IL 2r Antibody

Interleukin-2 Receptor (CD25), Mouse Anti-Human
Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

IL-2R antibodies are monoclonal or engineered antibodies that bind to subunits of the IL-2 receptor (IL-2R), a heterotrimeric complex composed of:

  • IL-2Rα (CD25): Low-affinity subunit, primarily expressed on activated T cells and regulatory T cells (Tregs).

  • IL-2Rβ (CD122): Intermediate-affinity subunit, expressed on memory T cells and natural killer (NK) cells.

  • IL-2Rγ (CD132): Common gamma chain shared with other cytokine receptors.

These antibodies are categorized based on their targets:

Antibody TypeTarget SubunitPrimary Application
Anti-CD25 (e.g., basiliximab)IL-2RαPrevention of organ transplant rejection
Anti-CD122IL-2RβCancer immunotherapy
Bispecific antibodiesIL-2Rβ + IL-2RγSelective Treg or effector T cell expansion

Mechanism of Action

IL-2R antibodies function through distinct mechanisms:

  • Blockade of IL-2 binding: Anti-CD25 antibodies (e.g., daclizumab, basiliximab) competitively inhibit IL-2 from binding to the high-affinity receptor, suppressing T cell proliferation .

  • Receptor agonism: Bispecific antibodies (e.g., IL-2Rβ/γ agonists) mimic IL-2 signaling to activate effector T cells or NK cells while avoiding Treg stimulation .

  • Covalent fusion strategies: Single-agent IL-2/antibody complexes (e.g., TCB2, UFKA-20) bias signaling toward Tregs or effector cells by altering receptor-binding dynamics .

Transplant Rejection Prevention

  • Basiliximab and daclizumab: Reduce acute rejection rates in renal transplants by 30–40% by blocking IL-2Rα on activated T cells .

  • Dosing: Pediatric regimens include 10–20 mg basiliximab (days 0 and 4 post-transplant) or 1 mg/kg daclizumab every 14 days .

Autoimmune Disease Management

  • Low-dose IL-2/antibody complexes: Expand Tregs to suppress autoimmunity in systemic lupus erythematosus (SLE) and ulcerative colitis .

  • TCB2-IL-2 fusion: Demonstrated superior efficacy in SLE mouse models by selectively activating Tregs .

Cancer Immunotherapy

  • Bispecific IL-2Rβ/γ agonists: Promote CD8+ T cell and NK cell proliferation without stimulating Tregs, enhancing antitumor responses .

  • IL-2 variants: Engineered to reduce vascular leak syndrome and improve half-life (e.g., PEGylated IL-2) .

Key Research Findings

StudyAntibody/ComplexOutcomeReference
Crystal structure of IL-2/TCB2 complexTCB2Allosteric enhancement of IL-2Rβ/γ binding
Bispecific IL-2Rβ/γ agonist (NGS-derived)UniAbsActivated cynomolgus CD8+ T cells
CD25-biased IL-2/UFKA-20 complexUFKA-20Selective Treg expansion in primates
IL-2/anti-IL-2 fusion for SLECovalent IL-2/antibodyDisease control in murine models

Challenges and Limitations

  • Short half-life: Native IL-2 and antibodies require frequent dosing .

  • Off-target effects: Activation of effector T cells can exacerbate autoimmunity or cause cytokine storms .

  • Treg selectivity: Achieving Treg-specific signaling without affecting effector cells remains difficult .

Future Directions

  • Engineered IL-2/antibody fusions: Improve pharmacokinetics and specificity (e.g., half-life extension via Fc fusion) .

  • Combination therapies: Pairing IL-2R antibodies with checkpoint inhibitors (e.g., anti-PD-1) to enhance antitumor immunity .

  • Precision targeting: Structural insights from crystallography (e.g., IL-2/TCB2 binding angles) guide rational drug design .

Product Specs

Introduction
IL2-Ra, a subunit of the IL2 receptor, is released into the serum upon elevated cellular expression, often observed in activated B and T cells. High IL2-Ra levels are associated with autoimmune disorders, leukemias, and lymphomas.
Formulation
The antibody is supplied at a concentration of 1 mg/ml in PBS following reconstitution.
Shipping Conditions
The antibody is shipped in lyophilized form at ambient temperature.
Storage Procedures
Store the lyophilized antibody at 4°C in a dry environment for long-term storage. After reconstitution, aliquot and store at -20°C if not intended for use within one month.
Solubility
Reconstitute the antibody with sterile water. Gently mix the solution, ensuring to wash the sides of the vial. Allow the solution to stand for 30-60 seconds before use.
Titer
A 1:50 dilution of the antibody effectively blocks 70-80% of the proliferative response of CTLL cells to 50 units of recombinant human IL-2. This dilution also stains 70% of PHA-activated human T cells as determined by flow cytometry. For FACS analysis or sorting, 10 µl of the antibody is sufficient to stain 10^6 IL-2R positive cells.
Synonyms
CD25, IL2R, TCGFR, IL-2RA, sIL-2RA, TAC antigen, sIL-2R, IDDM10, p55 TypeMouse Anti Human Monoclonal.
Purification Method
Ion Exchange.
Clone
YNRhIL2R.
Immunogen
Con A-activated human T cells.
Ig Subclass
Mouse IgG2a.

Q&A

What are the different IL-2 receptor subunits and how do they affect antibody selection?

The IL-2 receptor exists in three forms with varying affinities for IL-2:

  • High-affinity receptor: Trimeric complex (IL-2Rα/CD25, IL-2Rβ/CD122, IL-2Rγ/CD132)

  • Intermediate-affinity receptor: Dimeric complex (IL-2Rβ and IL-2Rγ)

  • Low-affinity receptor: Monomeric IL-2Rα

When selecting antibodies, consider which receptor subunit you need to target based on your research question. For detecting Treg cells, anti-IL-2Rα antibodies are often preferred as these cells constitutively express high levels of this subunit. For broader detection of IL-2 responsive cells, anti-IL-2Rβ antibodies may be more appropriate since effector T cells and NK cells express the intermediate-affinity receptor .

How can I reliably detect IL-2R expression on different immune cell populations?

For detecting IL-2R expression on immune cells:

  • Use flow cytometry with fluorochrome-conjugated antibodies specific for individual IL-2R subunits

  • Include proper isotype controls and gating strategy

  • Consider multiparameter analysis that includes lineage markers

For example, to identify IL-2Rβ expression on lymphocytes, you can stain peripheral blood lymphocytes with anti-IL-2Rβ antibodies alongside lineage markers like CD56 for NK cells . This approach allows for identification of receptor expression on specific cell subsets and provides more informative data than bulk analyses.

What are the key considerations when measuring soluble IL-2R (sIL-2R) in biological samples?

When measuring sIL-2R in serum or plasma:

  • Sample handling: Minimize freeze-thaw cycles and standardize collection procedures

  • Assay selection: ELISA-based methods are commonly used, but multiplex bead arrays may offer advantages for simultaneous cytokine profiling

  • Reference ranges: Establish appropriate healthy control ranges for your specific assay and population

  • Clinical context: Interpret results in the context of disease activity, as sIL-2R levels correlate with immune activation in autoimmune diseases and certain cancers

sIL-2R measurements can be particularly valuable in monitoring disease progression and therapy response in rheumatoid arthritis and various cancers, serving as a biomarker of immune activation .

How do different anti-IL-2R antibody clones vary in their epitope specificity and functional effects?

Anti-IL-2R antibodies can target different epitopes on the receptor subunits, leading to distinct functional outcomes:

Antibody TypeTarget EpitopeFunctional EffectResearch Application
Immunoenhancing (e.g., S4B6, JES6-5H4)Interface with IL-2RαPreferential stimulation of cells with IL-2Rβ/γEnhancing effector T cell and NK cell responses
Immunoregulatory (e.g., JES6-1A12)Region between IL-2Rα and IL-2Rβ/γ interfacesDual inhibition of both IL-2Rα and IL-2Rβ/γ interactionsModulating IL-2 signaling

Understanding the epitope specificity is crucial as it determines whether an antibody will block, enhance, or have no effect on IL-2 binding and signaling . When designing experiments, select antibodies based on their documented functional effects rather than just their binding specificity.

What controls should be included when using anti-IL-2R antibodies in functional assays?

For robust experimental design with anti-IL-2R antibodies:

  • Isotype controls: Include matched isotype control antibodies to account for non-specific effects

  • Blocking controls: Use known blocking antibodies against IL-2 or IL-2R to confirm specificity

  • Stimulation controls: Include positive controls like recombinant IL-2 to validate cell responsiveness

  • Cell type controls: Test antibodies on cells known to express or lack specific IL-2R subunits

  • Dose-response assessments: Titrate antibody concentrations to identify optimal working concentrations

Competition assays can be particularly informative, as they reveal whether different antibodies recognize overlapping epitopes. For instance, research shows that JES6-1A12 does not compete with S4B6 for binding to mouse IL-2, indicating they recognize different antigenic regions .

How can epitope mapping techniques be used to characterize novel anti-IL-2R antibodies?

For comprehensive epitope mapping of anti-IL-2R antibodies:

  • Phage display mutagenesis: Generate libraries of antigen variants with single or multiple mutations, then screen for interactions with the antibody

  • Competitive binding assays: Determine if the antibody competes with known antibodies or natural ligands

  • Cross-species reactivity analysis: Test binding to homologous proteins from different species to identify conserved epitope regions

  • Structural analysis: Use X-ray crystallography or cryo-EM to determine the atomic structure of antibody-antigen complexes

These approaches can reveal critical information about antibody function. For example, studies using phage display with humanized segments of mouse IL-2 helped delineate the JES6-1A12 epitope, explaining its immunoregulatory properties .

How can anti-IL-2R antibodies be engineered to selectively modulate different immune cell populations?

Engineering selective IL-2R-targeting antibodies involves several approaches:

  • Bispecific antibody design: Create antibodies that simultaneously bind IL-2Rβ and IL-2Rγ to mimic IL-2 signaling while avoiding IL-2Rα binding, thus preferentially activating effector cells over Tregs

  • Fusion protein development: Engineer covalently-linked fusions of IL-2 with anti-IL-2 antibodies that selectively direct activity toward specific receptor combinations

  • Affinity modulation: Modify antibody binding domains to adjust affinity for different receptor subunits

For example, researchers have developed bispecific heavy-chain only antibodies that bind to and activate signaling through the heterodimeric IL-2Rβγ receptor complex, circumventing the preferential Treg activation seen with native IL-2 while maintaining robust stimulatory effects on effector T cells and NK cells .

What strategies can resolve contradictory data when studying IL-2R antibody effects across different experimental systems?

When facing contradictory data with IL-2R antibodies:

  • Cell context analysis: Determine receptor expression levels across your experimental systems, as effects may differ based on the relative expression of IL-2Rα vs. IL-2Rβ/γ

  • Species-specific differences: Consider that human and mouse IL-2 systems have important differences; for example, some antibodies show species-specific effects due to sequence variations in the receptors

  • Antibody concentration effects: Test wide concentration ranges, as some antibodies exhibit bell-shaped dose-response curves or different effects at varying concentrations

  • Multiparameter readouts: Assess multiple downstream signaling pathways (STAT5 phosphorylation, cell proliferation, cytokine production) as antibodies may differentially affect these pathways

  • In vivo vs. in vitro discrepancies: Validate findings across both systems when possible, as the complex in vivo environment may alter antibody activity

How can IL-2R antibodies be applied in therapeutic development for autoimmune diseases?

IL-2R antibodies offer promising therapeutic approaches for autoimmune diseases:

  • Low-dose IL-2 therapy enhancement: IL-2/anti-IL-2 antibody complexes can selectively expand Treg cells at lower IL-2 doses than would otherwise be required

  • Selective receptor targeting: Bispecific antibodies or engineered fusions can direct IL-2 activity preferentially toward Tregs by exploiting their heightened IL-2 sensitivity

  • Combinatorial approaches: Pairing IL-2R modulation with antigen-specific therapies may enhance regulatory responses to self-antigens

For example, a single-agent fusion of human IL-2 and anti-IL-2 antibody demonstrated superior disease control in animal models of ulcerative colitis and systemic lupus erythematosus by selectively expanding Treg cells . This approach leverages the observation that Treg cells have heightened sensitivity to IL-2, potentially creating a therapeutic window to promote immune regulation .

What are common pitfalls in flow cytometric analysis of IL-2R expression and how can they be avoided?

Common pitfalls and solutions for IL-2R flow cytometry:

  • False negative results:

    • Ensure antibody clones can recognize both resting and activated forms of the receptor

    • Use freshly isolated cells when possible, as receptor expression may change during extended culture

    • Optimize staining buffers to prevent receptor internalization

  • Nonspecific binding:

    • Include proper blocking steps (Fc block) to prevent Fc receptor binding

    • Use carefully titrated antibody concentrations to minimize background

    • Include fluorescence-minus-one (FMO) controls for accurate gating

  • Epitope masking:

    • Be aware that ligand binding may block antibody access to certain epitopes

    • Consider using multiple antibody clones targeting different epitopes for validation

  • Receptor modulation:

    • Account for receptor downregulation following activation

    • Standardize the timing of analysis following stimulation

How can researchers distinguish between IL-2R blocking effects and agonistic activities of anti-IL-2R antibodies?

To distinguish blocking from agonistic antibody effects:

  • Comparative signaling analysis: Measure STAT5 phosphorylation, which is a direct downstream effect of IL-2R engagement, in the presence of:

    • Antibody alone

    • IL-2 alone

    • IL-2 plus antibody

  • Functional readouts: Assess cell proliferation, survival, and cytokine production to determine if the antibody:

    • Enhances IL-2 activity (agonistic)

    • Inhibits IL-2 activity (blocking)

    • Redirects IL-2 activity toward specific cell populations

  • Receptor occupancy studies: Use competitive binding assays to determine if the antibody prevents IL-2 binding or alters binding kinetics

  • Dose-response relationships: Analyze effects across a wide concentration range, as some antibodies may demonstrate biphasic effects depending on concentration

What approaches can improve reproducibility when measuring soluble IL-2R (sIL-2R) in clinical samples?

To enhance reproducibility of sIL-2R measurements:

  • Standardized sample processing:

    • Establish consistent blood collection protocols

    • Process samples within a defined timeframe

    • Use standardized centrifugation and storage conditions

  • Assay optimization:

    • Validate commercial kits for your specific sample types

    • Establish internal quality controls and standard curves

    • Perform dilution linearity testing to ensure accurate quantification

  • Analytical considerations:

    • Account for potential interfering factors (medications, inflammatory markers)

    • Consider circadian variations in sIL-2R levels

    • Establish appropriate reference intervals for your population

  • Data interpretation:

    • Correlate sIL-2R levels with clinical parameters and other biomarkers

    • Consider longitudinal monitoring rather than single time points

    • Establish clinically relevant cutoff values for specific diseases

How can next-generation sequencing (NGS) techniques enhance the discovery of novel anti-IL-2R antibodies?

NGS-based approaches offer powerful tools for anti-IL-2R antibody discovery:

  • Repertoire sequencing: Analyze antibody repertoires from immunized animals or human samples to identify candidates with desired binding properties

  • Rational library design: Create and screen focused antibody libraries based on structural knowledge of IL-2R epitopes

  • Humanized animal platforms: Generate fully human antibodies using humanized rat systems (UniRats) to identify a diverse collection of binding domains with varying agonist activities

  • Computational prediction: Apply machine learning algorithms to predict antibody-antigen interactions and design improved binding domains

These approaches have successfully yielded novel bispecific antibodies targeting both IL-2Rβ and IL-2Rγ subunits, enabling agonistic activity without preferential Treg activation .

What is the potential of combining IL-2R-targeted approaches with other immunomodulatory strategies?

Combining IL-2R targeting with other immunomodulatory approaches offers several advantages:

  • Synergistic immune modulation: Pairing IL-2R-targeted therapies with checkpoint inhibitors may enhance anti-tumor responses while controlling adverse events

  • Cell type-specific targeting: Combining IL-2R modulation with antigen-specific therapies could enhance regulatory responses to self-antigens in autoimmune conditions

  • Improved therapeutic window: Using IL-2R antibodies to direct IL-2 activity toward specific cell populations may reduce toxicities associated with high-dose IL-2 therapy

  • Sequential treatment strategies: Priming the immune system with one approach before applying IL-2R-targeted therapy may optimize therapeutic outcomes

This combinatorial approach represents a promising frontier in immunotherapy research, potentially addressing the limitations of current single-agent approaches.

Product Science Overview

Introduction

The Interleukin-2 receptor (IL-2R) is a critical component of the immune system, playing a pivotal role in the regulation of immune responses. The receptor is composed of three subunits: alpha (IL-2Rα or CD25), beta (IL-2Rβ), and gamma (IL-2Rγ). These subunits can combine in various ways to form receptors with different affinities for interleukin-2 (IL-2), a cytokine that is essential for the growth, proliferation, and differentiation of T cells .

Structure and Function

The alpha subunit, also known as CD25, is a 55 kDa protein that is primarily expressed on the surface of activated T cells, B cells, and monocytes. CD25 alone forms the low-affinity IL-2 receptor, while the combination of CD25 with IL-2Rβ and IL-2Rγ forms the high-affinity receptor. The intermediate-affinity receptor is formed by the combination of IL-2Rβ and IL-2Rγ .

The high-affinity IL-2 receptor plays a crucial role in intracellular signal transduction, leading to the activation and proliferation of T cells. This process is essential for the immune system’s ability to respond to infections and other immune challenges .

Expression and Regulation

CD25 is strongly expressed on the surface of T cells that have been activated by antigens, mitogens, or various pathogens such as viruses and bacteria. It is also expressed on B cells stimulated with anti-IgM antibodies and on monocytes/macrophages stimulated with lipopolysaccharides .

In normal peripheral blood, the expression of CD25 varies among lymphoid cells, with approximately 3-25% of these cells showing staining for CD25 .

Monoclonal Mouse Anti-Human CD25

Monoclonal antibodies against human CD25, such as those produced in mice, are valuable tools in both research and clinical diagnostics. These antibodies are typically produced from purified monoclonal mouse antibodies and are available in various conjugated forms, such as FITC (Fluorescein Isothiocyanate) and RPE (R-Phycoerythrin) .

These conjugates are used in flow cytometry to analyze the expression of CD25 on the surface of lymphocytes and other immune cells. The specificity of these antibodies is confirmed through immunoprecipitation and immunocytochemical labeling .

Applications

Monoclonal mouse anti-human CD25 antibodies are used in various applications, including:

  • Flow Cytometry: To analyze the expression of CD25 on immune cells.
  • Immunohistochemistry: To study the distribution and localization of CD25 in tissue samples.
  • Clinical Diagnostics: To assess the activation status of T cells in various diseases, including autoimmune disorders and cancers .

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.