EPO a Fc Human

Erythropoietin-Alpha Fc-Chimera Human Recombinant
Shipped with Ice Packs
In Stock

Description

Mechanism of Action

  • Erythropoiesis Stimulation: Binds homodimeric erythropoietin receptors (EpoR) on erythroid progenitors, activating JAK2/STAT5, PI3K/AKT, and MAPK pathways to promote RBC production .

  • FcRn-Mediated Recycling: The Fc domain binds FcRn in acidic endosomes, redirecting the protein to circulation instead of lysosomal degradation, yielding a half-life of 29.5–43.5 hours in primates (vs. 4–12 hours for native EPO) .

Table 1: Pharmacokinetics in Animal Models

SpeciesDose (µg/kg)Half-life (h)Bioavailability
Rhesus monkey8–8029.5–38.9≈40% (SC)
Rat16–16035.5–43.560–70% (SC)

Table 2: In Vitro Bioactivity

AssayED₅₀Receptor Binding (Kd)
TF-1 cell proliferation<2.0 ng/mL0.28–0.30 nM

Therapeutic Efficacy in Anemia Models

  • Rodent Studies: Weekly subcutaneous doses (7.5–30 µg/kg) restored RBC counts, hemoglobin, and hematocrit to baseline levels in irradiation- and chemotherapy-induced anemia .

  • Primate Studies: In renal ablation models, 5–10 µg/kg weekly matched the efficacy of thrice-weekly native EPO (5.4 µg/kg), with hematocrit stabilization within 4 weeks .

Production and Stability

  • Expression System: Produced in Chinese hamster ovary (CHO) cells, ensuring proper glycosylation and folding .

  • Formulation: Lyophilized powder stable at -18°C; reconstituted in PBS (≥100 µg/mL) retains activity for 2–7 days at 4°C .

Clinical and Regulatory Considerations

  • Advantages: Reduced dosing frequency, improved patient compliance, and comparable efficacy to existing ESAs (e.g., epoetin alfa) .

  • Detection Challenges: Anti-doping methods require specialized assays (e.g., SDS-PAGE/immunoblotting) due to altered glycosylation and molecular weight .

Emerging Applications

  • Neuroprotection: Preclinical studies suggest EPO-Fc mitigates neural injury via anti-apoptotic signaling, though clinical validation is pending .

  • Transgenic Production: Novel platforms (e.g., transgenic chickens) enable scalable, cost-effective synthesis with human-like glycosylation .

Product Specs

Introduction
As a member of the EPO/TPO family, this gene encodes a secreted, glycosylated cytokine composed of four alpha helical bundles. This protein is present in the plasma and plays a crucial role in regulating red blood cell production. It achieves this by promoting erythroid differentiation and initiating hemoglobin synthesis. Additionally, this protein exhibits neuroprotective properties against various potential brain injuries and demonstrates antiapoptotic functions in several tissue types.
Description
Erythropoietin-alpha Fc-Chimera Human Recombinant, produced in Chinese hamster ovary (CHO) cells using recombinant DNA technology, is a dimeric, glycosylated polypeptide chain. It consists of two mature human EPO molecules linked to the Fc portion of human IgG1. The Fc component includes the CH2 domain, the CH3 domain, and the hinge region, but excludes the CH1 domain of IgG1. Due to glycosylation, the recombinant protein exhibits an apparent molecular mass of 140 kDa in non-reducing SDS-PAGE.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The lyophilized powder is formulated with 1x PBS pH-7.4 at a concentration of 1 mg per mg of powder.
Solubility
To reconstitute the lyophilized Erythropoietin, it is recommended to dissolve it in sterile 18MΩ-cm H2O at a concentration of not less than 100 µg/ml. This solution can then be further diluted in other aqueous solutions as needed.
Stability
Lyophilized Erythropoietin-a, although stable at room temperature for 3 weeks, should be stored in a dry environment below -18°C. Once reconstituted, EPO-alpha should be stored at 4°C for short-term use (2-7 days) and below -18°C for future use. To ensure long-term stability, adding a carrier protein (0.1% HSA or BSA) is recommended. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined using two methods and exceeds 98.0%: (a) Analysis by RP-HPLC. (b) Analysis by SDS-PAGE.
Biological Activity
The ED50, determined by the dose-dependent stimulation of human megakaryoblastic leukemia cells, is less than 2.0 ng/ml. This corresponds to a Specific Activity of 5.0 x 105 IU/mg.
Synonyms
EPO-a, EPO-alpha, Epoetin, EP, MGC138142.
Source
Chinese Hamster Ovary Cells(CHO).

Q&A

What is EPO-Fc and how does it differ structurally from standard erythropoietin?

EPO-Fc is a recombinant fusion protein consisting of human erythropoietin (EPO) linked to the Fc region of human immunoglobulin. Specifically, in typical constructs, the extracellular domain of human EPO (amino acids 30-193) is fused to the N-terminus of the Fc region of a human immunoglobulin, such as a mutant IgG1 . Unlike standard EPO, this fusion provides extended serum half-life and potentially improved pharmacological properties. The design maintains EPO's receptor-binding domain while leveraging the Fc region to enhance circulation time through neonatal Fc receptor (FcRn) recycling mechanisms.

What expression systems are typically used for EPO-Fc production in laboratory settings?

For research-grade EPO-Fc production, Chinese Hamster Ovary (CHO) cells are the predominant expression system . This mammalian cell line provides appropriate post-translational modifications, particularly glycosylation patterns that are essential for EPO's biological activity. CHO cells ensure proper folding of the complex fusion protein structure and facilitate secretion of the correctly assembled fusion protein with appropriate glycosylation, which is critical for both the EPO and Fc components of the molecule.

What is the biological significance of the "non-lytic" property in EPO-Fc constructs?

The "non-lytic" property of certain EPO-Fc constructs refers to specific mutations introduced in the Fc region that eliminate effector functions while preserving FcRn binding. These mutations to the complement (C1q) and FcγR I binding sites render the fusion proteins incapable of antibody-directed cytotoxicity (ADCC) and complement-directed cytotoxicity (CDC) . This engineering approach creates a long-lasting fusion protein that binds to the EPO receptor without triggering unwanted immune destruction mechanisms, providing a cleaner pharmacological profile focused solely on erythropoietic effects.

How do the pharmacokinetic profiles of EPO-Fc compare to unmodified EPO?

EPO-Fc demonstrates significantly extended half-life compared to unmodified EPO. Pharmacokinetic studies in animal models have shown that the serum half-life of rhEPO-Fc ranges from 29.5 to 38.9 hours at doses of 8-80 μg/kg in rhesus monkeys and 35.5 to 43.5 hours at doses of 16-160 μg/kg in rats . This represents a substantial improvement over standard EPO formulations. Furthermore, advanced constructs like EPO-hyFc(H) with high sialic acid content show approximately double the serum half-life compared to hyperglycosylated EPO formulations like darbepoetin alfa .

What animal models are appropriate for evaluating EPO-Fc efficacy?

Multiple animal models have been validated for assessing EPO-Fc efficacy:

  • Irradiation-induced anemia models

  • Chemotherapy-induced anemia models (e.g., cyclophosphamide)

  • Partial renal ablation models

  • Cisplatin-induced anemia models

These models provide diverse pathophysiological backgrounds for evaluating EPO-Fc function across different anemic conditions. Researchers should select models based on their specific research questions, with consideration for species-specific differences in EPO receptor sensitivity and pharmacokinetics.

How do different Fc domain selections impact EPO-Fc functionality?

The selection of Fc domain significantly impacts the functionality and pharmacological profile of EPO-Fc fusion proteins:

  • IgG1 Fc: While providing extended half-life, it can exhibit unwanted antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), potentially limiting its utility for therapeutic applications .

  • IgG2 Fc: Selected in some constructs specifically because it does not bind to FcγR, reducing immune-mediated effects while still extending half-life through FcRn interaction .

  • Hybrid Fc (hyFc): Consisting of IgD and IgG4 components, this construct maintains the "Y-shaped" structure despite low amino acid homology (20.5%) between IgD Fc and IgG4 Fc. It cannot bind to FcγR I and C1q (unlike EPO-IgG1 Fc), eliminating cytotoxicity concerns while providing superior bioactivity compared to EPO-IgG1 Fc constructs .

The flexibility of the IgD component in hyFc constructs appears to contribute to better preservation of EPO bioactivity compared to more rigid IgG1-based constructs.

What methodological approaches exist for detecting EPO-Fc in research and anti-doping settings?

Detection of EPO-Fc presents unique challenges compared to standard EPO:

  • EPO-Fc cannot be directly detected by isoelectric focusing polyacrylamide gel electrophoresis (IEF-PAGE), the standard method for EPO detection .

  • Alternative methods required include sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) or sarcosyl polyacrylamide gel electrophoresis (SAR-PAGE) .

  • Detection typically reveals two characteristic bands: a strong band representing the monomer and a weaker band for the dimer .

  • Current methodological limitations include lack of consensus on whether both bands must be present for positive identification, as human administration studies are not available .

Researchers working with EPO-Fc should develop specialized protocols with appropriate controls to ensure accurate detection and quantification.

What are the comparative advantages of different EPO-Fc engineering strategies?

Different engineering approaches to EPO-Fc development offer distinct advantages:

Engineering ApproachKey FeaturesAdvantagesConsiderations
Non-mutated EPO with modified IgG2 FcPreserves native EPO structure; Fc mutations reduce CDC/ADCCMaintains full erythropoietic activity; Reduced immunogenicity riskMay have suboptimal flexibility at receptor binding interface
Hybrid Fc (hyFc) consisting of IgD and IgG4Creates highly flexible fusion proteinBetter in vitro and in vivo bioactivity than EPO-IgG1 Fc; No detectable antibody response in repeated administrationsRequires complex molecular engineering
High sialic acid content modifications (EPO-hyFc(H))Increases terminal sialic acid residues~2× longer half-life than darbepoetin alfa; Superior hemoglobin elevation in both normal and anemic ratsRequires specialized glycoengineering techniques

Research indicates that the hyFc-fusion strategy more effectively improves EPO's in vivo bioactivity compared to the hyperglycosylation approach used in darbepoetin alfa .

How does the pharmacodynamic profile of EPO-Fc differ across administration routes?

The route of administration significantly impacts EPO-Fc pharmacodynamics:

  • Subcutaneous administration: Provides sustained release with delayed Tmax (time to maximum concentration) and potentially twice the final area-under-curve (AUClast) compared to other EPO formulations . This results in more gradual onset but potentially more sustained erythropoietic effects.

  • Intravenous administration: Yields more immediate bioavailability but potentially shorter duration of action despite the extended half-life compared to standard EPO.

The pharmacodynamic differences translate to distinct patterns of reticulocyte production, followed by increases in red blood cell (RBC) count, hemoglobin, and hematocrit levels. These parameters should be monitored at appropriate intervals based on the administration route to accurately assess efficacy .

What considerations are important for dose optimization in EPO-Fc experimental design?

Dose optimization for EPO-Fc studies requires careful consideration of several factors:

  • Dose-response relationships: Studies demonstrate dose-dependent increases in reticulocyte levels followed by RBC increases across multiple species at dose ranges of:

    • 7.5–30.0 μg/kg in mice

    • 5.4–21.4 μg/kg in rats

    • 5.0–10.0 μg/kg in rhesus monkeys

  • Adjustment for molecular weight: EPO-Fc has significantly higher molecular weight than native EPO, requiring dose calculations based on molar equivalents rather than simple weight-based comparisons.

  • Species-specific sensitivity: Consideration of species differences in EPO receptor binding and downstream signaling is critical, as is accounting for potential differences in clearance mechanisms.

  • Administration frequency: The extended half-life permits less frequent dosing (e.g., weekly instead of three times weekly), which should be incorporated into study designs .

  • Comparator selection: When benchmarking against other long-acting EPO formulations (e.g., darbepoetin alfa), matching of molar concentrations rather than weight-based dosing provides more scientifically valid comparisons .

What analytical methods are recommended for characterizing EPO-Fc purity and integrity?

For rigorous characterization of EPO-Fc preparations, researchers should employ:

  • SDS-PAGE: To assess purity (≥98% purity is achievable) and to detect both monomeric and potential dimeric forms .

  • Endotoxin testing: Using methods such as the Limulus Amebocyte Lysate (LAL) test to ensure preparations contain <0.06EU/μg protein .

  • Glycan analysis: To characterize the glycosylation pattern, particularly for constructs where sialic acid content is engineered.

  • Size exclusion chromatography: To detect aggregates and confirm proper assembly of the fusion protein.

  • Biological activity assays: Using dose-dependent stimulation of human megakaryoblastic leukemia cells to confirm functional receptor binding and signaling .

How should stability studies be designed for EPO-Fc research preparations?

EPO-Fc stability studies should account for multiple storage and handling scenarios:

  • Long-term storage: EPO-Fc remains stable for at least 1 year when stored at -20°C .

  • Working solutions: Aliquots maintain stability for up to 3 months at -20°C .

  • Reconstitution protocols: Lyophilized preparations should be reconstituted at 100μg/ml in sterile PBS after centrifuging the vial before opening .

  • Handling procedures: Freeze/thaw cycles should be minimized as they can impact structural integrity and bioactivity .

Stability-indicating assays should monitor both structural integrity and functional activity at defined time points to establish evidence-based handling protocols.

What methodological approaches can address the detection challenges of EPO-Fc in research samples?

To overcome detection challenges:

  • Combined electrophoretic approaches: Utilizing both SDS-PAGE and SAR-PAGE to enhance detection specificity.

  • Antibody selection: Employing antibodies targeting either the EPO domain or the junction between EPO and Fc to distinguish from endogenous EPO.

  • Mass spectrometry: Implementing peptide mapping strategies to identify unique junction peptides between EPO and Fc domains.

  • Reference standards: Developing well-characterized reference standards that include both monomeric and dimeric forms for calibrating detection methods .

  • Western blotting optimization: Utilizing specialized transfer and detection conditions optimized for high molecular weight glycoproteins.

Since no approved EPO-Fc pharmaceuticals are currently available, and human administration data is lacking, researchers should include appropriate positive and negative controls in all analytical procedures .

How should researchers interpret comparative efficacy data between EPO-Fc and other erythropoiesis-stimulating agents?

When interpreting comparative efficacy:

  • Normalize for molecular differences: Account for the significantly higher molecular weight of EPO-Fc compared to standard EPO when comparing dose-response relationships.

  • Consider pharmacokinetic/pharmacodynamic relationships: More extended half-life may produce different temporal patterns of response that require longer observation periods.

  • Evaluate multiple parameters: Assess not only reticulocyte responses but subsequent RBC, hemoglobin, and hematocrit changes to fully characterize erythropoietic effects .

  • Account for model-specific factors: Different animal models of anemia (e.g., irradiation-induced versus renal impairment) may show varying responsiveness based on the underlying pathophysiology .

  • Apply appropriate statistical approaches: Time-series analyses and area-under-curve evaluations often provide more insight than single time-point comparisons for long-acting agents.

What are the key considerations for translating EPO-Fc research findings toward potential clinical applications?

Key translational considerations include:

  • Immunogenicity assessment: Despite engineering to reduce immunogenicity, comprehensive evaluation of antibody responses against both the EPO and Fc components remains essential .

  • Comparative risk-benefit profile: Assessment against established erythropoiesis-stimulating agents with consideration of potential advantages in dosing frequency and magnitude of response .

  • Target population selection: Identification of patient populations most likely to benefit from extended half-life formulations (e.g., chronic kidney disease patients with stable anemia versus acute anemia conditions).

  • Regulatory considerations: Understanding the additional characterization requirements for fusion proteins compared to modified versions of native proteins.

  • Manufacturing scalability: Evaluation of whether laboratory-scale production methods can be adapted to meet clinical-grade manufacturing requirements while maintaining critical quality attributes.

What emerging approaches might further enhance EPO-Fc design and functionality?

Several promising research directions may advance EPO-Fc technology:

  • Site-specific conjugation: Exploring engineered attachment sites for the Fc domain to optimize receptor interaction while minimizing steric hindrance.

  • Glycoengineering: Further refinement of glycosylation patterns to enhance both half-life and bioactivity simultaneously.

  • Multi-functional fusion constructs: Development of EPO-Fc variants that incorporate additional functional domains to address comorbidities in anemic patients.

  • Tissue-targeted variants: Engineering EPO-Fc constructs with additional targeting moieties to enhance delivery to specific tissues or cell populations.

  • Biosimilar development methodologies: Establishing analytical frameworks for comparing innovator and biosimilar EPO-Fc constructs as this technology advances toward clinical applications.

Product Science Overview

Structure and Function

Erythropoietin-Alpha Fc-Chimera (Human Recombinant), often abbreviated as EPO-α/Fc, is a recombinant form of EPO that has been engineered to include the Fc region of human IgG1. This fusion enhances the stability and half-life of the protein in the bloodstream . The Fc component contains the CH2 domain, the CH3 domain, and the hinge region, which contribute to the extended half-life by interacting with the neonatal Fc receptor (FcRn) and protecting the protein from lysosomal degradation .

The recombinant EPO-α/Fc is produced in Chinese hamster ovary (CHO) cells, which are commonly used in biotechnology for the production of therapeutic proteins due to their ability to perform complex post-translational modifications, including glycosylation . Glycosylation is essential for the biological activity and stability of EPO, as it affects the protein’s solubility, resistance to proteolysis, and receptor binding affinity .

Therapeutic Applications

Recombinant human EPO and its analogs, including EPO-α/Fc, are widely used as erythropoiesis-stimulating agents (ESAs) in the treatment of anemia associated with chronic kidney disease (CKD), cancer, and other conditions . By stimulating erythropoiesis, these agents help to increase red blood cell counts and improve oxygen delivery to tissues, thereby alleviating the symptoms of anemia .

In addition to its role in erythropoiesis, EPO has been found to have neuroprotective and anti-apoptotic effects in various tissues . These properties have sparked interest in the potential use of EPO and its derivatives in the treatment of neurological disorders and other conditions involving cell death and tissue damage .

Production and Purification

The production of recombinant EPO-α/Fc involves the transfection of CHO cells with the human EPO gene or cDNA linked to an expression vector . The transfected cells are then cultured, and the recombinant protein is secreted into the culture medium. The protein is subsequently purified using techniques such as affinity chromatography, which exploits the Fc region’s affinity for protein A or protein G, commonly used ligands in purification processes .

The purified EPO-α/Fc is typically lyophilized (freeze-dried) for stability and ease of storage. It is recommended to reconstitute the lyophilized protein in sterile water and store it at low temperatures to maintain its stability and biological activity .

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.