Anti-Human IFNy (Emapalumab) – Fc Muted™

Anti-Human IFNy (Emapalumab) – Fc Muted™

Product No.: LT2905

- -
- -
Product No.LT2905
Clone
NI-0501
Target
IFNγ
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Emapalumab,NI-0501,emapalumab-lzsg,IFNG
Isotype
Human IgG1 L2
Applications
ELISA
,
FA
,
FC
,
IP
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Recommended Isotype Controls
Immunogen
Human IFNγ
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
< 1.0 EU/mg as determined by the LAL method
Purity
≥95% by SDS Page
≥95% monomer by analytical SEC
Formulation
This biosimilar antibody is aseptically packaged and formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.2 - 7.4 with no carrier protein, potassium, calcium or preservatives added. Due to inherent biochemical properties of antibodies, certain products may be prone to precipitation over time. Precipitation may be removed by aseptic centrifugation and/or filtration.
State of Matter
Liquid
Product Preparation
Recombinant biosimilar antibodies are manufactured in an animal free facility using only in vitro protein free cell culture techniques and are purified by a multi-step process including the use of protein A or G to assure extremely low levels of endotoxins, leachable protein A or aggregates.
Storage and Handling
Functional grade preclinical antibodies may be stored sterile as received at 2-8°C for up to one month. For longer term storage, aseptically aliquot in working volumes without diluting and store at ≤ -70°C. Avoid Repeated Freeze Thaw Cycles.
Regulatory Status
Research Use Only (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Additional Applications Reported In Literature ?
ELISA
WB
IP
FA
FC
N
IF Microscopy
Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change.

Description

Description

Specificity
This non-therapeutic biosimilar antibody uses the same variable region sequence as the therapeutic antibody Emapalumab. This product is for research use only. Emapalumab activity is directed against human IFNγ.
Background
IFNγ plays roles in Th1 differentiation, macrophage function, leukocyte migration to sites of infection, and increasing major histocompatibility complex expression to improve T-cell recognition of infected or malignant cells 1. Hemophagocytic lymphohistiocytosis (HLH) is a rare but severe dysregulation of the immune system characterized by increased IFNγ production, macrophage and lymphocyte hyperactivity with tissue infiltration, hypersecretion of pro-inflammatory cytokines (CXCL9), hemophagocytosis, tissue damage, and multi-organ failure 2. IFNγ plays a central role in the pathophysiology of HLH, and blocking IFNγ leads to clinical improvement.

Emapalumab was developed by Novimmune and Swedish Orphan Biovitrum as an immunotherapeutic treatment for HLH 2. Emapalumab is a fully human IgG1 monoclonal antibody that targets and binds to IFNγ with high affinity. Emapalumab neutralizes IFNγ activity and inhibits interaction with its receptor by acting as a non-competitive inhibitor binding to free IFNγ and IFNγ-Receptor-1(IFNγR1)-bound IFNγ. Emapalumab inhibits receptor dimerization and transduction of interferon-γ signaling, impairing the interaction induced by IFNγ at the cell surface with IFNγR1 and IFNγ R2 and thereby neutralizing IFNγ biologic activity 2,3,4. Emapalumab prevents recruitment of IFNγR2 but has no effect on IFNγR1 endocytosis and internalization into lysosomes 2,4. In HLH patients, emapalumab reduces the plasma concentrations of the cytokine CXCL9.

Emapalumab is composed of anti-(human IFNγ) human monoclonal NI-0501 heavy chain, disulfide with human monoclonal NI-0501 light chain, dimer 2. Emapalumab is produced by recombinant DNA technology and is approximately 148 kDa 5.
Antigen Distribution
IFNγ is produced by natural killer and natural killer T cells, T-helper 1 (Th1) CD4 + T cells, and CD8 + and cytotoxic T-lymphocytes.
Ligand/Receptor
IFNAR
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Immunology
.
Other Molecules

Leinco Antibody Advisor

Powered by AI: AI is experimental and still learning how to provide the best assistance. It may occasionally generate incorrect or incomplete responses. Please do not rely solely on its recommendations when making purchasing decisions or designing experiments.

Research-grade emapalumab biosimilars are utilized as calibration standards and reference controls in pharmacokinetic bridging ELISAs through a carefully designed bioanalytical strategy that ensures accurate quantification of drug concentrations in serum samples.

Single Assay Methodology for Biosimilar Quantification

The current industry consensus favors developing a single PK assay using a single analytical standard for quantitative measurement of both biosimilar and reference products. This approach offers significant advantages by decreasing inherent variability that would be associated with running multiple methods and eliminating the need for crossover analysis when conducting blinded clinical studies.

Bioanalytical Comparability Assessment

Before implementing a single assay method, a comprehensive testing strategy must be executed to evaluate bioanalytical comparability. The process begins with a robust method qualification study that generates precision and accuracy data sets for both the biosimilar and reference products. Statistical analysis is then applied to determine if the test products are bioanalytically equivalent within the method.

The evaluation of analytical equivalence involves comparing the 90% confidence interval to pre-defined equivalence intervals [0.8, 1.25]. Bioanalytical equivalence is concluded by combining the totality of evidence, which provides stringent criteria around the measurement of test products within the assay to minimize confounding variability.

Validation Protocol and Standards Preparation

Once bioanalytical comparability is established, method validation proceeds using the biosimilar as the analytical standard for the single method. In the validation process, nine independent sets of biosimilar standards are prepared in human serum at nominal concentrations of 50, 100, 200, 400, 800, 1600, 3200, 6400, and 12800 ng/mL.

Additionally, two independent sets of validation samples are prepared using the biosimilar, FDA-licensed reference, and EU-authorized reference products in human serum at concentrations of 50, 150, 1250, 9600, and 12800 ng/mL. These samples are then quantified against the biosimilar standard curve during validation studies conducted across nine assays performed over three days by three different analysts.

Specialized Considerations for Emapalumab

For emapalumab specifically, special attention must be paid to the lower limit of quantification (LLOQ) requirements. Validated electrochemiluminescence immunoassays typically have an LLOQ of 50 pg/mL for relevant biomarkers. A critical procedural consideration involves pre-incubating serum samples with an excess of emapalumab (50 μg/mL) to push the equilibrium towards the bound form in all samples, thereby avoiding bias due to dissociation of the emapalumab-IFNγ complex during the bioanalysis process.

Quality Control Framework

The bioanalytical strategy requires that methods employed must be equally precise, accurate, and robust in measuring biosimilar and reference products sourced from different regions. This comprehensive approach with scientific rigor ensures that the PK assays are suitable for their intended use in supporting biosimilar drug development and regulatory submissions.

This standardized approach to using research-grade emapalumab biosimilars as calibration standards provides the foundation for reliable pharmacokinetic bioequivalence assessments and supports the regulatory pathway for biosimilar approval.

The primary animal models for in vivo administration of research-grade anti-IFNγ antibodies to study tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characteristics are syngeneic mouse models. Use of such antibodies in humanized models is less commonly reported for these specific mechanistic studies.

Key points and context:

  • Syngeneic Models: These are the standard for directly testing murine anti-IFNγ antibodies, since both the tumor and host immune system are murine, allowing full immune interactions. In these models, anti-IFNγ antibodies are administered to block murine IFNγ signaling, providing insights into how IFNγ shapes tumor growth and the immune cell composition within tumors. Such studies often analyze TIL phenotype and functionality after antibody treatment.

    • Example: In a syngeneic mouse model of melanoma, studies have shown that ablation or blockade of IFNγ signaling in T cells (via genetic deletion or pharmacologic blocking antibodies) alters TIL composition, particularly restricting expansion of stem-like CD8+ T cells, thereby impacting anti-tumor immunity and tumor growth control.
    • Tumor models used include MCA-induced sarcomas, RM-1 prostate carcinoma, DA3 mammary carcinoma, and B16 melanoma, most commonly in C57BL/6 or BALB/c backgrounds.
  • Why Syngeneic Models: They allow measurement of both tumor growth inhibition and detailed immune infiltration patterns, including modulation of TIL subsets by anti-IFNγ intervention. Antibodies employed in these studies are specific for murine IFNγ and are research-grade reagents.

  • Humanized Models: Humanized mouse models, where human immune cells are engrafted into immunodeficient mice, are invaluable for certain translational immunotherapy studies. However, use of anti-IFNγ antibodies in these settings is rare for classic TIL phenotyping and tumor growth inhibition because such experiments would require a human-reactive anti-IFNγ antibody, and the immune system and tumor-immune interactions are more complex and less standardized for this specific mechanistic analysis.

  • Outcome Measures: After anti-IFNγ antibody administration, both tumor growth and TIL subsets (e.g., stem-like CD8+ T cells, effector T cells, diversity, and spatial distribution within tumor) are typically assessed by flow cytometry, immunohistochemistry, or single-cell RNA-seq.

  • Limitations of Other Models: While xenograft and patient-derived xenograft (PDX) models are standard for human tumor biology, they lack a functional mouse immune system and thus are not commonly used to study in vivo cytokine blockade affecting TILs with research-grade antibodies.

Summary Table:
| Model Type | Anti-IFNγ Study Use | Immune/TIL Profiling | Relevance ||-------------------|---------------------|----------------------|--------------------------------------|| Syngeneic Mouse | Common | Robust | Standard for IFNγ mechanism studies || Humanized Mouse | Rare | Possible but complex | Less used for this specific purpose || Xenograft/PDX | Not compatible | Not relevant | Immune-deficient, not suited |

Conclusion:
Syngeneic mouse models (e.g., B16 in C57BL/6) are the primary and best-characterized systems in which anti-IFNγ antibodies are administered to assess effects on tumor growth and the tumor-infiltrating lymphocyte compartment in preclinical research. Humanized models are valuable for translational work but are infrequently used for mechanistic anti-IFNγ blockade and detailed TIL analysis.

Researchers are currently using Emapalumab—a fully human anti-IFN-γ monoclonal antibody—primarily to modulate cytokine release syndrome (CRS) and inflammatory toxicity, especially in settings such as CAR-T cell therapy, but direct evidence for its use in combination with classical checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3 biosimilars) for synergistic studies in immune-oncology models is limited in the available literature.

Key points from current research and rationale for combination strategies:

  • Emapalumab blocks IFN-γ, a cytokine implicated in severe immune-related toxicities such as CRS and hemophagocytic lymphohistiocytosis (HLH). Its use can mitigate these complications while preserving the efficacy and persistence of immune cell therapies like CAR-T in some contexts.

  • Checkpoint inhibitors such as anti-CTLA-4 (e.g., ipilimumab) and anti-LAG-3 are typically used to enhance anti-tumor immune responses by reversing T cell exhaustion or inhibition in the tumor microenvironment. Combination therapies involving multiple checkpoint inhibitors (e.g., CTLA-4 plus PD-1, or LAG-3 plus PD-1/PD-L1) have shown synergistic anti-tumor activity and improved clinical benefits in certain cancers.

  • The logical basis for combining agents: Combining checkpoint inhibitors targets multiple nonredundant immunosuppressive pathways, and may boost anti-tumor immunity more than monotherapy. In preclinical models, LAG-3/CTLA-4/PD-1 axis blockade has shown enhanced tumor rejection.

  • Hypothetical role of Emapalumab in combination models:

    • In preclinical or translational models, introducing Emapalumab alongside checkpoint inhibitors could potentially dissect the role of IFN-γ signaling in both efficacy and toxicity of combination immunotherapies.
    • For example, IFN-γ blockade with Emapalumab might reduce severe inflammatory side effects without fully suppressing the anti-tumor benefits of checkpoint-based immunotherapy, although there is concern about compromising some antitumor effects that rely on IFN-γ signaling for cytotoxicity, especially in solid tumors.
    • Synergistic or antagonistic effects would likely be model- and cancer-type specific, and such combinations would require detailed immune profiling (e.g., cytokine analysis, immune cell phenotyping, tumor growth assays) to understand mechanistic interactions.
  • Current limitations:

    • The literature contains robust data on Emapalumab for CRS management and on various combination checkpoint inhibitor strategies (e.g., anti-CTLA-4 plus anti-PD-1; anti-LAG-3 plus anti-PD-1).
    • No direct clinical or preclinical studies were found testing Emapalumab biosimilars in conjunction with anti-CTLA-4 or anti-LAG-3 biosimilars in complex immuno-oncology (IO) models, although such studies may be ongoing or in development.

In summary:
Researchers are using Emapalumab to manage immune-related toxicities in CAR-T and IO settings, and multiple checkpoint inhibitor combinations are under active investigation for synergistic effects. However, studies explicitly combining Emapalumab biosimilars with checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 for synergy in advanced IO models have not yet been published in the current scientific literature. The mechanistic rationale and emerging immune-profiling technologies suggest this is a promising future direction, but data are presently lacking.

Role of Emapalumab Biosimilar in Bridging ADA ELISA for Immunogenicity Testing

Bridging ADA (Anti-Drug Antibody) ELISAs are standard assays for detecting patient immune responses to therapeutic monoclonal antibodies (mAbs), including biosimilars like Emapalumab. These assays are designed to capture and detect ADAs, and sometimes the choice of reagent—whether the originator or biosimilar is used as the capture/detection agent—is critical for ensuring assay sensitivity and relevance.

How the Biosimilar Is Used in the Assay

  • Capture Reagent: In a typical bridging ELISA, the drug itself (e.g., a biosimilar Emapalumab) is immobilized on the plate to specifically capture any patient ADAs present in the sample that recognize that drug. The assay is highly dependent on the structural integrity and epitope presentation of the immobilized drug, so a biosimilar used here must be highly similar to the originator to ensure comparable ADA detection.
  • Detection Reagent: A labeled version of the drug (again, the biosimilar Emapalumab) is added to form a bridge between the captured ADA and the detection system. This labeled biosimilar binds to the patient’s ADA, completing the “bridge.” The label (e.g., biotin, HRP) allows for signal detection, which is proportional to the amount of ADA present.
  • Assay Validation: For regulatory comparability, the biosimilar must be shown to detect ADAs with equivalent sensitivity and specificity as the originator product. This is typically confirmed via head-to-head comparative immunogenicity studies during clinical development.

Practical Considerations

  • Drug Interference: High drug concentrations in patient samples can interfere with ADA detection. Some advanced bridging ELISA formats incorporate steps like acid dissociation or solid-phase extraction to mitigate this and improve drug tolerance.
  • Epitope Presentation: Since biosimilars and originators may have minor differences in post-translational modifications (e.g., glycosylation), it’s critical that the biosimilar’s use as a reagent does not lead to under- or over-detection of clinically relevant ADAs.
  • Regulatory Context: Regulatory guidelines (e.g., EMA, FDA) expect that biosimilars demonstrate comparable immunogenicity to the originator, both in terms of the product’s propensity to induce ADAs and in the ability to detect them. Discrepancies may warrant further investigation to ensure clinical relevance.

Example Workflow

A simplified workflow using a biosimilar Emapalumab in a bridging ADA ELISA might look like this:

  1. Plate Coating: Biosimilar Emapalumab is coated onto an ELISA plate.
  2. Sample Incubation: Patient serum is added; any ADAs specific to Emapalumab bind to the immobilized drug.
  3. Detection Step: Labeled biosimilar Emapalumab is added and binds to the captured ADAs, forming a “bridge.”
  4. Signal Development: A substrate is added to generate a signal proportional to the ADA concentration.

Key Points

  • A biosimilar Emapalumab can be used as both the capture and detection reagent in a bridging ADA ELISA to monitor a patient’s immune response against the therapeutic drug.
  • The biosimilar must be highly similar to the originator to ensure the assay’s clinical relevance and comparability.
  • Assay performance (sensitivity, specificity, drug tolerance) must be validated head-to-head against the originator to meet regulatory expectations for biosimilar immunogenicity assessment.
  • Minor differences in product quality between biosimilar and originator could affect assay performance and must be carefully evaluated during method development and validation.

This approach ensures that the immunogenicity profile of the biosimilar is accurately reflected and that patient monitoring is both sensitive and specific to the therapeutic in use.

References & Citations

1. Mah AY, Cooper MA. Crit Rev Immunol. 36(2):131-147. 2016.
2. Al-Salama ZT. Drugs. 79(1):99-103. 2019.
3. Locatelli F, Jordan MB, Allen C, et al. N Engl J Med. 382(19):1811-1822. 2020.
4. Hatterer E, Richard F, Malinge P, et al. Cytokine. 59(3):570. 2012.
5. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/761107lbl.pdf
Indirect Elisa Protocol
FA
Flow Cytometry
Immunoprecipitation Protocol
General Western Blot Protocol

Certificate of Analysis

- -
- -

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.