Eptifibatide Human

Eptifibatide Human
Shipped with Ice Packs
In Stock

Description

Eptifibatide selectively and reversibly binds to the GP IIb/IIIa receptor on activated platelets, blocking fibrinogen, von Willebrand factor, and other ligands from binding—a critical step in platelet aggregation . This inhibition is dose-dependent and rapidly reversible upon discontinuation .

Key Pharmacodynamic Findings:

  • Baboon Model: Complete inhibition of platelet aggregation at infusion rates >5.0 µg/kg/min .

  • Human Studies:

    • 135 µg/kg bolus + 0.5 µg/kg/min infusion: 69% platelet inhibition at 15 minutes, 40–50% at steady state .

    • 180 µg/kg bolus + 2.0 µg/kg/min infusion: >90% inhibition at steady state .

  • Bleeding Time: Prolonged <5-fold during infusion, returning to baseline within 6 hours post-infusion .

Pharmacokinetics

Eptifibatide exhibits linear pharmacokinetics with a plasma half-life of ~2.5 hours and 25% protein binding .

ParameterValueClinical Implications
Clearance55 mL/kg/h (coronary artery disease)Reduced by ~50% in renal impairment
Renal Excretion50% of total clearanceRequires dose adjustment for CrCl <50 mL/min
MetabolismMinimal hepatic involvementNo major metabolites detected

PURSUIT Trial (10,948 patients with unstable angina/NSTEMI)37:

  • Primary Endpoint: 1.5% absolute reduction in death/MI at 30 days (14.2% vs. 15.7%, p=0.004).

ESPRIT Trial (PCI patients)46:

  • Composite Endpoint: 37% relative risk reduction in death/MI/urgent revascularization at 48 hours (p=0.002).

  • 6-Month Outcomes: Sustained benefit with 6.8% vs. 10.5% event rate (p=0.002) .

IMPACT II (4,010 PCI patients)6:

  • Dose Response: 135 µg/kg bolus + 0.5 µg/kg/min infusion reduced ischemic complications by 22% (p=0.035) .

Standard Regimen189:

  • Acute Coronary Syndromes:

    • Bolus: 180 µg/kg IV.

    • Infusion: 2.0 µg/kg/min for up to 72–96 hours.

  • Percutaneous Coronary Intervention (PCI):

    • Double bolus (180 µg/kg ×2) + 2.0 µg/kg/min infusion for 18–24 hours post-procedure.

Recent Developments and Future Directions

  • COVID-19 Thrombosis: Investigated for salutary effects in COVID-related hypercoagulability .

  • Ongoing Trials:

    • EARLY-ACS: Upstream therapy in high-risk NSTE-ACS .

    • EVA-AMI: Comparison with abciximab in primary PCI .

Product Specs

Introduction
Eptifibatide is a cyclic peptide, derived from a protein found in the venom of the southeastern pygmy rattlesnake, that inhibits platelet aggregation. It achieves this by binding to the glycoprotein (GP) IIb/IIIa receptor on human platelets. As an arginin-glycin-aspartat-mimetic, it demonstrates reversible binding to platelets.
Description
Synthetic Eptifibatide is a hexapeptide with a molecular weight of 832.5 Daltons. Its amino acid sequence is: Met-Pro-Har-Gly-Asp-Trp-Pro-Cys-NH2.
Physical Appearance
White, sterile-filtered powder in lyophilized (freeze-dried) form.
Formulation
The lyophilization process of Eptifibatide does not involve any additives.
Solubility
For reconstitution of lyophilized Eptifibatide, it is recommended to use sterile 18MΩ-cm H2O at a minimum concentration of 100 µg/ml. This solution can be further diluted with other aqueous solutions.
Stability
Lyophilized Eptifibatide exhibits stability at room temperature for up to 3 weeks; however, for extended storage, it should be stored in a desiccated environment below -18°C. After reconstitution, Eptifibatide should be stored at 4°C for 2-7 days. For long-term storage, freezing at -18°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is suggested for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of this product is greater than 98.0%, determined by: (a) Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis and (b) Sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis.

Q&A

What is the primary mechanism of action for eptifibatide in human platelets?

Eptifibatide functions as a glycoprotein IIb/IIIa receptor antagonist, inhibiting platelet aggregation by preventing fibrinogen binding to activated platelets. In vitro studies demonstrate that eptifibatide effectively blocks platelet adhesion to fibrinogen-coated surfaces even at low concentrations. The compound achieves this by competitively binding to the receptor sites, preventing the cross-linking of platelets that would otherwise lead to thrombus formation. Research indicates that eptifibatide exhibits concentration-dependent inhibition of platelet aggregation in response to various agonists including thrombin, collagen, and ADP .

Experimental methodologies for evaluating this mechanism typically involve washed platelet preparations or platelet-rich plasma with measurements of aggregation responses using light transmission aggregometry and secretion assays utilizing luminescence techniques for ATP release detection .

How do standardized laboratory protocols measure eptifibatide's efficacy in platelet inhibition?

Laboratory assessment of eptifibatide's efficacy employs several standardized approaches:

  • Platelet Aggregation Studies: Using light transmission aggregometry to quantify percentage inhibition of aggregation in response to agonists (thrombin, collagen, ADP) at various eptifibatide concentrations. Studies have determined IC50 values of approximately 22 μg/mL for ADP-induced and 29 μg/mL for collagen-induced aggregation .

  • Adhesion Assays: Measuring platelet adhesion to fibrinogen-coated surfaces using microtiter plate-based methods, where collagen-stimulated adhesion is inhibited by eptifibatide in a dose-dependent manner .

  • Secretion Measurements: Quantifying dense granule secretion through ATP release assays and lysosomal secretion via β-hexosaminidase assay methods. Research demonstrates that eptifibatide inhibits secretion reactions differently based on the stimulating agonist .

  • Simultaneous Aggregation-Secretion Monitoring: Employing dual-channel recording techniques with luciferin/luciferase reagents to concurrently assess both aggregation and ATP release, providing integrated functional data .

These methodologies allow researchers to comprehensively characterize eptifibatide's pharmacodynamic profile across different platelet activation pathways.

What are the essential control conditions when designing in vitro experiments with eptifibatide?

When designing rigorous in vitro experiments with eptifibatide, researchers should implement the following control conditions:

  • Vehicle Controls: Include appropriate diluent controls matching the eptifibatide preparation to account for potential solvent effects.

  • Positive Control Inhibitors: Incorporate alternative glycoprotein IIb/IIIa inhibitors (such as tirofiban) for comparative pharmacology.

  • Agonist Concentration Optimization: Establish threshold concentrations for each platelet agonist (e.g., 0.2-0.25 U/mL for thrombin, 15-20 μg/mL for collagen, 8-12 μM for ADP) to ensure sensitivity to inhibition .

  • Concentration Range Selection: Test a broad eptifibatide concentration range (typically from sub-therapeutic to supra-therapeutic) to generate complete dose-response relationships.

  • Multiple Donor Sampling: Conduct experiments across platelet preparations from multiple donors (n≥7 recommended) to account for interpersonal variability in receptor density and signaling efficiency .

  • Time-Course Evaluations: Measure responses at multiple time points (e.g., immediate, 3 minutes, and extended intervals) to capture both acute inhibition and potential recovery phenomena.

These controls ensure experimental rigor and facilitate meaningful data interpretation in eptifibatide research.

How were the pivotal PURSUIT and ESPRIT trials designed to evaluate eptifibatide efficacy in acute coronary syndromes?

The PURSUIT (Platelet Glycoprotein IIb/IIIa in Unstable Angina: Receptor Suppression Using Integrilin Therapy) and ESPRIT trials represent landmark studies in eptifibatide research, with distinct methodological approaches:

PURSUIT Trial Design:

  • Scope: 726 centers across 27 countries with 10,948 patients presenting with unstable angina or non-Q-wave myocardial infarction

  • Inclusion Criteria: Cardiac ischemia at rest (≥10 minutes) within 24 hours plus either ST-segment changes (depression >0.5mm of <30 minutes or persistent ST elevation >0.5mm not requiring reperfusion therapy) or T-wave inversion (>1mm) or increased CK-MB

  • Randomization: Double-blind allocation to placebo, eptifibatide 180μg/kg bolus followed by 2.0μg/kg/min infusion, or eptifibatide 180μg/kg bolus followed by 1.3μg/kg/min infusion

  • Duration: Infusion continued until hospital discharge, CABG, or up to 72 hours (extended to 96 hours if PCI performed)

  • Primary Endpoint: Death or myocardial infarction at 30 days

ESPRIT Trial Design:

  • Focus: Double-blind, multicenter, randomized, parallel-group, placebo-controlled evaluation of eptifibatide safety and efficacy in patients undergoing non-emergent percutaneous coronary intervention

  • Specific methodological details beyond this are not fully elaborated in the provided search results

The PURSUIT trial demonstrated that eptifibatide (180/2.0 dosing) significantly reduced the primary endpoint incidence from 15.8% (placebo) to 14.3% (eptifibatide), representing approximately 15 events avoided per 1,000 patients treated (p=0.034) . Notably, in the United States subgroup analysis, the treatment effect was more pronounced, with a reduction from 15.4% to 11.9% (p=0.003) .

What methodological considerations explain regional variations in eptifibatide efficacy outcomes?

The PURSUIT trial revealed significant regional variations in eptifibatide efficacy, with more substantial benefits observed in U.S. patients compared to those enrolled elsewhere. Several methodological considerations explain these variations:

  • Practice Pattern Differences: Prospectively planned analysis acknowledged expected differences in medical practice patterns across regions, which influenced both baseline risk and treatment approaches .

  • Revascularization Strategy Variability: The PURSUIT design allowed discretionary revascularization decisions by treating physicians, leading to regional differences in PCI and CABG utilization rates that potentially modified the drug's impact .

  • Timing of Interventions: Regional variations in door-to-balloon times and timing of invasive procedures likely influenced the opportunity for eptifibatide to prevent periprocedural myocardial infarction.

  • Adjunctive Pharmacotherapy: Differences in concurrent medication use (anticoagulants, antiplatelet agents, statins) may have created regional interaction effects.

  • Endpoint Ascertainment: Despite standardized definitions, regional differences in diagnostic thresholds and biomarker utilization may have affected endpoint detection sensitivity.

  • Population Heterogeneity: Genetic factors influencing platelet reactivity and drug metabolism could contribute to regional response variations.

Among U.S. patients, eptifibatide reduced the primary endpoint of death or MI at 30 days from 15.4% to 11.9% (p=0.003), with benefits maintained at 6 months (18.9% vs. 15.2%, p=0.004) . This methodological heterogeneity underscores the importance of region-specific subgroup analyses in multinational cardiovascular trials.

How should researchers interpret and reconcile bleeding risk data from eptifibatide clinical trials?

Interpreting bleeding risk data from eptifibatide trials requires sophisticated analytical approaches:

How do researchers differentiate between direct pharmacological effects of eptifibatide and downstream consequences of platelet inhibition?

Differentiating direct pharmacological effects from downstream consequences requires sophisticated experimental designs:

  • Receptor Binding vs. Functional Assays: Direct pharmacological effects are measured through specific receptor occupancy assays using radiolabeled eptifibatide or competitive binding assays, while functional consequences are assessed through aggregation and secretion measurements .

  • Temporal Resolution Studies: Time-course experiments can separate immediate receptor-mediated effects from delayed downstream signaling events. Direct effects typically manifest within seconds to minutes while downstream consequences evolve over extended timeframes.

  • Isolated Signaling Pathway Analysis: Using specific pathway inhibitors in combination with eptifibatide helps determine whether observed effects are directly receptor-mediated or result from secondary signaling cascades.

  • Concentration-Effect Relationship Analysis: Direct effects typically show simple concentration-dependent relationships while downstream consequences often exhibit complex, non-linear relationships across concentration ranges.

  • Comparative Pharmacology: Comparing eptifibatide effects with other GPIIb/IIIa antagonists of different chemical structures helps distinguish class effects (shared mechanisms) from compound-specific actions.

Research demonstrates that while eptifibatide directly inhibits fibrinogen binding to GPIIb/IIIa receptors, its effects on granule secretion may represent both direct and indirect mechanisms depending on the activation pathway being studied. For instance, at concentrations inhibiting collagen-induced aggregation by ~50%, eptifibatide inhibits dense granule secretion by only ~30%, suggesting mechanistic complexity beyond simple aggregation inhibition .

What are the established methodologies for studying rare adverse events like eptifibatide-induced thrombocytopenia and thrombosis?

Investigating rare adverse events like eptifibatide-induced thrombocytopenia and thrombosis requires specialized methodological approaches:

  • Ex Vivo Antibody Detection Assays: Researchers have developed serum testing methods where patient serum (collected 2-3 days after eptifibatide administration) is incubated with normal platelets in the presence of eptifibatide. Simultaneous measurement of platelet aggregation and granule secretion using lumi-aggregometry can identify antibody-mediated effects .

  • IgG Depletion Studies: Confirmatory experiments employ protein G Sepharose beads to deplete IgG from patient serum samples. Loss of platelet-activating capability after IgG depletion confirms the antibody-mediated nature of the adverse reaction .

  • Comparative Activation Assessment: Standardizing granule secretion measurements against strong agonists (like collagen-related peptide) provides quantitative comparison of antibody-induced activation potential. In documented cases, patient antibodies plus eptifibatide induced granule secretion comparable to that of strong physiological agonists .

  • Flow Cytometry Analysis: Advanced methods utilize flow cytometry to detect drug-dependent antibody binding to platelets and subsequent activation marker expression (P-selectin, activated GPIIb/IIIa).

  • Longitudinal Platelet Monitoring: Serial platelet count measurements in high-risk patients, with standardized thresholds for thrombocytopenia definition, enable early detection of this rare complication.

These methodologies have revealed that eptifibatide-induced thrombocytopenia results from patient antibodies that, paradoxically, activate platelets in the presence of the drug, potentially explaining the simultaneous occurrence of thrombocytopenia and thrombosis in some patients .

How can researchers quantify the relative contribution of eptifibatide to outcomes in multimodal treatment approaches?

Isolating eptifibatide's specific contribution within multimodal treatment regimens presents significant methodological challenges. Researchers employ several sophisticated approaches:

  • Propensity Score Matching: As demonstrated in the EPOCH and ANGEL-ACT studies examining eptifibatide with mechanical thrombectomy (MT) for acute ischemic stroke, propensity matching creates comparable treatment groups, allowing isolation of eptifibatide's effects. This approach matched 81 eptifibatide+MT patients with 81 MT-only patients to evaluate outcomes .

  • Multivariable Regression Models with Interaction Terms: Mixed-effects logistic regression models account for within-hospital clustering while simultaneously adjusting for confounding variables. These models can include interaction terms to assess whether eptifibatide's effect differs across subgroups .

  • Sequential Treatment Introduction Designs: Some trials implement a phased introduction of therapies to isolate individual treatment contributions through temporal comparisons.

  • Mediation Analysis: Statistical approaches that quantify how much of a treatment's total effect occurs through specific mechanistic pathways.

  • Biomarker-Based Attribution: Correlating pharmacodynamic markers specific to eptifibatide (e.g., degree of platelet inhibition) with outcomes to establish mechanistic links.

The EPOCH study demonstrated that MT combined with eptifibatide resulted in significantly higher rates of successful recanalization (91.3% versus 81.5%; p=0.043) and good clinical outcomes at 3 months (53.1% versus 33.3%; p=0.016) compared to MT alone, without significant increases in intracranial hemorrhage complications . These findings suggest substantial contributions from adjunctive eptifibatide in this context.

What methodological approaches best assess eptifibatide's efficacy in acute ischemic stroke research?

Research investigating eptifibatide in acute ischemic stroke requires specialized methodological approaches:

  • Propensity-Matched Control Studies: The EPOCH trial utilized propensity matching with the ANGEL-ACT trial data to create comparable treatment groups (81 eptifibatide+MT patients matched with 81 MT-only patients), controlling for baseline characteristics that might confound treatment effect measurement .

  • Standardized Outcome Measures:

    • Primary efficacy outcome: Modified Rankin Scale (mRS) score 0-2 at 3 months, representing good functional outcomes

    • Secondary measures: Full mRS distribution analysis, poor outcome rates (mRS 5-6), and successful recanalization rates

    • Safety outcomes: Any intracranial hemorrhage, symptomatic intracranial hemorrhage, and 3-month mortality

  • Multicentered Design with Standardized Protocols: The EPOCH trial enrolled patients across 15 hospitals between April 2019 and March 2020, while ANGEL-ACT included 111 hospitals, ensuring diverse patient populations and enhancing generalizability .

  • Mixed-Effects Statistical Models: Using mixed-effects logistic regression models to account for within-hospital clustering while adjusting for relevant clinical variables .

  • Standardized Reperfusion Assessment: Employing validated angiographic scales to quantify reperfusion success after mechanical thrombectomy with or without eptifibatide.

This methodological framework demonstrated that MT combined with eptifibatide achieved significantly higher rates of successful recanalization (91.3% vs. 81.5%; p=0.043) and good clinical outcomes at 3 months (53.1% vs. 33.3%; p=0.016) compared to MT alone, without significant increases in hemorrhagic complications .

How do researchers control for variables that impact platelet response to eptifibatide in experimental systems?

Controlling for variables affecting platelet responses to eptifibatide requires rigorous experimental design:

  • Standardized Platelet Preparation:

    • For washed platelets: Consistent isolation protocols with specified centrifugation speeds/times and buffer compositions

    • For PRP: Standardized centrifugation (typically 190×g for 12-15 minutes) and platelet count adjustment

  • Agonist Concentration Optimization: Establishing threshold concentrations for each agonist through preliminary dose-finding experiments (e.g., 0.2-0.25 U/mL for thrombin, 15-20 μg/mL for collagen, 8-12 μM for ADP) .

  • Temperature and pH Control: Maintaining consistent experimental conditions (37°C, physiological pH) throughout all assays .

  • Donor Selection Criteria:

    • Medication restrictions: No antiplatelet medications for ≥10 days

    • Dietary controls: Fasting status or standardized meals before blood collection

    • Demographic balancing: Age, sex, and ethnicity considerations

  • Timing Standardization:

    • Consistent incubation periods before agonist addition

    • Uniform measurement timepoints (e.g., 3 minutes post-stimulation)

  • Technical Replication: Multiple determinations (≥6) from different platelet preparations (≥3) to account for biological variability .

  • Calibration Procedures: Using standard curves or internal controls for secretion assays to enable quantitative comparisons across experiments .

Research demonstrates that these controls are essential, as platelet responses to eptifibatide vary significantly based on the activation pathway studied. For example, eptifibatide exhibits differential inhibitory potency against thrombin-, collagen-, and ADP-induced platelet responses, highlighting the importance of standardized experimental conditions .

What are the optimal approaches for translating in vitro eptifibatide findings to clinical applications?

Translating in vitro eptifibatide findings to clinical applications involves several methodological bridges:

  • Concentration-Effect Relationship Mapping: Establishing in vitro IC50 values (e.g., ~22 μg/mL for ADP-induced and ~29 μg/mL for collagen-induced aggregation) and correlating these with plasma concentrations achieved clinically with standard dosing regimens (180 μg/kg bolus followed by 2.0 μg/kg/min infusion) .

  • Ex Vivo Confirmation Studies: Testing blood samples from patients receiving eptifibatide to verify that in vitro observations translate to the clinical setting. This approach bridges the gap between controlled laboratory conditions and the complex in vivo environment.

  • Mechanistic Biomarker Integration: Identifying pathway-specific biomarkers that can be measured both in vitro and in patients to create translational linkages. For example, correlating in vitro dense granule secretion inhibition with clinical markers of platelet activation.

  • Pharmacokinetic/Pharmacodynamic (PK/PD) Modeling: Developing mathematical models that integrate in vitro potency data with clinical pharmacokinetics to predict therapeutic effects across diverse patient populations.

  • Targeted Clinical Endpoint Selection: Designing clinical trials with endpoints specifically linked to mechanisms established in vitro. For instance, the PURSUIT trial's focus on death or myocardial infarction aligns with eptifibatide's demonstrated anti-aggregatory effects .

  • Surrogate Endpoint Validation: Establishing correlations between in vitro parameters (e.g., degree of aggregation inhibition) and clinical outcomes to develop validated surrogate endpoints for future research.

Through systematic application of these approaches, researchers have successfully translated fundamental observations on eptifibatide's platelet inhibitory mechanisms to clinical protocols that demonstrate significant reductions in ischemic events in both coronary syndromes and stroke management .

What methodological limitations affect current understanding of eptifibatide's effects in specific patient populations?

Current eptifibatide research faces several methodological limitations when applied to specific populations:

  • Regional Variation Interpretation: The PURSUIT trial revealed significant efficacy differences between U.S. and non-U.S. populations, but lacked standardized variables to explain these variations comprehensively. This limits understanding of which population characteristics predict optimal treatment response .

  • Limited Representation in Clinical Trials: Pivotal studies like PURSUIT had inclusion criteria requiring specific ECG changes or biomarker elevations, potentially excluding patients with atypical presentations who might still benefit from treatment .

  • Co-medication Interaction Data Gaps: Insufficient research exists on interactions between eptifibatide and newer antiplatelet or anticoagulant medications, creating uncertainty in contemporary practice settings.

  • Special Population Characterization:

    • Limited pharmacokinetic/pharmacodynamic data in hepatic impairment

    • Insufficient characterization in elderly populations (>75 years)

    • Unknown effects during pregnancy (though labeled for hospital use only)

  • Mechanistic Understanding in Comorbid Conditions: Incomplete data on how diabetes, chronic kidney disease, or inflammatory conditions modify platelet responses to eptifibatide.

  • Pharmacogenomic Insights: Limited exploration of genetic polymorphisms affecting GPIIb/IIIa receptor density or function that might influence treatment efficacy.

  • Dosing Optimization: Current dosing (180 μg/kg bolus, 2.0 μg/kg/min infusion) derives from population-level optimization rather than individualized approaches based on platelet function or receptor density testing .

Addressing these limitations through targeted research would enhance precision in eptifibatide application across diverse patient populations.

How can researchers design studies to investigate potential expanded indications for eptifibatide?

Designing studies for expanded eptifibatide indications requires strategic methodological approaches:

  • Adaptive Trial Designs for Stroke Management: Building on promising results from the EPOCH trial (showing 91.3% vs. 81.5% successful recanalization with eptifibatide plus mechanical thrombectomy), researchers should implement adaptive designs that allow protocol modifications based on interim results, potentially accelerating clinical translation .

  • Enrichment Strategies: Identifying patient subgroups most likely to benefit based on biomarkers or clinical characteristics to enhance statistical power in smaller, more focused trials.

  • Surrogate Endpoint Validation: Establishing correlation between early imaging findings (e.g., reperfusion grades) and clinical outcomes to enable shorter trials with mechanistically relevant endpoints .

  • Combination Therapy Optimization: Factorial design studies examining eptifibatide with complementary antithrombotic or thrombolytic agents to identify synergistic combinations with favorable benefit-risk profiles.

  • Dose-Finding Methodologies: Implementing Bayesian adaptive dosing algorithms to optimize concentration-effect relationships for specific indications, potentially identifying lower effective doses with reduced bleeding risk.

  • Pragmatic Trial Frameworks: Embedding research in routine clinical practice through registry-based randomized trials to enhance generalizability and facilitate recruitment.

  • Targeted Population Selection:

    • High-risk TIA/minor stroke patients

    • Peripheral arterial intervention subjects

    • Venous thromboembolism scenarios resistant to standard approaches

The methodological foundation established in the EPOCH trial, using propensity matching and mixed-effects models to account for clinical heterogeneity, provides a template for these expanded indication studies .

What novel experimental approaches might address contradictions in current eptifibatide research data?

Resolving contradictions in eptifibatide research data requires innovative experimental strategies:

  • Single-Cell Analytics: Implementing high-dimensional flow cytometry and mass cytometry to characterize platelet subpopulations with differential eptifibatide responsiveness, potentially explaining heterogeneous clinical outcomes.

  • Systems Pharmacology Modeling: Developing comprehensive mathematical models integrating receptor binding, signaling cascades, and functional outcomes to reconcile apparently contradictory observations across different experimental systems.

  • Advanced Imaging Techniques:

    • Intravital microscopy to visualize eptifibatide effects on thrombus formation in real-time

    • Correlative light-electron microscopy to link ultrastructural changes with functional outcomes

  • Paradoxical Activation Investigation: Further exploring the mechanisms behind eptifibatide-induced thrombocytopenia and thrombosis, where patient antibodies paradoxically activate platelets in drug presence. This research requires ex vivo antibody detection assays and IgG depletion studies as demonstrated in previous investigations .

  • Parallel In Vitro System Comparison: Simultaneously evaluating eptifibatide effects in multiple model systems (washed platelets, PRP, whole blood) under identical conditions to identify system-specific artifacts versus genuine pharmacological effects.

  • Temporal Resolution Enhancement: Implementing high-speed analytical techniques to capture rapid, transient signaling events that might explain divergent downstream outcomes.

  • Multi-Omics Integration: Combining proteomics, metabolomics, and transcriptomics to create comprehensive molecular signatures of eptifibatide response, potentially identifying previously unrecognized mechanistic pathways.

These approaches could help resolve apparent contradictions, such as why eptifibatide sometimes shows differential effects on aggregation versus secretion pathways, or why clinical outcomes vary significantly across different patient populations despite similar pharmacokinetic profiles .

Product Science Overview

Mechanism of Action

Eptifibatide works by reversibly binding to the platelet receptor glycoprotein IIb/IIIa, which is crucial for platelet aggregation . By blocking this receptor, eptifibatide prevents the binding of fibrinogen, von Willebrand factor, and other adhesive ligands, thereby inhibiting platelet aggregation in a dose- and concentration-dependent manner .

Clinical Uses

Eptifibatide is primarily used to reduce the risk of acute cardiac ischemic events, such as death and myocardial infarction, in patients with unstable angina or non-ST-segment elevation myocardial infarction (NSTEMI) . It is often administered in conjunction with aspirin or clopidogrel and heparin (either low molecular weight or unfractionated) . Additionally, it is used as an adjunct to percutaneous coronary intervention (PCI) to prevent complications during and after the procedure .

Pharmacokinetics

Eptifibatide has a relatively short half-life of approximately 2.5 hours and is primarily excreted through the kidneys . It has a bioavailability of around 25% when administered intravenously .

Development and Approval

Eptifibatide was developed by Millennium Pharmaceuticals and is marketed under the brand name Integrilin . It has gained broad acceptance as the third inhibitor of glycoprotein IIb/IIIa, following the specific antibody abciximab and the non-peptide tirofiban .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.