Selexipag
Selexipag is an oral, selective prostacyclin IP receptor agonist prodrug used to treat pulmonary arterial hypertension (PAH, WHO Group I) in adults by delaying disease progression and reducing the risk of PAH-related complications such as hospitalization or death.[1][2] Marketed as Uptravi, it was approved by the U.S. Food and Drug Administration in December 2015 following the phase 3 GRIPHON trial, which enrolled 1,156 patients and showed a 40% relative risk reduction in the composite endpoint of death or PAH complications compared to placebo over a median follow-up of 1.4 years.[2][3] Unlike traditional prostacyclin analogs, selexipag's non-prostanoid structure allows oral administration with a pharmacokinetic profile enabling twice-daily dosing after gradual titration to minimize side effects like headache and diarrhea.[4][5] Its active metabolite, ACT-333679, selectively binds the IP receptor to induce pulmonary vasodilation, inhibit vascular smooth muscle proliferation, and suppress platelet aggregation, addressing core PAH pathophysiological mechanisms.[6][7]
Clinical Applications
Indications and Efficacy
Selexipag is indicated for the treatment of pulmonary arterial hypertension (PAH, World Health Organization Group 1) in adults to delay disease progression and reduce the risk of clinical worsening and hospitalization related to PAH.[8][9] This approval, granted by the U.S. Food and Drug Administration on December 21, 2015, for oral administration and extended to intravenous use on July 30, 2021, for patients unable to take oral therapy, is based on evidence from randomized controlled trials demonstrating benefits in reducing PAH-associated morbidity and mortality events.[8][10] Selexipag is not approved for other forms of pulmonary hypertension or non-PAH cardiovascular conditions.[8] The primary evidence for efficacy comes from the GRIPHON trial (NCT01106014), a phase 3, event-driven, multicenter, double-blind, placebo-controlled study enrolling 1,156 patients with PAH (WHO functional class I–IV) on stable background therapy, randomized 1:1 to selexipag (n=574, titrated from 200 mcg twice daily to a maximum of 1,600 mcg twice daily) or placebo (n=582), with a median treatment duration of 1.4 years for selexipag and 1.3 years for placebo.[2][11] The primary composite endpoint—time to first occurrence of death or PAH-related complications (worsening PAH symptoms, hospitalization for PAH, worsening WHO functional class, need for lung transplantation or balloon atrial septostomy, or initiation of parenteral prostacyclin analog therapy)—was reduced by 40% with selexipag versus placebo (hazard ratio 0.60, 99% confidence interval 0.46–0.78, P<0.001), with 27% of selexipag patients and 42% of placebo patients reaching an event.[2] This benefit was consistent across subgroups, including those on monotherapy, dual therapy, or triple therapy background regimens, and irrespective of disease duration at enrollment (≤6 months or >6 months from diagnosis).[12][13] Secondary endpoints in GRIPHON supported these findings: selexipag improved WHO functional class in 40.8% of patients (versus 30.2% with placebo) and showed numerical improvements in 6-minute walk distance (mean change +12 meters versus -10 meters) and pulmonary vascular resistance (geometric mean ratio 0.85), though the latter did not reach statistical significance after multiplicity adjustment.[2] Long-term data from the GRIPHON open-label extension (involving 953 patients initiating selexipag, 81% on background therapy) reported Kaplan-Meier survival estimates of 92.0% at 1 year, 84.0% at 3 years, 69.3% at 5 years, and 59.1% at 7 years among those randomized to selexipag in the parent study, with tolerability maintained over extended exposure despite dose adjustments for adverse events.[14][15] Real-world observational analyses have corroborated risk reduction in progression, emphasizing the value of serial risk assessments in guiding selexipag use within combination regimens.[16] Efficacy appears independent of dose level (low, medium, or high) but more pronounced with treatment durations exceeding 6 months.[17] No evidence supports efficacy in connective tissue disease-associated PAH beyond general PAH populations, though small cohort studies suggest potential improvements in exercise capacity and estimated right ventricular systolic pressure.[18]Dosage and Administration
Selexipag is administered orally as tablets twice daily, with the recommended starting dose of 200 micrograms (mcg) twice daily.[19] [20] Tolerability may be enhanced by taking the tablets with food, as this reduces peak plasma concentrations and associated adverse effects such as headache or nausea.[1] Dose escalation occurs in increments of 200 mcg twice daily, typically at weekly intervals, to reach the highest tolerated dose, which may go up to a maximum of 1600 mcg twice daily.[19] [21] Individualization is based on clinical response and tolerability, with slower titration possible if side effects like prostacyclin-related symptoms emerge; in clinical trials, the median achieved dose was 1200 mcg twice daily after 12 weeks.[19] Tablets are available in strengths of 200, 400, 600, 800, 1000, 1200, 1400, and 1600 mcg to facilitate precise dosing without splitting.[22] For patients with moderate hepatic impairment (Child-Pugh class B), the starting dose is reduced to 200 mcg once daily, with subsequent increases of 200 mcg at weekly intervals, not exceeding 1000 mcg daily in total.[23] No dose adjustment is required for mild hepatic impairment or severe renal impairment (creatinine clearance <30 mL/min), but use is not recommended in severe hepatic impairment (Child-Pugh C).[19] Elderly patients may require more cautious titration due to potentially higher exposure.[19] An intravenous formulation of selexipag, approved in 2021, is indicated for temporary use (up to 72 hours initially, extendable) when oral administration is not feasible, such as during interruptions.[10] It is infused twice daily over 80 minutes at a dose equivalent to the patient's current oral maintenance dose (225 to 1800 mcg, adjusted from tablet equivalents), prepared by reconstituting lyophilized powder and diluting in saline.[24] Transition back to oral therapy occurs once feasible, resuming at the pre-interruption dose if tolerated.[24]Patient Selection and Monitoring
Selexipag is indicated for adult patients with pulmonary arterial hypertension (PAH, World Health Organization Group 1) to delay disease progression and reduce the risk of hospitalization related to PAH.[24] Patient selection prioritizes those in WHO functional classes II or III, with confirmed diagnosis via right heart catheterization showing mean pulmonary artery pressure greater than 20 mmHg, pulmonary artery wedge pressure 15 mmHg or less, and pulmonary vascular resistance greater than 3 Wood units (updated from prior thresholds in recent guidelines).[25] Efficacy data derive primarily from patients with idiopathic or heritable PAH (58%), PAH associated with connective tissue disease (29%), or congenital heart disease with repaired shunts (10%), though use in other Group 1 subtypes follows similar risk-benefit assessment.[19] Selection incorporates comorbidity evaluation, including avoidance in severe hepatic impairment (Child-Pugh class C), hypersensitivity to selexipag, or concurrent strong CYP2C8 inhibitors like gemfibrozil, due to elevated exposure risks.[24] Dose initiation requires 200 μg twice daily, with weekly increments of 200 μg to the maximum tolerated dose up to 1600 μg twice daily, guided by tolerance to prostacyclin-mediated effects.[24] Adjustments apply for moderate hepatic impairment (Child-Pugh B: start 200 μg once daily, titrate by 200 μg weekly) or moderate CYP2C8 inhibitors (reduce to once-daily dosing at maintenance); no routine changes for mild-to-moderate renal impairment (eGFR >15 mL/min/1.73 m²) or age up to 75 years, though caution is advised in elderly patients due to potential comorbidities.[24] PAH-specific risk stratification, including WHO functional class, 6-minute walk distance, and echocardiography, informs ongoing suitability, with specialist oversight recommended for combination therapy or high-risk profiles.[26] Monitoring emphasizes adverse effect surveillance during titration, including headache (65% incidence), diarrhea (42%), jaw pain (26%), and nausea (33%), with temporary dose holds or reductions as needed.[19] Laboratory assessments include hemoglobin (decreases to <10 g/dL in 8.6% of patients versus 5% placebo) and thyroid-stimulating hormone (TSH reductions observed, warranting periodic testing).[24] Vigilance for pulmonary edema signs is critical, with immediate discontinuation if pulmonary veno-occlusive disease is confirmed; standard PAH follow-up—such as serial functional assessments and right heart catheterization—applies without selexipag-specific additions beyond these.[24] No enhanced monitoring is required for renal function in non-severe impairment, but dialysis patients lack data.[24]Safety and Risks
Contraindications
Selexipag is contraindicated in patients with hypersensitivity to the active substance or to any of the excipients in the formulation.[27][24] Concomitant use with strong inhibitors of cytochrome P450 2C8 (CYP2C8), such as gemfibrozil, is also contraindicated. This interaction substantially increases systemic exposure to selexipag's active metabolite MRE-269 by inhibiting its metabolism, potentially elevating the risk of adverse effects including hypotension and other prostacyclin-related toxicities.[27][24][20] While not formally contraindicated, selexipag should be avoided in patients with severe hepatic impairment (Child-Pugh class C) due to markedly elevated drug exposure; dosage adjustments or alternative therapies are recommended in such cases.[27][24]Adverse Effects
The most common adverse effects of selexipag, observed in the pivotal GRIPHON phase 3 trial involving 1,156 patients with pulmonary arterial hypertension, are consistent with the vasodilatory and prostacyclin-mimetic properties of the drug class, including headache (65% incidence versus 17% with placebo), diarrhea (42% versus 18%), jaw pain (26% versus 6%), nausea (33% versus 18%), myalgia (17% versus 7%), vomiting (18% versus 9%), pain in extremity (17% versus 8%), flushing (12% versus 4%), and arthralgia (11% versus 7%).[1][28] These effects led to treatment discontinuation in 14.3% of selexipag-treated patients compared to 7.1% on placebo, with headache, diarrhea, and nausea being the most frequent causes.[1] Serious adverse events attributable to selexipag were primarily extensions of common effects, such as severe headache or gastrointestinal intolerance, though disease progression (e.g., right ventricular failure) confounded attribution in the trial setting.[1] Hyperthyroidism occurred in 0.9% of patients (versus 0.2% placebo), potentially linked to prostacyclin pathway activation, while hypothyroidism was reported in 2.6% (versus 0.4%).[1] No significant increase in bleeding events was noted beyond background rates, despite theoretical risks from vasodilatory effects.[1] Long-term data from the GRIPHON open-label extension, following patients for up to 7 years (mean exposure 4.2 years), confirmed the safety profile remained consistent, with no new signals emerging; adverse events decreased over time as patients titrated to maintenance doses, and 78% of completers continued therapy.[14] Real-world pharmacovigilance from the FDA Adverse Event Reporting System (FAERS) through 2023 identified disproportionate signals for headache, diarrhea, and jaw pain, aligning with trial data but highlighting underreporting limitations in spontaneous systems.[29] Dose-dependent management, including gradual titration from 200 mcg twice daily, mitigates many effects, with reductions resolving symptoms in most cases.[1]Drug Interactions and Precautions
Concomitant administration of selexipag with strong inhibitors of CYP2C8, such as gemfibrozil, is contraindicated due to doubled exposure to selexipag and approximately 11-fold increased exposure to its active metabolite, the primary contributor to pharmacological activity.[27][20] Moderate CYP2C8 inhibitors, including clopidogrel, deferasirox, and teriflunomide, elevate active metabolite exposure by about 2.7-fold; in such cases, selexipag dosing should be reduced to once daily during coadministration, with reversion to twice-daily dosing upon discontinuation of the inhibitor.[27][20] CYP2C8 inducers like rifampin halve active metabolite exposure, potentially necessitating an increase in selexipag dose up to twice the targeted amount, with re-titration upon inducer discontinuation.[27][20] Selexipag and its active metabolite do not inhibit or induce cytochrome P450 enzymes or relevant transporters at clinically relevant concentrations, minimizing risks of selexipag precipitating interactions with other drugs.[27] In patients with moderate hepatic impairment (Child-Pugh class B), initiate selexipag at 200 mcg once daily and titrate cautiously up to a maximum of 1600 mcg daily, as systemic exposure to selexipag increases and active metabolite exposure may rise substantially.[20] Avoid selexipag in severe hepatic impairment (Child-Pugh class C) due to lack of data and potential for excessive exposure.[20] No initial dose adjustment is required for mild hepatic impairment (Child-Pugh class A) or renal impairment with eGFR greater than 15 mL/min/1.73 m², though titration should proceed with caution in severe renal impairment (eGFR less than 15 mL/min/1.73 m²) or dialysis due to limited data.[27][20] Discontinue selexipag if signs of pulmonary veno-occlusive disease, such as pulmonary edema, emerge, as it may worsen this condition.[27] Hypersensitivity to selexipag or its excipients contraindicates use.[20] During pregnancy, use only if benefits outweigh risks, given animal studies indicating developmental toxicity; limited human data exist.[27] Advise discontinuation of breastfeeding or selexipag, as the drug and metabolites may appear in milk and cause unknown effects in infants.[27]Pharmacological Profile
Mechanism of Action
Selexipag is a prodrug that undergoes hydrolysis primarily by carboxylesterase 1 in the liver and intestines to form its active metabolite, ACT-333679 (also known as MRE-269), which exhibits approximately 37-fold greater potency at the prostacyclin IP receptor compared to the parent compound.[1] [7] The active metabolite binds with high affinity to the human IP receptor (Ki = 0.5 nM), demonstrating over 130-fold selectivity for the IP receptor relative to other prostanoid receptors such as EP, FP, TP, DP1, and DP2.[1] [30] Activation of the IP receptor, a G-protein-coupled receptor expressed on vascular smooth muscle cells, endothelial cells, fibroblasts, and platelets, triggers downstream signaling via increased cyclic AMP (cAMP) levels, which promotes pulmonary vasodilation, reduces pulmonary vascular resistance, inhibits platelet aggregation, and exerts antiproliferative effects on vascular smooth muscle cells.[1] [31] Unlike traditional prostacyclin analogs (e.g., epoprostenol, treprostinil), selexipag and its metabolite possess a non-prostanoid chemical scaffold, enabling oral administration and potentially mitigating off-target effects associated with broader prostanoid receptor activation.[32] [33] This selective IP agonism addresses the prostacyclin pathway deficiency observed in pulmonary arterial hypertension, where reduced endogenous prostacyclin production contributes to vasoconstriction, thrombosis, and vascular remodeling.[7] Preclinical studies confirm that ACT-333679 induces potent relaxation of human pulmonary arteries and inhibits proliferation of pulmonary arterial smooth muscle cells in a concentration-dependent manner, supporting its role in counteracting disease pathophysiology.[34]Pharmacokinetics
Selexipag, a prodrug, is rapidly absorbed following oral administration, achieving median peak plasma concentrations (T<sub>max</sub>) of 1–3 hours for the parent compound and 3–4 hours for its active metabolite, ACT-333679.[1] Absolute bioavailability is approximately 49%, with food intake delaying T<sub>max</sub> and reducing maximum concentration (C<sub>max</sub>) by about 30% without altering overall exposure (area under the curve, AUC).[1] The pharmacokinetics of both selexipag and ACT-333679 remain consistent in patients with pulmonary arterial hypertension regardless of disease severity and do not change over time with repeated dosing.[1] Selexipag and its active metabolite exhibit high plasma protein binding of approximately 99%.[1] The apparent volume of distribution at steady state is 11.7 L for selexipag.[1] Metabolism begins with hydrolysis of selexipag by hepatic carboxylesterases to the more potent active metabolite ACT-333679, which accounts for the majority of pharmacological activity.[33] ACT-333679 undergoes further oxidative metabolism primarily via CYP2C8 (major pathway) and CYP3A4, along with glucuronidation by UGT1A3 and UGT2B7; minor metabolites constitute less than 3% of total drug-related material.[1][33] Elimination of selexipag occurs predominantly via hepatic metabolism, with over 90% of the dose excreted in feces (approximately 93%) and only about 12% in urine, reflecting primarily biliary clearance.[1][35] The terminal elimination half-life is 0.8–2.5 hours for selexipag and 6.2–13.5 hours for ACT-333679, yielding an effective half-life of 3–4 hours; mean total clearance is 17.9 L/hour.[1] Exposure increases in hepatic impairment (2- to 4-fold in mild to moderate cases) and severe renal impairment (40–70% higher), with no clinically significant variations by age, sex, race, or body weight.[1][35]Pharmacodynamics
Selexipag functions as a prodrug that is rapidly hydrolyzed by hepatic carboxylesterase-1 to its primary active metabolite, ACT-333679, which is approximately 37 times more potent as an agonist at the prostacyclin IP receptor than the parent compound.[32] This metabolite exhibits high selectivity for the IP receptor, activating downstream signaling pathways that promote vasodilation of pulmonary and systemic arteries, inhibit vascular smooth muscle proliferation, and suppress platelet aggregation in preclinical models.[32] Unlike prostacyclin analogs, ACT-333679 demonstrates reduced β-arrestin recruitment and lower potential for receptor desensitization, potentially contributing to sustained efficacy with oral dosing.[36] In vitro pharmacodynamic assessments reveal dose-dependent inhibition of platelet aggregation by ACT-333679 with an IC<sub>50</sub> of 0.21 μM, whereas selexipag itself has an IC<sub>50</sub> of 5.5 μM; however, therapeutic plasma concentrations achieved with up to 1600 mcg twice daily do not produce clinically meaningful antiplatelet effects in vivo.[27] Selexipag and ACT-333679 also lack significant agonism at other prostanoid receptors, minimizing off-target vasoconstrictive risks observed with less selective agents.[32] Cardiac electrophysiology studies confirm no prolongation of the QT interval at maximum tolerated doses.[27] In patients with pulmonary arterial hypertension, selexipag elicits measurable hemodynamic improvements, including a 30.3% reduction in pulmonary vascular resistance (95% CI: -44.7% to -12.2%) and a 0.41 L/min/m² increase in cardiac index (95% CI: 0.10 to 0.71) after 17 weeks of titration to 800 mcg twice daily in a phase 2 trial.[27] Early treatment with 200 mcg twice daily yields a modest 3% decrease in NT-proBNP levels at week 4, though this effect is not sustained long-term.[32] No clinically relevant pharmacodynamic interactions occur with warfarin, as steady-state exposure to selexipag at 400 mcg twice daily does not alter international normalized ratio values.[27]Clinical Evidence
Pivotal Clinical Trials
The GRIPHON trial (AC-065A302) was a multicenter, double-blind, event-driven phase 3 study evaluating selexipag in patients with pulmonary arterial hypertension (PAH).[11] It randomized 1,156 adults (aged 18-75 years) with symptomatic PAH (WHO functional class I-IV) in a 1:1 ratio to receive selexipag or matching placebo, alongside standard background therapy (endothelin receptor antagonists, phosphodiesterase type 5 inhibitors, or both).[2] Selexipag was initiated at 200 μg twice daily and up-titrated by 200 μg twice daily every 4 days to the highest tolerated dose (maximum 1,600 μg twice daily), with 80% of patients reaching at least 800 μg twice daily.[2] The primary composite endpoint was time from randomization to the first morbidity or mortality event, defined as death from any cause, PAH-related complications (sustained worsening of WHO functional class, PAH symptoms leading to hospitalization or IV prostanoid initiation, or <15% decline in 6-minute walk distance with need for prostanoid), or other events requiring intervention.[2] Selexipag reduced the risk of this endpoint by 40%, with a hazard ratio of 0.60 (95% CI, 0.46-0.78; P<0.001); events occurred in 27% of the selexipag group versus 42% in placebo over a median follow-up of 1.4 years (maximum 4.2 years).[2] All-cause mortality was similar between groups (HR 0.87; 95% CI, 0.47-1.62), but PAH-related death or hospitalization showed benefit (HR 0.64; 95% CI, 0.46-0.88).[2] Secondary endpoints included change in 6-minute walk distance (6MWD) from baseline to 26 weeks, WHO functional class, and N-terminal pro-brain natriuretic peptide levels.[37] Selexipag did not significantly improve 6MWD (least squares mean difference +12 meters; 95% CI, -5 to 29; P=0.16) or functional class distribution at week 26, though improvements trended positive.[2] Exploratory analyses indicated consistent primary endpoint benefits across subgroups, including treatment-naïve patients and those on monotherapy or combination therapy.[12] This trial formed the basis for selexipag's FDA approval on December 21, 2015, for delaying disease progression and reducing PAH-related hospitalization risk in WHO group 1 PAH patients (functional class II-III).[37] Long-term data from the open-label extension (GRIPHON OL), involving 1,447 patients (including GRIPHON completers and de novo entrants), confirmed sustained tolerability over up to 7 years, with Kaplan-Meier survival estimates of 92% at 1 year, 84% at 3 years, 70% at 5 years, and 53% at 7 years among original selexipag-randomized patients.[15] Adverse events were primarily prostacyclin-related (headache, diarrhea, jaw pain, nausea), leading to discontinuation in 14.3% versus 7.1% on placebo in GRIPHON.[2]Real-World Effectiveness and Safety
In real-world settings, selexipag has demonstrated effectiveness in managing pulmonary arterial hypertension (PAH), particularly when initiated early. Analysis of U.S. claims data from 2,966 PAH patients on background endothelin receptor antagonists and phosphodiesterase-5 inhibitors showed that adding selexipag in 351 patients (11.8%)—especially within 6 months of starting dual therapy—was associated with reduced risks of all-cause hospitalization, PAH-related hospitalization, and disease progression (e.g., initiation of parenteral prostacyclin, death, lung transplant, or atrial septostomy); benefits attenuated if added after 12 months.[38] The SPHERE registry, capturing U.S. clinical practice, enrolled patients typically at WHO functional class II/III and intermediate REVEAL 2.0 risk despite background therapies, with most showing stable or improved functional class and risk category after selexipag initiation.[13] [39] Comparative real-world evidence indicates favorable survival outcomes with selexipag. In a retrospective analysis, selexipag treatment was associated with a lower relative risk of death compared to other PAH-specific therapies, after adjusting for confounders.[40] Persistence data from routine care highlight sustained use, with individualized up-titration enabling maintenance doses across strata without dose-dependent differences in outcomes.[41] [42] Safety in post-marketing use aligns with prostacyclin-class effects, without emergent signals beyond trials. In the SPHERE registry, adverse event-related discontinuations occurred in approximately 7.2% of patients, most commonly due to headache (6.5%), with overall tolerability supporting long-term adherence in intermediate-risk cohorts.[16] FDA Adverse Event Reporting System analysis confirmed a profile limited to known events like headache, diarrhea, and nausea, with no disproportionate signals relative to other oral prostacyclins.[43] Real-world cohorts report comparable event rates to GRIPHON across doses, emphasizing the role of gradual titration in mitigating prostacyclin-associated side effects.[42]Comparative Studies
A network meta-analysis of randomized controlled trials evaluating oral prostacyclin pathway therapies, including selexipag and oral treprostinil, found no significant differences between selexipag and oral treprostinil in improvements to 6-minute walk distance (6MWD) or reductions in clinical worsening risk.[44] Selexipag demonstrated a relative risk (RR) of 0.47 (95% CI 0.35–0.65) for clinical worsening versus placebo, comparable to oral treprostinil's RR of 0.65 (95% CI 0.46–0.90).[44] In real-world evidence from a retrospective U.S. claims database analysis (2015–2017) involving 123 selexipag and 99 oral treprostinil initiators with pulmonary hypertension, selexipag was associated with a 46% lower risk of all-cause hospitalization (HR 0.54, 95% CI 0.31–0.92) and a 47% lower risk of pulmonary hypertension-related hospitalization (HR 0.53, 95% CI 0.31–0.93).[45] Corresponding hospitalization rates were 42% and 46% lower with selexipag (RR 0.58, 95% CI 0.39–0.87 for all-cause; RR 0.54, 95% CI 0.35–0.82 for pulmonary hypertension-related).[45] A broader network meta-analysis of prostacyclin-based therapies across formulations ranked selexipag highest for reducing clinical worsening (P-score 0.95; RR 0.62, 95% CI 0.51–0.74 versus placebo) but lowest for 6MWD improvement (P-score 0.15).[46] Epoprostenol ranked superior for 6MWD gains (mean difference 46.84 m, 95% CI 21.90–71.78) and hemodynamic improvements, while treprostinil showed mortality benefits (RR 0.66, 95% CI 0.49–0.90 versus placebo).[46] Observational data from the EXPOSURE study (initiated 2017), a prospective post-authorization safety study of over 2,000 PAH patients, indicated a 45% lower mortality risk with selexipag versus propensity score-weighted other PAH therapies (rate ratio 0.55, 95% CI 0.31–0.99; 10.5% mortality in selexipag cohort versus 17.6% in comparators over ~830 person-years follow-up).[40] Selexipag was predominantly used in triple combination regimens.[40] Compared to inhaled iloprost in real-world use, selexipag initiation correlated with lower rates of hospitalization and outpatient visits among PAH patients, alongside comparable drug persistence.[47] These findings derive from indirect comparisons and observational cohorts, as direct head-to-head randomized trials remain scarce; limitations include potential confounding by baseline differences and combination therapy variations.[40][45]Chemical and Manufacturing Aspects
Molecular Structure and Properties
Selexipag has the molecular formula C<sub>26</sub>H<sub>32</sub>N<sub>4</sub>O<sub>4</sub>S and a molecular weight of 496.62 g/mol.[6] Its IUPAC name is 2-{4-[(5,6-diphenylpyrazin-2-yl)(propan-2-yl)amino]butoxy}-N-(methanesulfonyl)acetamide.[48] The core structure features a pyrazine ring substituted with phenyl groups at the 5- and 6-positions, an N-isopropylamino group at the 2-position linked to a butoxy chain, and terminating in an N-methanesulfonylacetamide moiety.[6] Selexipag is characterized by low water solubility of approximately 0.004 mg/mL and a logP value of 4.4, indicating significant lipophilicity.[33] Solubility is pH-dependent, with insolubility in aqueous media at pH 2–4, free solubility at pH 8, and high solubility at pH 9–12.[49] These properties influence its formulation and bioavailability as an oral prostacyclin receptor agonist prodrug.[6]Synthesis and Formulation
Selexipag is synthesized through a multi-step organic process that constructs the central 5,6-diphenylpyrazine moiety followed by attachment of the substituted butoxyacetamide side chain. A key intermediate, 4-[(5,6-diphenylpyrazin-2-yl)(isopropyl)amino]butan-1-ol, is prepared by nucleophilic aromatic substitution of 2-chloro-5,6-diphenylpyrazine with 4-(isopropylamino)butan-1-ol in the presence of a base such as sodium hydride in a solvent like dimethylformamide.[50] This alcohol is then converted to the mesylate or tosylate, displaced with glycolic acid derivative, and amidated with methanesulfonamide to yield selexipag, often under conditions involving coupling agents like dicyclohexylcarbodiimide.[51] Alternative routes involve protection of the alcohol as tert-butyl ether during pyrazine assembly from precursors like 2,3-diphenylpyrazine carboxylic acid derivatives, followed by deprotection and side chain elaboration.[7] These processes, detailed in patents held by Actelion Pharmaceuticals, emphasize control of impurities and yield optimization for large-scale production.[52] The active pharmaceutical ingredient (API) is a pale yellow crystalline powder with low aqueous solubility, necessitating specific formulation strategies for oral bioavailability. Selexipag is primarily formulated as film-coated tablets in strengths of 200 μg to 1600 μg, employing a wet granulation process to achieve content uniformity at microgram doses.[53] The manufacturing involves dry blending the API with excipients like mannitol and microcrystalline cellulose, followed by wet granulation with a binder solution, drying, milling, lubrication, compression into tablets, and coating with a polymer film containing titanium dioxide for protection and identification.[53] This approach ensures stability and consistent release, with the tablets also containing croscarmellose sodium as a disintegrant and magnesium stearate as a lubricant.[24] Polymorphic forms, such as solvates or specific crystal habits, are controlled during synthesis and formulation to maintain consistent physicochemical properties and dissolution profiles.[54] An intravenous formulation, approved in 2021, consists of selexipag in a lyophilized powder with glycine, phosphoric acid, polysorbate 20, and sodium hydroxide for reconstitution, intended for temporary use in patients unable to take oral therapy.[24]Development History
Preclinical and Early Development
Selexipag, chemically known as ACT-293987, was discovered and synthesized by Nippon Shinyaku Co., Ltd. as a non-prostanoid, selective prostacyclin IP receptor agonist designed for oral administration in the treatment of pulmonary arterial hypertension (PAH). It functions as a prodrug that undergoes hydrolysis to its active metabolite ACT-333679 (also denoted MRE-269), which binds the IP receptor with high affinity (Ki = 20 nM) and demonstrates greater than 130-fold selectivity over other prostanoid receptors.[7] [55] Preclinical in vitro pharmacology revealed that the active metabolite elevates cyclic AMP (cAMP) levels in cells expressing the human IP receptor with an EC<sub>50</sub> of 11 nM and inhibits proliferation of human pulmonary artery smooth muscle cells with an IC<sub>50</sub> of 2.9 nM, supporting its antiproliferative and vasodilatory potential in PAH pathology.[7] In animal models, selexipag exhibited robust efficacy without inducing tachyphylaxis. In the monocrotaline-induced PAH rat model, oral dosing at 1 mg/kg twice daily reduced right ventricular pressure and enhanced survival rates. In the Sugen 5416/hypoxia (SuHx) model, administration to Sprague-Dawley rats at 30 mg/kg twice daily for three weeks lowered right ventricular systolic pressure from 141.1 mmHg to 102.1 mmHg, decreased the right ventricle-to-left ventricle plus septum weight ratio from 0.63 to 0.50, reduced occlusive lesion prevalence from 55% to 26%, and diminished medial wall thickness from 29.5% to 16%, alongside reductions in vascular proliferation (Ki-67-positive cells) and fibrosis (collagen type I expression). Comparable improvements in right ventricular hypertrophy and 100% survival (versus 30% in vehicle controls) were observed in Fischer rats under similar dosing. These effects occurred without altering systemic mean arterial pressure or heart rate, indicating pulmonary selectivity.[7] [56] Early development advanced through a 2008 exclusive worldwide collaboration between Nippon Shinyaku and Actelion Pharmaceuticals Ltd., enabling progression to clinical evaluation while leveraging the compound's favorable pharmacokinetic profile, including sustained IP receptor activation via the long-acting metabolite.[57]
Regulatory Milestones and Approvals
Selexipag received orphan drug designation in the European Union on August 26, 2005, for the treatment of pulmonary arterial hypertension (PAH).[58] It was granted orphan status in Japan in September 2014 and in the United States prior to its initial approval, recognizing PAH as a rare disease qualifying for incentives to support development.[59][60] The new drug application for oral selexipag was submitted to the U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA) in December 2014. The FDA approved selexipag (as Uptravi tablets) on December 21, 2015, for the treatment of adults with PAH (WHO Group I) to delay disease progression and reduce the risk of hospitalization, based on the GRIPHON trial demonstrating a 40% reduction in morbidity/mortality events.[61][60] The EMA followed with marketing authorization on May 12, 2016, for the same indication in adults with WHO Functional Class II–III PAH.[62] Subsequent approvals expanded access: Health Canada authorized it on January 21, 2016; Swissmedic on August 15, 2016; and Japan's Ministry of Health, Labour and Welfare enabled launch in November 2016 for PAH.[63] In July 2021, the FDA approved an intravenous formulation of selexipag for adult PAH patients when oral therapy cannot be continued, providing a bridge during interruptions with equivalent exposure to oral dosing.[10]| Regulatory Agency | Approval Date | Formulation and Indication |
|---|---|---|
| FDA (US) | December 21, 2015 | Oral tablets; PAH (WHO Group I) to delay progression and reduce hospitalization risk[61] |
| Health Canada | January 21, 2016 | Oral; PAH[63] |
| EMA (EU) | May 12, 2016 | Oral tablets; PAH (WHO FC II–III)[62] |
| PMDA (Japan) | November 2016 (launch) | Oral; PAH[64] |
| FDA (US) | July 30, 2021 | Intravenous; PAH when oral interrupted[10] |