Anti-Human IFNy (Emapalumab)

Anti-Human IFNy (Emapalumab)

Product No.: LT2900

- -
- -
Product No.LT2900
Clone
NI-0501
Target
IFNγ
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Emapalumab,NI-0501,emapalumab-lzsg,IFNG ,1709815-23-5
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
Active
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 serve as critical analytical tools in pharmacokinetic bridging ELISA assays, functioning primarily as calibration standards and reference controls to enable accurate quantitative measurement of drug concentrations in serum samples.

Role as Calibration Standards

In PK bridging ELISA assays, research-grade Emapalumab biosimilars are used to construct calibration curves that establish the relationship between known drug concentrations and measured assay signals. The most optimal approach involves developing a single PK assay using a single analytical standard for quantitative measurement of both the biosimilar and reference products. This methodology creates a series of calibration standards at defined concentrations, typically ranging across the expected therapeutic concentration range.

The calibration standards are prepared by serial dilution of the research-grade biosimilar in human serum matrix, creating a concentration gradient that spans the anticipated pharmacokinetic profile. For example, standard concentrations might range from 50 ng/mL to 12,800 ng/mL, establishing nine independent calibration points. These standards enable the construction of a dose-response curve that allows for back-calculation of unknown concentrations in clinical samples.

Function as Reference Controls

Research-grade Emapalumab biosimilars also serve as quality control (QC) samples at multiple concentration levels to monitor assay performance throughout the analytical run. These QC samples are prepared independently from the calibration standards and are analyzed alongside study samples to ensure the assay maintains acceptable precision and accuracy.

The QC samples typically include low, medium, and high concentration controls that bracket the expected sample concentrations. For instance, validation samples might be prepared at concentrations of 50, 150, 1,250, 9,600, and 12,800 ng/mL and quantified against the biosimilar standard curve.

Bioanalytical Comparability Assessment

A critical aspect of using research-grade biosimilars involves establishing bioanalytical equivalence between the test biosimilar and reference products within the assay system. This requires implementing a comprehensive testing strategy that evaluates whether both products can be accurately measured using the same analytical method.

The comparability assessment involves statistical analysis comparing the precision and accuracy data sets of the biosimilar and reference products. The 90% confidence interval approach is commonly applied, comparing results to pre-defined equivalence intervals (typically 0.8 to 1.25) to conclude bioanalytical equivalence. Only after establishing this comparability can the biosimilar be confidently used as the analytical standard for quantifying both test products.

Practical Implementation

The research-grade biosimilar selected as the analytical standard undergoes rigorous method validation consistent with FDA bioanalytical guidance requirements. This includes evaluation of precision, accuracy, selectivity, stability, and robustness across multiple days and analysts. The validation ensures the method can reliably quantify drug concentrations in serum samples throughout the expected analytical range.

The specificity of the ELISA system is particularly important, as it must accurately distinguish the target Emapalumab molecule from other serum components and potential interferents. This specificity validation ensures that the research-grade biosimilar standard provides reliable quantification despite the complex biological matrix.

Through this comprehensive approach, research-grade Emapalumab biosimilars enable accurate, reproducible quantification of drug concentrations in clinical samples, supporting critical pharmacokinetic assessments necessary for biosimilar development and regulatory approval.

The primary models used for in vivo administration of research-grade anti-IFNγ antibodies to study tumor growth inhibition and TIL characterization are syngeneic mouse models, with humanized models used less commonly but increasingly for translational relevance.

Key details:

  • Syngeneic models involve transplanting mouse-derived tumor cells into genetically identical, immunocompetent mice (most often inbred strains like C57BL/6 or BALB/c).

    • These models allow for mechanistic studies of immune modulation, including cytokine blockade (e.g., with anti-IFNγ antibodies), on both tumor growth and composition of TILs.
    • Syngeneic systems are well-established for assessing how interferon signaling influences tumor immunity. For example, studies used genetic knockout or antibody blockade of IFNγ or its receptor to show alterations in CD8+ T cell subsets and expansion, changes in tumor growth rates, and subsequent effects on stem-like TILs.
    • Representative tumors used in these models include B16 melanoma (C57BL/6) and MCA-induced sarcoma (BALB/c).
  • Humanized mouse models—where human immune cells are engrafted into immunodeficient mice—are applied to bridge experimental findings to human biology, particularly for understanding human TIL responses.

    • These models are less commonly referenced for specific anti-IFNγ antibody studies, but are important in validating discoveries made in murine systems before clinical translation.

Common experimental design:

  • Mouse (mAb) monoclonal antibodies against IFNγ are administered to mice bearing syngeneic tumor grafts.
  • Researchers then analyze the impact on tumor growth kinetics and profile TIL populations (e.g., expansion of stem-like CD8+ T cells, clonal diversity, effector phenotype).
  • Models such as melanoma (B16), sarcoma (MCA-induced tumors), and metastasis models (RM-1 prostate, DA3 mammary carcinoma) are specifically described as platforms for dissecting IFNγ’s role in vivo.

Summary Table of Model Types:

Model TypeImmune SystemTumor OriginUse in Anti-IFNγ Studies
Syngeneic MouseMurineMurineStandard; enables detailed study of TILs, IFNγ blockade
Humanized MouseHuman (engraft)Murine/HumanTranslational extension; less common for anti-IFNγ

Supporting insights:

  • Syngeneic models provide direct measurement of immune mechanisms—including TIL infiltration, activation, and exhaustion—as a result of anti-IFNγ therapy, and facilitate biomarker discovery (e.g., IFNγR expression patterns correlating with immune response and tumor suppression).
  • Humanized models are ideal for validating whether findings regarding IFNγ and TILs translate to human cell biology but face limitations in technical complexity and cost.

In summary, syngeneic mouse models are the standard in vivo platform for administering anti-IFNγ antibodies and studying the resulting effects on tumor growth and TIL phenotypes; humanized mouse models provide supporting translational data but are less commonly used for mechanistic anti-IFNγ studies.

Researchers are actively exploring combinations of Emapalumab—a monoclonal antibody targeting interferon-gamma (IFN-γ)—with other checkpoint inhibitors such as anti-CTLA-4 and anti-LAG-3 agents to investigate synergistic effects in complex immune-oncology models. The rationale is that targeting multiple, distinct immunoregulatory pathways may improve anti-tumor responses compared to single-agent therapies.

Emapalumab has primarily been studied for its ability to suppress immune hyperactivation by neutralizing IFN-γ, particularly in conditions like hemophagocytic lymphohistiocytosis (HLH) and in mitigating cytokine release syndrome (CRS) associated with therapies such as CAR T-cell treatment. While published clinical or preclinical studies on direct combinations of emapalumab biosimilars with classical checkpoint inhibitors (CTLA-4 or LAG-3 blockade) are limited or emerging, the foundational principles are supported by broader combination immunotherapy research:

  • Distinct Mechanisms: Anti-CTLA-4 agents act primarily in lymph nodes to facilitate the activation and proliferation of T cells, whereas agents like anti-PD-1 act at tumor sites to prevent suppression of cytotoxic T cells. Targeting IFN-γ with emapalumab could further modulate the tumor microenvironment, especially in settings where excessive immune activation or CRS threatens patient safety or reduces efficacy.
  • Preclinical and Clinical Models: Studies often use in vivo mouse models engrafted with human tumors, or ex vivo humanized immune system models, to test the effects of checkpoint combinations. By introducing an emapalumab biosimilar alongside anti-CTLA-4 or anti-LAG-3 biosimilars, researchers assess:
    • Immune cell infiltration and activation in tumors
    • Cytokine and chemokine profiles (reducing CRS while preserving antitumor immunity)
    • Overall tumor regression, survival, and adverse events
  • Immunotoxicity and Efficacy: The combination approach is also designed to address toxicity: emapalumab may be used to limit IFN-γ-mediated adverse effects (such as CRS or HLH), potentially allowing higher dosing or prolonged use of other checkpoint inhibitors without unacceptable side effects.

Synergistic Effects Evaluation:

  • Multiparametric flow cytometry and multiplex cytokine assays are used to monitor changes in immune cell subsets and functional markers.
  • Tumor growth rates, response rates, and biomarker modulation (e.g., IFN-γ levels, T-cell exhaustion markers) are primary endpoints in both preclinical and early-phase clinical studies.

While robust head-to-head synergy data for emapalumab biosimilars specifically combined with CTLA-4 or LAG-3 biosimilars are not yet extensively published, current research frameworks adapted from multi-checkpoint inhibitor strategies set the foundation for ongoing studies. As the understanding of immune-related adverse events deepens, these combinatorial approaches are expected to play an increasingly significant role in optimizing immune-oncology therapies.

In the context of immunogenicity testing, a biosimilar monoclonal antibody such as Emapalumab can be used in a bridging ELISA to monitor a patient's immune response against the therapeutic drug. While specific details about Emapalumab's use in this context are not available, the general approach can be inferred from how other monoclonal antibodies are used in bridging ELISAs.

Basic Principle of Bridging ELISA:

  1. Capture Reagent: The biosimilar monoclonal antibody (e.g., Emapalumab biosimilar) is immobilized on a microtiter plate to serve as the capture reagent. This step captures anti-drug antibodies (ADAs) present in patient serum samples.

  2. Detection Reagent: Another form of the biosimilar or the same biosimilar is labeled (e.g., with HRP) and used as the detection reagent. This labeled reagent binds to the captured ADAs, forming a "bridge" between the capture and detection reagents.

  3. Detection: The bound labeled reagent is then detected using a chromogenic substrate (e.g., TMB), which changes color in the presence of the enzyme, indicating the presence of ADAs.

Steps in a Bridging ELISA Assay:

  1. Preparation: Coating the plates with the capture reagent (biosimilar monoclonal antibody).

  2. Sample Addition: Adding patient serum samples containing potential ADAs.

  3. Detection Reagent Addition: Adding the labeled biosimilar monoclonal antibody as the detection reagent.

  4. Substrate Addition and Detection: Adding a chromogenic substrate to visualize the bound ADAs.

  5. Data Analysis: Quantifying the immune response based on the optical density (OD) of the substrate reaction.

Considerations for Emapalumab Biosimilar in Bridging ELISA:

  • Interference Minimization: Methods like acid dissociation can be used to minimize interference from high drug concentrations, ensuring accurate ADA detection.
  • Sensitivity and Specificity: The assay conditions must be optimized to ensure high sensitivity and specificity for detecting ADAs, which is crucial for monitoring immunogenicity.
  • Validation: The assay should be thoroughly validated to ensure that it accurately measures the immune response against the therapeutic drug.

While there isn't specific information on Emapalumab's use in bridging ELISAs, the approach described above outlines how biosimilars can generally be used in this context. Emapalumab, like other monoclonal antibodies, could be adapted to this method if it has sufficient binding properties and is amenable to the bridging ELISA format.

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.