Recombinant Human EPO-alpha/Fc Chimera protein (EPOFc) (Active)

Shipped with Ice Packs
In Stock

Description

Amino Acid Composition

The EPOFc fusion protein consists of two mature human EPO molecules (aa 28–193) linked to the Fc region of IgG1 (Pro100–Lys330). The EPO component retains its native tertiary structure with four alpha-helical bundles, while the Fc portion includes the CH2 and CH3 domains but excludes the CH1 domain .

ComponentAmino Acid RangeFunction
EPO (alpha subunit)28–193Binds EPOR, induces erythropoiesis
Fc fragment (IgG1)100–330Enhances stability, prolongs half-life

Molecular Mass and Glycosylation

  • Apparent Molecular Mass: ~140 kDa (non-reducing SDS-PAGE) .

  • Glycosylation: Contains N-linked glycans from both EPO and Fc domains. O-linked glycans may also contribute to glycosylation patterns .

Expression System

Produced in Chinese Hamster Ovary (CHO) cells via recombinant DNA technology, ensuring proper post-translational modifications such as glycosylation .

Purification and Quality Control

  • Purification: Proprietary chromatographic techniques (e.g., affinity, ion-exchange) .

  • Purity: >98% as confirmed by SDS-PAGE and HPLC .

  • Formulation: Lyophilized in PBS (pH 7.4) or sodium citrate buffer .

EPO-EPOR Interaction

EPO binds to the erythropoietin receptor (EPOR) on erythroid progenitor cells, triggering receptor dimerization and activation of Janus kinase 2 (JAK2). This initiates downstream signaling cascades:

  1. JAK2/STAT5 Pathway: Phosphorylation of STAT5, which translocates to the nucleus to upregulate pro-survival genes (e.g., Bcl-xL) .

  2. PI3K/AKT and MAPK Pathways: Promote cell proliferation and differentiation .

Role of the Fc Domain

The Fc fragment enhances:

  • Serum Half-Life: By binding to Fc receptors (e.g., FcRn), prolonging systemic exposure .

  • Stability: Reduces renal clearance and proteolysis .

In Vitro Assays

ParameterValueReference
ED50 (Cell Proliferation)<2.0 ng/mL
Specific Activity>5.0 × 10⁵ IU/mg
Target CellsHuman megakaryoblastic leukemia cells (e.g., UT-7)

Functional Specificity

  • EPOR Binding: High affinity (Kd ~100–200 pM) .

  • Hypoxia Sensitivity: Regulated by hypoxia-inducible transcription factors (HIF-1/2α) .

Preclinical Efficacy

StudyModelOutcomeReference
Reticulocyte StimulationRatsSingle-dose EPOFc increased peripheral reticulocyte count significantly
Half-Life ExtensionMice, PrimatesEPOFc showed prolonged half-life vs. native EPO; no immunogenicity observed
Anemia TreatmentChronic Kidney DiseaseImproved hemoglobin levels in primate models

Glycosylation’s Role

  • Sialic Acid Content: Higher sialylation correlates with reduced clearance and enhanced in vivo activity .

  • Receptor-Mediated Uptake: EPOR internalization accounts for ~40% of EPO degradation; EPOFc may resist this pathway .

Indications

ConditionPhaseStatusReference
Anemia in Chronic Kidney DiseasePhase 3NDA/BLA filed (CN)
Chemotherapy-Induced AnemiaPreclinicalInvestigational

Advantages Over Traditional EPO

  • Dosing Frequency: Reduced due to extended half-life .

  • Stability: Tolerates repeated freeze-thaw cycles with minimal loss of activity .

Pharmacokinetic Profile

ParameterValueReference
Half-LifeProlonged vs. native EPO
ClearanceReduced renal excretion

Product Specs

Buffer
Lyophilized from a 0.2µm filtered sodium citrate buffer (1 liter of ddH2O containing 5.9 g of sodium citrate, 5.8 g of sodium chloride, and 0.06 g of citric acid).
Form
Lyophilized powder
Lead Time
Typically, we can ship products within 5-10 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial before opening to ensure the contents settle to the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We suggest adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%, which can serve as a reference.
Shelf Life
Shelf life is influenced by various factors, including storage conditions, buffer components, temperature, and the inherent stability of the protein itself. Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. Lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
C-terminal hFc-tagged
Synonyms
EPOErythropoietin; Epoetin
Datasheet & Coa
Please contact us to get it.
Expression Region
28-193aa
Mol. Weight
45.3 kDa
Protein Length
Full Length of Mature Protein
Purity
>98% as determined by SDS-PAGE and HPLC.
Research Area
Cancer
Source
Mammalian cell
Species
Homo sapiens (Human)
Target Names
EPO
Uniprot No.

Target Background

Function
Erythropoietin is a hormone that plays a crucial role in regulating erythrocyte proliferation and differentiation, maintaining the physiological levels of circulating red blood cells. It binds to the erythropoietin receptor (EPOR), triggering EPOR dimerization and activation of JAK2, subsequently activating downstream effectors, including STAT1 and STAT3.
Gene References Into Functions
  1. Elevated plasma erythropoietin and erythropoietin receptor activation are implicated in the increase of plasma FGF23 in acute kidney injury. PMID: 29395333
  2. The alpha-7-nAChR-JAK-2/STAT-3-Nrf-2 signaling cascade is involved in the radiomitigative potential of EPO against ARS. PMID: 29220591
  3. Pro-inflammatory proteins S100A9 and tumor necrosis factor-alpha suppress erythropoietin production in myelodysplastic syndromes. PMID: 28983059
  4. EPO levels in the coronary artery disease (CAD) group were higher than those in the non-CAD group. A statistically significant correlation was observed between red cell distribution width and EPO levels among CAD patients. PMID: 28885393
  5. CD133(+) cells contribute to the local production of erythropoietin, as evidenced by the detection of circulating human erythropoietin. CD133(+) cells serve as an effective source for cell repair, capable of restoring renal functions, including erythropoietin release, and limiting long-term maldifferentiation and fibrosis. PMID: 27853265
  6. Circulating anti-EPO antibodies are detected in a significant proportion of treatment-naive HCV-infected patients and are independently associated with anemia, suggesting a further implication of autoimmunity in the pathophysiology of HCV-related anemia. PMID: 28603097
  7. The T allele of SNP rs60684937 located at 67,419,130 bp on chromosome 17 was associated with increased plasma EPO and a relatively increased expression of a non-coding transcript of PRKAR1A in sickle cell disease patients. PMID: 28173069
  8. This study describes a gain-of-function variant in EPO in an extended kindred with familial erythrocytosis, including 10 affected family members across four generations; this mutation, a single-nucleotide deletion (c.32delG), introduces a frameshift in exon 2. PMID: 29514032
  9. Using zebrafish, murine, and human models, this study reveals that erythropoietin (EPO) signaling, in conjunction with the GATA1 transcriptional target, AKAP10, regulates heme biosynthesis during erythropoiesis at the outer mitochondrial membrane. PMID: 28553927
  10. A decrease in central venous blood pressure prompts an increase in plasma EPO concentration independent of hemoconcentration, suggesting that CVP itself acts as an acute regulator of EPO synthesis. PMID: 27169519
  11. EPO (7q22) and SEC-61(7p11) emerged as new candidate genes susceptible to genetic losses, with 57.7% deletions identified in regions on chromosome 7. PMID: 27282568
  12. The current controversy may arise from a context-dependent mode of action of Epo, exhibiting opposite skeletal actions during bone regeneration and steady-state bone remodeling. PMID: 26822707
  13. High EPO expression is associated with monoclonal gammopathy of undetermined significance and multiple myeloma. PMID: 26919105
  14. Plasma levels of EPO at age 3 were found to be related to childhood asthma. PMID: 27434124
  15. EPO induces an EMT-like process in mammary non-tumorigenic epithelial cells. PMID: 28247960
  16. These results suggest that quercetin's cytoprotective effects in HepG2 cells are mediated via EPO production. PMID: 29080630
  17. Serum Epo and VEGF may serve as markers of the severity of hypoxia-ischaemia and brain injury due to their close association with hypoxic exposure. PMID: 27902983
  18. CIS interacts with phosphorylated EpoR at Y401, which is essential for the activation of STAT5 and ERK. PMID: 28038963
  19. EPO-dependent regulation pathway of FGF23 gene expression. PMID: 29073196
  20. Fetal plasma EPO concentrations are selectively increased in monochorionic twin pregnancies with intrauterine growth restriction. PMID: 27161360
  21. This study demonstrates that EPO is involved in the pathogenesis of sepsis-induced acute kidney injury. PMID: 27266727
  22. Erythropoietin surpasses the standard prognostic scores in predicting 28-day mortality in patients with acute-on-chronic liver failure. PMID: 27981303
  23. EPO levels were also found to be positively correlated with heme, TNF-alpha, IL-10, IP-10, and MCP-1 during cerebral malaria. PMID: 27441662
  24. Three single nucleotide polymorphisms are associated with an increased risk of diabetic retinopathy in a Chinese Han population. PMID: 27190272
  25. Pharmacokinetic animal studies revealed a significant 15.6-fold plasma half-life extension for the PASylated EPO (83.16 +/- 13.28 h) compared to epoetin alpha (8.5 +/- 2.4 h) and darbepoetin alpha (25.3 +/- 2.2h). PMID: 28168382
  26. Secreted MIR122 reached the kidney and reduced expression of erythropoietin, contributing to inflammation-induced anemia. PMID: 27477940
  27. This paper demonstrates that Epo can directly down-regulate pro-inflammatory T cell responses without affecting T cell activation status. PMID: 27208431
  28. These findings suggest that erythropoietin levels in anemia of unknown etiology, although elevated, remain inappropriately low, particularly when compared to other forms of anemia. This indicates a relative erythropoietin deficiency or a blunted erythroid cell response. PMID: 26747131
  29. Plasma IGFBP-1 was significantly associated with plasma EPO concentration in acute kidney injury, suggesting an unknown mechanism related to systemic stress conditions for EPO regulation in AKI. PMID: 26479890
  30. Our results suggest that the EPO/EPOR pathway promotes gastric cancer formation, proliferation, migration, and decreases apoptosis. PMID: 27086036
  31. These results indicate that both EpoR-positive and EpoR-negative cancer cells can be regulated by exogenous Epo. However, an increased response to erythropoietin was observed in EpoR-positive cells. Therefore, erythropoietin increases the risk of tumor progression in colon cancer and should not be used to treat anemia in this type of cancer. PMID: 27543111
  32. Overexpression of EPO is associated with clear cell renal cell carcinoma. PMID: 27468719
  33. EPO may play a significant role in stem cell mobilization by upregulating HGF in mesenchymal stem cells and inducing the migration of hematopoietic stem/progenitor cells. PMID: 27865586
  34. A review of contemporary aspects of EPO relating to chronic liver disease. [review] PMID: 26919118
  35. Hepatic EPO synthesis is not enhanced in cirrhosis. PMID: 26924722
  36. Conclusion: Anemia in cancers was not due to inadequate Epo or Fe levels, but rather due to an improper Epo response. PMID: 26838000
  37. In multivariate survival analysis, age, Epo, and EpoR were independent prognostic factors related to overall survival in hepatocellular carcinoma. PMID: 26097591
  38. This suggests that hypoxia prevents EPO suppression and exacerbates the plasma volume reduction induced by bed rest. PMID: 27081163
  39. Inadequate erythropoietin response may partly explain anemia in anorexia nervosa. PMID: 26049959
  40. These findings suggest that TGF-beta suppression and EPO stimulation promote erythropoiesis of CD34(+)CD31(+) progenitor cells derived from hPSCs. PMID: 26012423
  41. Our findings have important potential clinical implications, indicating that EPO supplementation in rhabdomyosarcoma patients may have the unwanted side effect of tumor progression. PMID: 26412593
  42. This suggests that rhEPO regulates apoptosis-related genes and affects apoptosis in the hippocampus of aging rats by upregulating SIRT. PMID: 26261574
  43. Higher levels of endogenous erythropoietin are associated with incident heart failure in older adults. PMID: 26721912
  44. Erythropoietin protects mouse renal tubular basement membrane by promoting bone marrow cells to generate and secrete miR-144, which, in turn, inhibits activation of the tPA/MMP9-mediated proteolytic network. PMID: 26469975
  45. This review describes the induction of erythropoietin gene expression in the liver, reproductive, and hemopoietic systems during hypoxia or a state of proliferation. PMID: 26995951
  46. Our data suggest that rs507392 and rs551238 in the erythropoietin gene likely act to lessen the risk for diabetic retinopathy (DR) in a Chinese cohort with type 2 diabetes mellitus (T2DM). PMID: 25675872
  47. Data suggest that maternal circulating 25-hydroxyvitamin D during mid-pregnancy and at delivery is inversely related to serum EPO; an indirect relation observed between circulating vitamin D and circulating hemoglobin is at least partly mediated by EPO. PMID: 26447159
  48. This review examines these different strategies and highlights the leading molecular recognition elements that have potential roles in rHuEPO doping detection. PMID: 25058943
  49. The addition of salt (even low concentrations of the strong chaotrope salt guanidinium hydrochloride) also exponentially decreased the initial rate of soluble erythropoietin non-native aggregation at 37 degrees C storage. PMID: 25628168
  50. This study investigated whether elevated perinatal erythropoietin (EPO) concentrations in very preterm infants are associated with increased risks of indicators of brain damage. PMID: 25793991

Show More

Hide All

Database Links

HGNC: 3415

OMIM: 133170

KEGG: hsa:2056

STRING: 9606.ENSP00000252723

UniGene: Hs.2303

Involvement In Disease
Microvascular complications of diabetes 2 (MVCD2)
Protein Families
EPO/TPO family
Subcellular Location
Secreted.
Tissue Specificity
Produced by kidney or liver of adult mammals and by liver of fetal or neonatal mammals.

Q&A

What is Recombinant Human EPO-alpha/Fc Chimera protein (EPOFc)?

Recombinant Human EPO-alpha/Fc Chimera protein (EPOFc) is a fusion protein consisting of human erythropoietin (EPO) conjugated to the Fc portion of human immunoglobulin G1 (IgG1). This chimeric protein combines the biological activity of EPO with the extended half-life and unique transport properties of the Fc domain. EPOFc can be produced in both monomeric and dimeric forms, with the monomeric form consisting of a single EPO molecule attached to a dimeric Fc, while the dimeric form contains two EPO molecules attached to a dimeric Fc . The fusion protein maintains the ability to bind to EPO receptors while gaining the additional capability to interact with the neonatal Fc receptor (FcRn), which facilitates transport across biological barriers and extends the protein's serum half-life .

How does the structure of EPOFc differ from standard recombinant EPO?

EPOFc differs structurally from standard recombinant EPO by the addition of the Fc domain of human IgG1. Typically, the N-terminus of the Fc region is fused to the C-terminus of the EPO molecule via a short linker sequence. In the case of monomeric EPOFc, the structure includes:

ComponentPositionDetails
Human EPON-terminusContains the active domain that binds to EPO receptor
LinkerMiddleShort peptide sequence (e.g., IEGRMD)
Human IgG1 FcC-terminusTypically Pro100-Lys330 region of IgG1

This chimeric structure enables EPOFc to maintain similar binding affinity to the EPO receptor as unconjugated EPO while gaining FcRn-binding capabilities, although with somewhat lower affinity for FcRn compared to full IgG1 .

What are the functional differences between monomeric and dimeric EPOFc?

Monomeric and dimeric EPOFc exhibit distinct functional properties that are important to consider in research applications:

Monomeric EPOFc (single EPO molecule + dimeric Fc):

  • Demonstrates enhanced pharmacokinetic properties compared to both dimeric EPOFc and unconjugated EPO

  • Shows bioavailability through pulmonary delivery approximately equal to subcutaneously delivered unconjugated EPO in humans

  • Offers potentially improved tissue penetration due to smaller size compared to dimeric form

  • Demonstrates more consistent absorption patterns in delivery studies

Dimeric EPOFc (two EPO molecules + dimeric Fc):

  • Potentially higher avidity for EPO receptor due to dual EPO molecules

  • May show different biodistribution patterns

  • Demonstrates different pharmacokinetic and pharmacodynamic properties compared to the monomeric form

The choice between monomeric and dimeric forms should be based on the specific research objectives and delivery routes being investigated .

What critical variables should researchers control when designing experiments with EPOFc?

When designing experiments involving EPOFc, researchers should carefully control several key variables to ensure valid and reproducible results:

  • Independent variables: Clearly define what you are manipulating (e.g., EPOFc dosage, administration route, timing of administration)

  • Dependent variables: Precisely specify what outcomes you are measuring (e.g., serum concentration, hematocrit levels, receptor binding)

  • Extraneous variables to control:

    • Storage conditions of EPOFc (temperature, freeze-thaw cycles)

    • Expression system used for protein production

    • Purification method consistency

    • Animal model characteristics (age, weight, sex, genetic background)

    • Administration technique standardization

    • Timing of measurements

  • Experimental treatments:

    • Include appropriate controls (vehicle-only, unconjugated EPO, non-functional EPOFc mutants)

    • Consider dose-response relationships

    • Account for potential species differences in receptor binding

  • Subject assignment:

    • Use randomization to minimize bias

    • Consider between-subjects or within-subjects designs based on your research question

    • Ensure adequate sample size through power analysis

The success of EPOFc experiments heavily depends on controlling these variables, particularly when comparing across different administration routes or protein variants .

How can researchers effectively assess EPOFc binding to FcRn versus EPO receptor?

Assessing EPOFc binding to different receptors requires specific methodological approaches:

For EPO receptor binding:

  • Cell-based proliferation assays: Use EPO-dependent cell lines like TF-1 to assess biological activity. The stimulation of TF-1 cell proliferation serves as a functional readout of EPO receptor binding and activation .

  • Direct binding assays: Compare binding affinity of EPOFc to the EPO receptor against standard unconjugated EPO (e.g., Epogen). Studies have shown that properly engineered EPOFc binds to the EPO receptor with affinity not significantly different from Epogen .

For FcRn binding:

  • Surface plasmon resonance: This technique can directly measure binding kinetics and affinity. Studies have shown that standard EPOFc binds FcRn, though with somewhat lower affinity than full IgG1 .

  • Mutational analysis: Creating mutant versions of EPOFc with substitutions in critical FcRn-binding residues (e.g., I253A, H310A, H435A) can serve as negative controls. These mutants typically show no detectable binding to FcRn in surface plasmon resonance assays .

  • pH-dependent binding assays: Since FcRn binding is pH-dependent, comparing binding at pH 6.0 versus pH 7.4 can help distinguish specific FcRn interactions.

A comprehensive assessment would include both functional readouts (e.g., half-life extension in vivo) and direct binding measurements to fully characterize the dual functionality of EPOFc .

What experimental design is optimal for comparing pulmonary versus subcutaneous delivery of EPOFc?

When comparing pulmonary versus subcutaneous delivery of EPOFc, the following experimental design elements are critical:

  • Study design: A crossover design where each subject receives both delivery methods (with appropriate washout periods) minimizes inter-subject variability .

  • Key controlled variables:

    • Equivalent deposited doses between routes

    • For pulmonary delivery: breathing pattern standardization (shallow vs. deep breathing affects deposition patterns)

    • For subcutaneous delivery: injection site consistency

    • Timing of administration and sampling

  • Pharmacokinetic measurements:

    • Primary endpoints: Peak serum concentrations (Cmax), area under the curve (AUC), half-life

    • Secondary endpoints: Time to peak concentration (Tmax), clearance rate, volume of distribution

    • Sampling schedule: More frequent early sampling to accurately capture absorption phase

  • Deposition confirmation for pulmonary delivery:

    • Radiolabeling of a portion of the test material

    • Gamma scintigraphy to visualize and quantify lung deposition

    • Analysis of peripheral-to-central deposition ratio (values around 0.4 suggest greater central airway deposition)

Example data from non-human primate studies comparing delivery routes:

Delivery MethodMean Peak Concentration (ng/ml)Mean AUC (ng·hr·ml⁻¹)Notes
Pulmonary (shallow breathing)800-900Not specified300 μg/kg deposited dose
Pulmonary (deep breathing)Significantly lower4,529 ± 900*300 μg/kg deposited dose
Subcutaneous (reference)Similar to shallow pulmonarySimilar to shallow pulmonaryBased on human Epo data

*Excluding one outlier animal with unusually high absorption

This design enables direct comparison of bioavailability between routes while controlling for critical variables that influence drug delivery efficiency.

How do mutations in the Fc domain affect EPOFc functionality and applications?

Mutations in the Fc domain of EPOFc can dramatically alter its functionality and are powerful tools for investigating mechanism of action and optimizing therapeutic properties:

  • FcRn binding mutations:

    • Triple mutation I253A/H310A/H435A completely abolishes FcRn binding

    • This mutant (EPOFc/IHH) shows no detectable interaction with FcRn in surface plasmon resonance assays

    • Serves as a critical control to demonstrate FcRn-dependent transport across biological barriers

    • Helps distinguish FcRn-mediated effects from passive diffusion or other transport mechanisms

  • Fc effector function modifications:

    • Mutations in the lower hinge and CH2 domain can reduce or eliminate binding to Fcγ receptors

    • LALA mutations (L234A/L235A) reduce unwanted immune cell activation

    • These modifications are valuable when studying EPOFc in inflammatory settings or where immune activation would confound results

  • Half-life modulating mutations:

    • Mutations like M428L/N434S (LS mutant) can enhance FcRn binding and potentially extend half-life

    • YTE mutations (M252Y/S254T/T256E) similarly enhance FcRn binding under acidic conditions

    • These variants allow researchers to study how extended circulation affects EPOFc efficacy

  • Glycosylation site modifications:

    • Altering N-glycosylation sites on either EPO or Fc portions affects protein stability and receptor interactions

    • N-glycan modifications can fine-tune pharmacokinetic and pharmacodynamic properties

    • These variants help elucidate the role of glycosylation in EPOFc functionality

The ability to generate these mutant variants makes EPOFc an excellent platform for structure-function studies and optimization of protein therapeutics .

What are the pharmacokinetic differences between monomeric EPOFc, dimeric EPOFc, and unconjugated EPO?

The pharmacokinetic profiles of these three erythropoietin variants differ significantly, with important implications for research applications:

ParameterMonomeric EPOFcDimeric EPOFcUnconjugated EPO
Half-lifeExtendedExtended but different from monomericShorter
Distribution volumeIntermediatePossibly more restrictedLarger
Clearance mechanismFcRn recycling dominantFcRn recycling dominantPrimarily renal filtration
Bioavailability (pulmonary)Similar to SC unconjugated EPOLower than monomericVery low without enhancers
Bioavailability (subcutaneous)Enhanced compared to unconjugatedNot directly compared~40% in humans
Receptor bindingMaintainedMaintained, possibly with avidity effectsReference standard

Key differences in absorption and distribution:

  • Monomeric EPOFc:

    • When delivered through lung using shallow breathing techniques targeting central airways, shows excellent bioavailability

    • Peak serum concentrations of 800-900 ng/ml observed at 300 μg/kg deposited dose in non-human primates

    • Absorption appears to be FcRn-dependent, as evidenced by studies with FcRn-binding mutants

  • Unconjugated EPO:

    • Rapidly cleared by kidneys

    • Poor bioavailability via pulmonary route

    • Requires more frequent dosing due to shorter half-life

The pharmacokinetic advantages of EPOFc are primarily mediated by FcRn interaction, which protects the protein from degradation and facilitates transport across biological barriers .

How can researchers address contradictions in experimental data when working with EPOFc?

When researchers encounter contradictory results in EPOFc studies, a systematic approach to addressing these contradictions includes:

  • Identify the specific contradiction type:

    • Contradictions between your findings and published literature

    • Internal contradictions between experiments in your own research

    • Contradictions between in vitro and in vivo results

    • Unexpected response variations between experimental subjects

  • Examine experimental variables:

    • Production batch variability (expression system, purification method)

    • Storage conditions and protein stability

    • Experimental subject variability (genetic background, age, health status)

    • Administration technique differences

    • Assay sensitivity and specificity issues

  • Statistical analysis approaches:

    • Identify outliers through statistical tests (e.g., the unusually high-absorbing monkey in deep breathing EPOFc studies)

    • Consider whether to include or exclude outliers with clear justification

    • Reanalyze data with and without outliers to understand their impact

    • Apply appropriate statistical tests for your experimental design

  • Mechanistic investigations:

    • Design experiments to test specific hypotheses about the contradiction

    • Use mutant variants (e.g., FcRn-binding mutants) to isolate mechanism-specific effects

    • Consider species differences in receptor binding or expression

    • Investigate time-dependent or dose-dependent effects that might explain contradictions

  • Documentation and transparency:

    • Report all contradictions openly in publications

    • Provide detailed methods to enable reproduction

    • Include all relevant data, even when it doesn't support your hypothesis

For example, in the EPOFc pulmonary delivery study, researchers encountered one monkey that absorbed significantly more EPOFc during deep breathing (>400 ng/ml peak and 24,120 ng·hr·ml⁻¹ AUC) compared to other animals in the same treatment group. They addressed this by: (1) reporting the outlier data, (2) analyzing results both with and without the outlier, and (3) providing a plausible mechanistic explanation (the possibility that more fusion protein was deposited in central airways rather than deep lung in this animal) .

What are the optimal production and purification methods for research-grade EPOFc?

The production and purification of high-quality EPOFc for research applications involves several critical steps and considerations:

  • Expression system selection:

    • Chinese Hamster Ovary (CHO) cells are the preferred mammalian expression system

    • CHO DG44 cells specifically have been successfully used for EPOFc production

    • Mammalian expression ensures proper glycosylation and folding of both EPO and Fc domains

  • Transfection and selection strategy for monomeric EPOFc:

    • Co-transfection approach using two plasmids:
      a) pEDdC.natEpoFc (encoding EPO-Fc fusion)
      b) pcDNA3.1/FLAGFc (encoding FLAG-tagged Fc)

    • Selection using dual markers (methotrexate and G418)

    • This strategy produces three protein products that can be separated: EpoFc/EpoFc homodimer, EpoFc/FLAG-Fc heterodimer (monomeric EPOFc), and FLAG-Fc/FLAG-Fc homodimer

  • Multi-step purification protocol:

    • Initial capture: Protein A chromatography exploiting the Fc domain

    • Intermediate purification: Size exclusion chromatography to separate monomeric and dimeric forms

    • Polishing step for monomeric form: Immunoaffinity chromatography using anti-FLAG-Sepharose

    • Final formulation in appropriate buffer and storage at -80°C

  • Quality control assessments:

    • SDS-PAGE and Western blotting to confirm size and identity

    • Surface plasmon resonance to verify FcRn binding

    • Cell-based assays (e.g., TF-1 proliferation) to confirm EPO receptor binding and biological activity

    • Endotoxin testing to ensure safety for in vivo applications

This methodological approach enables the production of well-characterized monomeric EPOFc with defined purity and functionality for research applications.

How should researchers measure and compare the biological activity of EPOFc variants?

Comprehensive assessment of EPOFc biological activity requires multiple complementary approaches:

  • In vitro receptor binding assays:

    • Direct measurement of EPO receptor binding affinity compared to reference standards

    • Surface plasmon resonance to determine binding kinetics (kon, koff) and equilibrium dissociation constant (KD)

    • FcRn binding assays at both pH 6.0 and pH 7.4 to characterize pH-dependent interaction

  • Cell-based functional assays:

    • TF-1 cell proliferation assay: EPOFc stimulates proliferation of this EPO-dependent cell line

    • Determination of EC50 values (typically 1-4 ng/mL for standard EPO)

    • Comparison of potency relative to reference standards (e.g., Epogen)

    • Assessment of dose-response relationships

  • Pharmacokinetic measurements in animal models:

    • Serum concentration over time following different routes of administration

    • Calculation of standard PK parameters: Cmax, Tmax, AUC, t1/2

    • Comparison between different variants and administration routes

    • Use of radiolabeled protein to track biodistribution

  • Pharmacodynamic readouts:

    • Reticulocyte count increase

    • Hematocrit and hemoglobin changes

    • Bone marrow response

    • Correlation between PK parameters and PD effects

  • Specialized activity assessments for modified variants:

    • For FcRn-binding mutants: Compare transport across biological barriers

    • For glycosylation variants: Assess stability and receptor binding

    • For fragment variants: Evaluate activity relative to molecular size

When comparing variants, researchers should normalize data to protein concentration, use appropriate statistical analyses, and include reference standards in each experiment to account for inter-assay variability.

What administration techniques are optimal for studying pulmonary delivery of EPOFc?

Studying pulmonary delivery of EPOFc requires specialized techniques to ensure consistent and measurable lung deposition:

  • Controlled breathing parameters:

    • Shallow breathing techniques target central airways, which have higher FcRn expression

    • Deep breathing targets alveoli, which have different absorption characteristics

    • Using ventilator-controlled breathing in animal models ensures consistency

    • Specific breathing patterns should be defined by tidal volume, respiratory rate, and inspiratory/expiratory ratios

  • Aerosol generation and characterization:

    • Particle size is critical: 3-5 μm mass median aerodynamic diameter typically targets conducting airways

    • Characterize aerosol properties before administration

    • Measure and report aerosol concentration and protein stability after aerosolization

    • Document nebulizer type and operating parameters

  • Deposition confirmation and quantification:

    • Radiolabel a portion of EPOFc to track deposition without affecting biological activity

    • Use gamma scintigraphy to visualize and quantify lung deposition patterns

    • Calculate the peripheral-to-central ratio to characterize regional deposition

    • In non-human primate studies, a peripheral-to-central ratio of 0.4 ± 0.2 suggested greater central airway deposition

  • Dose calculation and normalization:

    • Calculate lung-deposited dose rather than nominal dose

    • For comparison between routes, normalize pharmacokinetic data to the actual deposited dose

    • Report both the nominal dose in the nebulizer and the estimated lung-deposited dose

  • Sampling protocol optimization:

    • Collect both serum samples (for PK) and bronchoalveolar lavage samples (for local concentration)

    • Use appropriate sampling frequency to capture absorption phase

    • Consider collecting tissue samples at experimental endpoints to assess local distribution

Using these specialized techniques, researchers have demonstrated that monomeric EPOFc delivered to the lungs during shallow breathing can achieve bioavailability approximately equal to subcutaneous administration of unconjugated EPO in humans, highlighting the potential of this administration route .

What are common pitfalls in EPOFc research and how can they be addressed?

Researchers working with EPOFc may encounter several challenges that can affect experimental outcomes:

  • Protein stability issues:

    • Pitfall: Loss of activity during storage or handling

    • Solution: Store at -80°C, minimize freeze-thaw cycles, verify activity before experiments

    • Use freshly prepared working solutions for critical experiments

  • Variable pulmonary deposition:

    • Pitfall: Inconsistent dosing in pulmonary delivery studies

    • Solution: Use radiolabeled protein to verify deposition, standardize breathing parameters

    • Report both nominal and estimated deposited doses

    • Account for individual variation in deposition efficiency

  • Fc-mediated effects beyond FcRn binding:

    • Pitfall: Unexpected immune activation or off-target effects

    • Solution: Include appropriate Fc-only controls

    • Consider using Fc variants with reduced effector functions for certain applications

    • Monitor for immune responses in animal studies

  • Species differences in receptor binding:

    • Pitfall: Variable response between animal models

    • Solution: Characterize binding to species-specific receptors before in vivo studies

    • Consider potential differences in FcRn expression and distribution

    • Validate key findings across multiple species when possible

  • Contradictory results from different experimental approaches:

    • Pitfall: In vitro and in vivo results may not align

    • Solution: Use multiple complementary assays to build a comprehensive understanding

    • Identify potential methodological limitations in each approach

    • Design experiments that can reconcile contradictions

  • Sample analysis artifacts:

    • Pitfall: Inaccurate quantification due to interfering substances or matrix effects

    • Solution: Validate analytical methods in relevant matrices

    • Include appropriate calibration standards and quality controls

    • Consider using orthogonal detection methods for critical measurements

Addressing these common pitfalls requires careful experimental design, appropriate controls, and thorough validation of methods before conducting large-scale studies with EPOFc.

What are the current limitations in understanding EPOFc mechanism of action?

Despite significant advances in EPOFc research, several knowledge gaps and limitations remain:

Addressing these limitations will require combining advanced structural biology techniques, refined animal models, and eventually carefully designed human studies to fully understand and optimize EPOFc therapeutics.

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.