IL2RA Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target

IL2RA monoclonal antibodies are immunotherapies that inhibit or deplete cells expressing the high-affinity IL-2 receptor alpha chain (CD25). This receptor is upregulated on activated T cells and Tregs, making it a strategic target for immune modulation .

Table 1: Primary Applications

ApplicationMechanism/OutcomeSources
Cancer TherapyDepletes leukemic stem cells in AML; synergizes with chemotherapy .
Transplant RejectionReduces acute rejection in renal transplants by 25–28% vs. placebo .
Autoimmune DiseasesLow-dose IL-2 therapy restores Treg function in rheumatoid arthritis .
Research ToolsValidated for WB, IHC, ICC, IP, and flow cytometry (e.g., Alexa Fluor® conjugates) .

Table 2: Clinical Trial Outcomes

ConditionAntibody UsedKey ResultCitation
Acute Myeloid Leukemia (AML)Anti-IL2RA mAbsInhibited leukemic cell proliferation; enhanced survival in mouse models .
GlioblastomaDaclizumabReduced Tregs by 48% post-lymphodepletion; improved vaccine efficacy .
Kidney TransplantationBasiliximab/DaclizumabLower acute rejection rates (RR 0.72) and CMV risk vs. ATG .

Mechanism of Action

  • Signal Blockade: Competitively inhibits IL-2 binding to CD25, disrupting JAK1/JAK3 signaling and downstream T-cell activation .

  • Treg Depletion: Preferential depletion of CD4+CD25+Foxp3+ Tregs in lymphopenic environments (e.g., post-chemotherapy) .

  • Synergy: Enhances efficacy of antileukemic agents and vaccines by reducing immunosuppressive Treg activity .

Clinical Considerations

  • Safety: Fewer infections and malignancies vs. polyclonal antibodies (e.g., ATG) .

  • Dosing: Renal transplant protocols: Basiliximab (20 mg on days 0/4) or daclizumab (1 mg/kg biweekly) .

  • Limitations: Ineffective against established acute rejection due to transient CD25 expression .

Future Directions

Ongoing trials explore IL2RA antibodies in combination with checkpoint inhibitors for solid tumors and low-dose IL-2 regimens for autoimmune diseases . Challenges include optimizing Treg-specific depletion while sparing effector T cells .

Product Specs

Buffer
PBS, pH 7.4, with 0.02% sodium azide as a preservative and 50% glycerol.
Form
Liquid
Lead Time
Product shipment typically occurs within 1-3 business days of order receipt. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Synonyms
IL2RA; Interleukin-2 receptor subunit alpha; IL-2 receptor subunit alpha; IL-2-RA; IL-2R subunit alpha; IL2-RA; TAC antigen; p55; CD antigen CD25
Target Names
Uniprot No.

Target Background

Function

IL2RA is a receptor for interleukin-2, playing a crucial role in immune tolerance regulation by modulating regulatory T cell (Treg) activity. Tregs suppress the activation and expansion of autoreactive T cells.

Gene References Into Functions
IL2RA Gene and Associated Diseases: Research Highlights

The following studies demonstrate the significant role of IL2RA in various disease processes:

  1. Early-stage ALS is characterized by neuroprotective helper T cell infiltration in the lumbar spinal cord, indicated by increased peripheral CD4+ helper T cells and higher FOXP3 and IL2Ralpha expression. (PMID: 29574662)
  2. An association exists between IL2RA SNPs and alopecia areata susceptibility in an Iranian cohort. (PMID: 29979892)
  3. IL2RA gene polymorphisms (rs2104286 and rs12722489) are linked to multiple sclerosis (MS) susceptibility in Chinese populations. (PMID: 30352019)
  4. While one study found no significant association between IL2RA SNPs and MS, this might be due to limited power and sample size. (PMID: 29141792)
  5. Meta-analysis suggests the rs2104286 A allele increases MS risk in Caucasians and Asians, while the rs12722489 C allele elevates risk in Caucasians. (PMID: 29648897)
  6. Soluble IL-2R (sIL-2R) shows promise as a marker for inflammatory disease activity in complex regional pain syndrome (CRPS). (PMID: 28634419)
  7. Research explores the relationship between sCD25 and gene expression in healthy individuals and MS patients. (PMID: 28511943)
  8. Elevated serum sIL-2R levels correlate with a poorer response to interferon alpha and VEGF-targeting therapy in metastatic clear cell renal cell carcinoma. (PMID: 28545581)
  9. IL2Rα, TNF receptor 1, sSTimulation-2 (IL1RL1), and regenerating islet-derived 3-alpha are significantly associated with non-relapse mortality. (PMID: 28126963)
  10. High IL2RA expression is associated with CRLF2-rearranged acute lymphoblastic leukemia. (PMID: 28866095)
  11. LKB1-AMPK-mTOR pathway regulation inhibits CD25 translation, suppressing T cells. (PMID: 28230853)
  12. Post-treatment sIL-2R levels in patients with a positive response to R-CHOP and low pretreatment sIL-2R may indicate poor prognosis. (PMID: 28413914)
  13. CD25 expression is a prognostic factor in Philadelphia chromosome-positive (Ph+) and Ph- acute lymphoblastic leukemia (ALL), potentially linked to underlying molecular abnormalities. (PMID: 28430957)
  14. Genome-wide association studies (GWAS) reveal a sex-specific association between emotion dysregulation and an IL2RA SNP in men. (PMID: 27643478)
  15. CD25 acts as a negative growth regulator of chronic myeloid leukemia leukemic stem cells. (PMID: 28457753)
  16. Decidual stromal cells influence IL-2 production and IL-2R expression and signaling. (PMID: 27651429)
  17. The single nucleotide variant rs12722489 affects ERα binding and enhancer properties of an IL2RA intronic region. (PMID: 28234966)
  18. CDK6-mediated CD25 suppression is essential for Notch1-activated T-cell acute lymphoblastic leukemia (T-ALL) initiation. (PMID: 26707936)
  19. CD4+ CD25+ GARP+ Treg cells are deficient in dilated cardiomyopathy patients, suggesting GARP as a potential regulatory phenotype marker. (PMID: 28207945)
  20. sIL-2R measurement offers potential diagnostic and therapeutic applications in hemophagocytic syndromes. (PMID: 28497365)
  21. Differential methylation of the IL2RA promoter in T cells may be a key pathogenic mechanism in MS. (PMID: 28077880)
  22. Sustained STAT5 phosphorylation is crucial for long-term CD25 expression in T lymphocytes. (PMID: 27936140)
  23. Gastric adenocarcinoma patients exhibit increased PD-1+ lymphocytes and CD4+CD25+FOXP3+ Tregs in peripheral blood. (PMID: 28031121)
  24. Increased serum sIL-2R levels are observed in Kawasaki disease. (PMID: 28081636)
  25. CD25 is an independent prognostic factor in elderly acute myeloid leukemia (AML) patients, highlighting the need for alternative therapies. (PMID: 28097942)
  26. Elevated CD25 levels are found in patients with active vitiligo. (PMID: 27556155)
  27. CD45RA distinguishes CD4+CD25+CD127-/low Treg subpopulations with varying stability and susceptibility to tacrolimus. (PMID: 28118317)
  28. Combining anti-CD25 with anti-PD-1 antibodies promotes complete tumor rejection, suggesting CD25 as a therapeutic target in immuno-oncology. (PMID: 28410988)
  29. IL-7-mediated homeostatic proliferation compensates for CD24+Foxp3+ Treg cell inhibition after anti-CD25 therapy in islet allotransplantation. (PMID: 27306531)
  30. (99)Tc-methylene diphosphonate may enhance rheumatoid arthritis (RA) activity by upregulating γδ T cells and CD4+CD25+Foxp3+ Tregs, and modifying cytokine levels. (PMID: 24467668)
  31. IL-2R antagonists reduce the risk of new-onset diabetes after liver transplantation (NODAT). (PMID: 26588180)
  32. CD8+CD25+FoxP3bright Tregs correlate with mean peak expiratory flow. (PMID: 25921629)
  33. Certain treatments efficiently downregulate CD4+CD25+Foxp3+ Treg cells. (PMID: 27431260)
  34. CD25 and CD135 values offer potential alternatives to FLT3-ITD mutation testing. (PMID: 27087256)
  35. PTPN22 polymorphisms contribute to type 1 diabetes mellitus predisposition in Egyptian children. (PMID: 27288719)
  36. Analysis of IL2RA gene intron 1 SNPs reveals potential influence on gene regulation in autoimmune disease. (PMID: 27876533)
  37. sIL-2R serves as a reliable prognostic marker in head and neck cancer (HNC), either alone or in a biomarker panel. (PMID: 27466555)
  38. CD25 negatively impacts prognosis in elderly AML patients, even with transplantation, suggesting a need for additional therapy. (PMID: 26422713)
  39. IL2RA and TAGAP are vitamin D target genes, with responses observed in both MS patients and controls, irrespective of MS-associated SNP genotypes. (PMID: 26765264)
  40. The anti-CD25 immunotoxin LMB-2 demonstrated limited phase I activity due to immunogenicity and rapid growth. (PMID: 26350263)
  41. Elevated pretreatment CD25 expression on CD4+ T cells correlates with reduced survival in AML patients. (PMID: 26721345)
  42. Untreated ulcerative colitis patients exhibit decreased CD4+CD25+CD127lowFoxP3+ Tregs with impaired suppression. (PMID: 26333292)
  43. Ultra-high serum sIL-2R levels at diagnosis are a poor prognostic biomarker for angioimmunoblastic T-cell lymphoma (AITL). (PMID: 25563559)
  44. The rs2104286 G allele in IL2RA is more frequent in neuromyelitis optica patients compared to controls in a Southern Han Chinese population. (PMID: 24257225)
  45. The CD34+/CD25+/CD123+/CD99+ leukemia-associated immunophenotype (LAIP) is strongly associated with FLT3-ITD-positive cells. (PMID: 25957287)
  46. IL2RA expression predicts poor prognosis in AML. (PMID: 26375984)
  47. IL2RA SNP rs2104286 and serum sIL2Rα levels are associated with rheumatoid arthritis persistence. (PMID: 26350950)
  48. The CD4+CD45RO+CD25-/lowCD127+ : CD4+CD45RO+CD25hiCD127-/low ratio in peripheral blood identifies heart transplant recipients at risk for cardiac allograft vasculopathy. (PMID: 25539460)
  49. Analysis of sIL-2R levels in sarcoidosis patients with renal insufficiency. (PMID: 25745051)
  50. Five IL2RA gene SNPs are identified as risk factors for type 1 diabetes. (PMID: 26249556)
Database Links

HGNC: 6008

OMIM: 147730

KEGG: hsa:3559

STRING: 9606.ENSP00000369293

UniGene: Hs.231367

Involvement In Disease
Diabetes mellitus, insulin-dependent, 10 (IDDM10); Immunodeficiency 41 with lymphoproliferation and autoimmunity (IMD41)
Subcellular Location
Membrane; Single-pass type I membrane protein.

Q&A

What is IL2RA and what is its functional significance in immunology?

IL2RA (Interleukin-2 receptor subunit alpha), also known as CD25, is a transmembrane glycoprotein with a molecular mass of approximately 55 kDa. In humans, the canonical protein consists of 272 amino acid residues with a mass of 30.8 kDa and is primarily localized in the cell membrane . IL2RA functions as part of the high-affinity IL-2 receptor complex, which also includes IL-2 receptor beta (IL2RB) and the common gamma chain (IL2RG). Homodimeric alpha chains result in low-affinity receptors, while heterotrimeric complexes form high-affinity IL-2 receptors .

The receptor is critically involved in the regulation of immune tolerance by controlling regulatory T cell (Treg) activity. Specifically, IL2RA-expressing Tregs suppress the activation and expansion of autoreactive T cells, thereby maintaining immune homeostasis . IL2RA demonstrates notable expression in lymphoid tissues including tonsil, spleen, lymph node, and cerebellum . Post-translational modifications, particularly glycosylation, significantly influence the receptor's functional properties and cellular interactions .

The IL2RA marker is particularly valuable for identifying specific immune cell populations:

  • Regulatory T cells (Tregs)

  • T follicular regulatory cells

  • Large intestine lamina propria lymphocytes

  • Activated B and T lymphocytes

  • Activated monocytes/macrophages

How are IL2RA monoclonal antibodies generated and validated for research use?

IL2RA monoclonal antibodies are typically generated through immunization with native purified IL2RA from specific sources, such as PHA-activated peripheral blood leukocytes . The generation process follows these methodological steps:

  • Immunization: Animals (typically mice) are immunized with either recombinant IL2RA protein or cell preparations containing IL2RA .

  • Hybridoma production: B cells from immunized animals are fused with myeloma cells to create hybridomas that continuously produce antibodies.

  • Screening: Hybridomas are screened for specific binding to IL2RA through ELISA, flow cytometry, or other binding assays.

  • Clonal selection: Hybridomas producing antibodies with desired specificity are subcloned to ensure monoclonality.

  • Scale-up: Selected clones are expanded for antibody production and purification.

Validation of IL2RA monoclonal antibodies involves multiple methodological approaches:

  • Binding specificity: Confirmation through ELISA binding assays using recombinant IL2RA proteins .

  • Functional testing: Assessment of neutralizing capacity, epitope specificity, and cross-reactivity.

  • Application validation: Testing in relevant applications such as flow cytometry, immunoprecipitation, and functional assays .

Researchers should verify that their selected antibody has been validated for their specific application and species of interest, as epitope recognition can vary significantly between different antibody clones .

What are the critical differences between neutralizing and non-neutralizing IL2RA monoclonal antibodies?

Neutralizing and non-neutralizing IL2RA monoclonal antibodies differ fundamentally in their mechanisms of action and research applications:

Neutralizing IL2RA antibodies:

  • Directly block the interaction between IL-2 and its receptor by binding to functionally critical epitopes

  • Inhibit downstream signaling pathways activated by IL-2 binding

  • Deplete IL2RA-expressing cells in vivo, particularly regulatory T cells (93% reduction observed in some studies)

  • Can augment therapeutic antitumor efficacy (66% reduction in tumor growth reported)

  • Particularly useful for functional studies examining the consequences of IL2RA signaling blockade

  • May have different effects depending on the immunological context (e.g., lymphodepleted versus normal conditions)

Non-neutralizing IL2RA antibodies:

  • Bind to IL2RA without interfering with IL-2 binding or signaling

  • Primarily used for detection and quantification rather than functional modulation

  • Important for phenotypic characterization of cell populations via flow cytometry

  • Valuable for tracking IL2RA-expressing cells without altering their function

  • Often preferred for immunoprecipitation studies to maintain protein-protein interactions

Methodologically, researchers should select between these antibody types based on their experimental goals. For functional studies investigating IL2RA signaling blockade, neutralizing antibodies are appropriate. For phenotypic studies requiring detection without functional interference, non-neutralizing antibodies are preferable .

How should researchers optimize experimental conditions when using IL2RA monoclonal antibodies for flow cytometry?

Optimizing experimental conditions for IL2RA monoclonal antibody use in flow cytometry requires systematic methodological consideration of multiple variables:

Antibody titration:
Begin with a concentration range based on manufacturer recommendations (typically 20 μl per 100 μl whole blood or 10^6 cells in suspension) . Perform a titration series to determine the optimal concentration that provides maximum signal-to-noise ratio. This prevents both insufficient staining and excessive background from non-specific binding.

Sample preparation protocol:

  • Fresh samples yield optimal results for IL2RA detection

  • If fixation is necessary, use paraformaldehyde (0.5-2%) for 10-15 minutes

  • For intracellular staining (when examining Foxp3+ Tregs), use specialized permeabilization buffers

  • Include a viability dye to exclude dead cells, which can bind antibodies non-specifically

Fluorophore selection:
Select fluorophores based on the cytometer configuration and other markers in the panel. For IL2RA, which is typically highly expressed on activated cells, fluorophores with moderate brightness (e.g., PE, APC) are often suitable . For detecting low-level expression, brighter fluorophores (PE, PE-Cy7) are recommended.

Control samples:
The following controls are essential for accurate interpretation:

  • Unstained controls

  • Single-color controls for compensation

  • Fluorescence-minus-one (FMO) controls

  • Isotype controls (using matching IgG1 isotype antibodies)

  • Biological controls (stimulated vs. unstimulated cells)

Gating strategy for IL2RA+ populations:
Start with standard gating to identify lymphocytes based on forward and side scatter, exclude doublets and dead cells, then apply lineage markers before analyzing IL2RA expression. For Treg identification, gate on CD4+CD25+Foxp3+ cells.

What methodological approaches should be used to investigate IL2RA monoclonal antibody-mediated depletion of regulatory T cells?

Investigating IL2RA monoclonal antibody-mediated depletion of regulatory T cells requires systematic methodological approaches that account for multiple biological and experimental variables:

In vivo assessment protocol:

  • Baseline measurement: Obtain blood samples before antibody administration to establish baseline Treg levels.

  • Dose determination: Test multiple doses (typically starting at 10-50 μg/mouse or 1-10 mg/kg in humans).

  • Timing analysis: Sample at multiple timepoints (e.g., 24h, 72h, 7d) post-administration to determine depletion kinetics.

  • Tissue-specific effects: Analyze both peripheral blood and lymphoid organs (spleen, lymph nodes) as depletion efficiency may vary between compartments.

Flow cytometry analysis protocol:

  • Stain with antibody combinations to identify Tregs: CD4, CD25 (using a non-competing clone), and Foxp3

  • Include functional markers (CTLA-4, GITR, Helios) to characterize remaining Tregs

  • Use viability dye to exclude dead cells

  • Analyze both percentage and absolute numbers of Tregs

Depletion efficiency measurement:
Research has demonstrated variable depletion efficiency depending on the immunological context. For example, one study showed 73% reduction in normal mice versus 93% reduction during lymphodepletion (P = 0.0001) . This context-dependent effect should be carefully assessed in each experimental system.

Functional assessment of remaining Tregs:
After antibody treatment, assess the suppressive capacity of remaining Tregs using:

  • In vitro suppression assays with conventional T cells

  • Cytokine production analysis (IL-10, TGF-β)

  • Expression of functional markers (CTLA-4, CD39)

Considerations for combination therapies:
When combining IL2RA monoclonal antibody treatment with other immunotherapies (e.g., vaccination, checkpoint inhibitors), assess the timing and sequence of administration. Research has shown that during lymphodepletion, IL2Rα blockade can decrease Tregs without impairing effector T-cell responses, while in normal mice, it may abolish vaccine-induced immune responses .

How do different epitope specificities of IL2RA monoclonal antibodies affect experimental outcomes?

Different epitope specificities of IL2RA monoclonal antibodies significantly influence experimental outcomes through multiple mechanisms that researchers must consider:

Functional domain targeting:
IL2RA has distinct structural domains involved in different functions:

  • The IL-2 binding site (critical for cytokine recognition)

  • The receptor trimerization interface (important for forming the high-affinity receptor complex)

  • Intracellular signaling regions

Antibodies targeting the IL-2 binding domain typically demonstrate stronger neutralizing capacity than those targeting other regions . The specific epitope targeted determines whether the antibody will:

  • Block IL-2 binding

  • Prevent receptor complex formation

  • Induce receptor internalization

  • Trigger antibody-dependent cellular cytotoxicity (ADCC)

Experimental impact analysis:
The following table summarizes how epitope specificity affects different experimental applications:

Epitope RegionEffect on IL-2 BindingCell Depletion EfficiencyBest Applications
IL-2 Binding SiteHigh blockadeVariableSignaling inhibition studies
Trimerization InterfaceModerate blockadeModerateReceptor assembly research
Non-functional DomainsMinimal interferenceLowDetection and localization

Clone-specific considerations:
Different monoclonal antibody clones (e.g., MEM-181, 7D4) recognize distinct epitopes on IL2RA . When selecting an antibody for research:

  • Review the epitope information provided by manufacturers

  • Consider testing multiple clones if the exact epitope is unknown

  • Select clones validated for your specific application

  • Be consistent with clone selection throughout a research project

Cross-species reactivity implications:
Epitope conservation between species varies considerably. Some epitopes are highly conserved between human and mouse IL2RA, while others differ significantly. Consider that:

  • Antibodies raised against human IL2RA may not recognize the same epitope in mouse IL2RA

  • IL2RA orthologs have been identified in multiple species including mouse, rat, bovine, chimpanzee, and chicken

  • Cross-reactivity testing is essential when working with different species models

Methodological recommendation:
When studying IL2RA functionality, researchers should characterize the epitope specificity of their chosen antibody through competitive binding assays with known ligands or other antibodies with defined epitopes. This characterization will enable more precise interpretation of experimental results.

How should researchers interpret flow cytometry data when analyzing IL2RA expression in heterogeneous cell populations?

Interpreting flow cytometry data for IL2RA expression in heterogeneous cell populations requires nuanced analytical approaches that account for biological variability and technical considerations:

Gating strategy methodology:
Implement a systematic gating approach:

  • Exclude debris using forward/side scatter

  • Apply doublet discrimination

  • Use viability dye to exclude dead cells

  • Apply lineage markers to identify major populations

  • Within each population, analyze IL2RA expression using properly set thresholds

Expression pattern analysis:
IL2RA expression follows distinct patterns in different cell populations:

  • High expression: Activated T cells, particularly Tregs (CD4+CD25+Foxp3+)

  • Intermediate expression: Recently activated conventional T cells, some activated B cells

  • Low/inducible expression: Resting lymphocytes (expression is lost on resting B and T lymphocytes)

  • Tissue-specific variations: Expression is notably higher in tonsil, spleen, lymph node, and cerebellum compared to other tissues

Quantitative assessment approaches:
Beyond simple positive/negative classification, consider:

  • Mean/median fluorescence intensity (MFI): Quantifies expression level per cell

  • Percent positive: Determines fraction of cells expressing IL2RA

  • Expression density calculation: Combines percent positive and MFI for comprehensive assessment

Multiparameter analysis strategy:
IL2RA should be analyzed in conjunction with other markers:

  • For Tregs: CD4, Foxp3, CD127(low)

  • For activated T cells: CD4/CD8, CD69, HLA-DR

  • For activated B cells: CD19, CD69, CD86

  • For T follicular regulatory cells: CXCR5, PD-1, Bcl-6

Common interpretation challenges and solutions:

ChallengeCauseSolution
False positivesNon-specific bindingInclude isotype controls; optimize staining buffer
Low signalReceptor internalizationUse freshly isolated cells; optimize fixation protocol
Heterogeneous expressionBiological variationUse density plots rather than histograms; analyze subpopulations separately
Background stainingDead cells, Fc receptor bindingInclude viability dye; use Fc blocking reagents

Technical considerations:
When analyzing samples from different experimental conditions (e.g., treatment vs. control):

  • Use standardized instrument settings

  • Apply consistent gating strategy

  • Include biological controls for accurate interpretation

  • Consider batch effects in longitudinal studies

By systematically applying these methodological approaches, researchers can generate robust and reproducible interpretations of IL2RA expression patterns in heterogeneous cell populations.

What control strategies are essential when using IL2RA monoclonal antibodies in immunoprecipitation experiments?

Implementing comprehensive control strategies is critical for generating reliable data when using IL2RA monoclonal antibodies in immunoprecipitation (IP) experiments:

Essential control methodologies:

  • Input controls:

    • Analyze a fraction (5-10%) of pre-IP lysate to confirm IL2RA presence

    • Use this control to assess IP efficiency by comparing band intensity

    • Essential for quantitative analysis of enrichment

  • Antibody controls:

    • Isotype control: Use matched isotype (typically IgG1 for IL2RA antibodies) from the same species

    • No-antibody control: Perform IP procedure without adding antibody

    • Blocking peptide control: Pre-incubate antibody with excess IL2RA peptide to confirm specificity

  • Sample controls:

    • Negative cell line: Use cells known not to express IL2RA

    • Positive cell line: Use cells with confirmed high IL2RA expression (e.g., activated T cells)

    • Knockdown/knockout validation: Use IL2RA-silenced cells to confirm specificity

  • Technical controls:

    • Heavy chain control: Use anti-IgG light chain secondary antibodies for detection to avoid heavy chain interference

    • Cross-linking efficiency control: When using cross-linking agents, include controls to assess cross-linking efficiency

    • Buffer composition controls: Test multiple lysis buffers to optimize extraction while maintaining protein-protein interactions

Protocol optimization strategies:

For IL2RA immunoprecipitation, consider these methodological modifications:

  • Use gentle lysis buffers (e.g., 1% NP-40 or 0.5% CHAPS) to preserve membrane protein integrity

  • Pre-clear lysates with protein A/G beads to reduce non-specific binding

  • Optimize antibody concentration based on IL2RA expression level

  • Include protease inhibitors to prevent degradation of IL2RA (55 kDa glycoprotein)

  • For co-immunoprecipitation studies, consider crosslinking to stabilize transient interactions

Validation methodology:

After immunoprecipitation, validate results through complementary approaches:

  • Reverse IP: Use antibodies against potential interacting partners to IP IL2RA

  • Mass spectrometry: Confirm identity of precipitated proteins

  • Functional assays: Validate biological significance of identified interactions

Troubleshooting guidance:

IssuePotential CauseSolution
No IL2RA detectionInsufficient expressionUse activated lymphocytes with high expression
Antibody epitope disruptionTry alternative clone or lysis conditions
High backgroundIncomplete washingIncrease wash stringency
Non-specific bindingInclude additional blocking steps
Multiple bandsPost-translational modificationsConfirm with deglycosylation experiments
DegradationAdd additional protease inhibitors

By implementing these control strategies, researchers can generate robust and reproducible immunoprecipitation data when studying IL2RA and its interactions.

How can researchers resolve discrepancies in IL2RA detection between different experimental techniques?

Resolving discrepancies in IL2RA detection across different experimental techniques requires systematic analysis of technique-specific variables and biological considerations:

Methodological comparison analysis:

Different techniques detect IL2RA through distinct mechanisms:

  • Flow cytometry: Detects surface/intracellular protein in intact cells

  • Western blot: Detects denatured protein based on molecular weight

  • Immunohistochemistry: Detects protein in fixed tissue with spatial context

  • qPCR: Measures mRNA expression, not protein

  • ELISA: Detects soluble or captured protein in solution

Common discrepancy patterns and resolution strategies:

  • Flow cytometry positive, Western blot negative:

    • Cause: Epitope denaturation during SDS-PAGE

    • Resolution: Try native gel electrophoresis or use antibodies recognizing linear epitopes

    • Verification: Use multiple antibody clones targeting different epitopes

  • qPCR positive, protein detection negative:

    • Cause: Post-transcriptional regulation or rapid protein turnover

    • Resolution: Assess protein stability using proteasome inhibitors

    • Verification: Measure half-life through pulse-chase experiments

  • Discrepancies between antibody clones:

    • Cause: Different epitope specificity, accessibility, or affinity

    • Resolution: Map epitopes or use antibody panels

    • Verification: Test with recombinant IL2RA protein as positive control

Systematic troubleshooting protocol:

  • Sample preparation analysis:

    • Ensure consistent preparation across techniques

    • Consider effects of fixation, permeabilization, and denaturation

    • Compare fresh vs. frozen samples

  • Antibody validation approach:

    • Confirm antibody compatibility with each technique

    • Test multiple antibody clones (e.g., MEM-181, 7D4)

    • Validate using positive and negative controls

  • Biological variable assessment:

    • Consider IL2RA's dynamic expression patterns

    • Account for post-translational modifications (glycosylation)

    • Evaluate alternative splicing (multiple IL2RA mRNA variants exist)

  • Technical optimization strategy:
    For each technique, optimize:

    • Antibody concentration and incubation conditions

    • Blocking procedures to reduce background

    • Detection systems to ensure appropriate sensitivity

Unified experimental approach:
When discrepancies persist, implement a unified experimental design:

  • Split single samples for parallel processing by different techniques

  • Include gradient of positive controls (e.g., unstimulated, partially activated, fully activated T cells)

  • Analyze time-course samples to capture dynamic expression changes

  • Document all protocol variables for comprehensive comparison

Alternative validation methods:
When conventional techniques yield conflicting results:

  • Use genetic approaches (siRNA knockdown, CRISPR knockout)

  • Employ functional assays (IL-2 binding, downstream signaling)

  • Consider mass spectrometry for unbiased protein detection

By systematically applying these approaches, researchers can identify the sources of discrepancies and develop a coherent understanding of IL2RA expression across different experimental contexts.

How can IL2RA monoclonal antibodies be optimally used in cancer immunotherapy research models?

Optimizing IL2RA monoclonal antibodies in cancer immunotherapy research requires careful consideration of multiple variables across experimental design, dosing strategies, and combination approaches:

Mechanistic foundations for experimental design:
IL2RA monoclonal antibodies can function through multiple mechanisms in cancer immunotherapy:

  • Regulatory T cell depletion: Removing immunosuppressive Tregs from the tumor microenvironment

  • Effector T cell modulation: Altering IL-2 signaling in effector cells

  • Immune checkpoint regulation: Modifying T cell activation thresholds

  • Antibody-dependent cellular cytotoxicity (ADCC): Triggering immune-mediated killing of IL2RA+ cells

Dosing and administration optimization:
Research has demonstrated that timing and context significantly impact outcomes:

  • During lymphodepletion, IL-2Rα blockade can decrease Tregs (93% reduction; P = 0.0001) without impairing effector T-cell responses

  • This approach significantly augments therapeutic antitumor efficacy (66% reduction in tumor growth; P = 0.0024)

  • In contrast, identical treatment in normal mice impaired vaccine-induced effector responses

Combination therapy experimental design:
The following methodological approaches optimize combinatorial strategies:

  • With chemotherapy:

    • Sequence IL2RA antibody after lymphodepleting chemotherapy (e.g., temozolomide)

    • This sequencing enhances Treg depletion while preserving effector function

    • Monitor recovery kinetics to determine optimal timing

  • With cancer vaccines:

    • In non-lymphodepleted models: Caution required as IL2RA blockade may abolish vaccine responses

    • In lymphodepleted models: Synergistic enhancement observed with sequential administration

    • Assess tumor-specific immune responses to confirm efficacy

  • With checkpoint inhibitors:

    • Test varying sequences (concurrent vs. sequential)

    • Monitor for potential enhancement of immune-related adverse events

    • Evaluate both antitumor efficacy and toxicity profiles

Monitoring parameters for comprehensive assessment:

ParameterMethodologySignificance
Treg depletionFlow cytometry (CD4+CD25+Foxp3+)Confirms primary mechanism
Effector responseIFN-γ ELISPOT, cytotoxicity assaysAssesses functional immunity
Tumor infiltrationImmunohistochemistry, flow cytometryEvaluates local immune effects
Systemic effectsCytokine profiling, immune cell phenotypingMonitors off-target effects

Tumor model selection considerations:
Different tumor models may respond distinctly to IL2RA antibody therapy:

  • Immunologically "hot" tumors: May show more pronounced responses due to pre-existing T cell infiltration

  • Immunologically "cold" tumors: May require combination approaches to enhance infiltration

  • Regulatory T cell-dependent models: Likely to show greater benefit from IL2RA targeting

Translational considerations:
When designing preclinical studies with potential clinical translation:

  • Use humanized models where possible

  • Consider IL2RA polymorphisms that may influence response

  • Include biomarker analysis to identify predictors of response

  • Assess potential toxicities, particularly autoimmune manifestations

By systematically applying these methodological approaches, researchers can optimize the use of IL2RA monoclonal antibodies in cancer immunotherapy models and generate more translatable preclinical data.

What are the critical considerations when using IL2RA monoclonal antibodies in autoimmune disease research?

Using IL2RA monoclonal antibodies in autoimmune disease research requires careful methodological consideration of multiple factors that influence experimental outcomes and translational relevance:

Genetic and polymorphism considerations:
Research has identified IL2RA polymorphisms associated with autoimmune conditions:

  • Polymorphisms in IL2RA (rs2104286, rs41295061, rs35285258) show significant associations with multiple sclerosis

  • The minor allele frequency for rs2104286 was significantly lower in MS patients compared with controls

  • These genetic variations may influence antibody binding and therapeutic efficacy

Experimental design strategies:
When designing autoimmune disease studies:

  • Model selection:

    • Choose models that accurately reflect the role of IL2RA in human disease

    • Consider both induced models (EAE for MS) and spontaneous models (NOD for type 1 diabetes)

    • Humanized models may better reflect human IL2RA biology

  • Timing optimization:

    • Test prophylactic (before disease onset) vs. therapeutic (after establishment) administration

    • Assess intervention during different disease phases (initiation, progression, remission)

    • Evaluate long-term vs. short-term blockade effects

  • Dose-response assessment:

    • Conduct comprehensive dose-titration studies

    • Examine both partial and complete IL2RA blockade effects

    • Monitor Treg depletion efficiency at different doses

Monitoring parameters:
Comprehensive assessment should include:

ParameterMethodologyRelevance
Disease activityClinical scoring, histopathologyPrimary outcome measure
Regulatory T cell dynamicsFlow cytometry, functional assaysMechanism verification
Effector T cell functionCytokine production, proliferationOff-target effects
Autoantibody productionELISA, immunofluorescenceB cell-mediated effects
Tissue-specific inflammationHistology, imagingLocalized disease effects

Paradoxical effects management:
IL2RA blockade may produce seemingly contradictory effects in autoimmune contexts:

  • Treg depletion: May worsen autoimmunity by removing suppressive cells

  • Activation inhibition: May improve autoimmunity by blocking pathogenic T cell activation

  • IL-2 availability: Blocking IL2RA may increase IL-2 availability for cells using intermediate-affinity receptors

Human disease-specific considerations:
For translational relevance, consider:

  • Multiple sclerosis: IL2RA-specific humanized monoclonal antibody showed promising therapeutic effects

  • Type 1 diabetes: Consider IL2RA's association with IDDM10 (insulin-dependent diabetes mellitus locus 10)

  • Inflammatory bowel disease: Disruption of IL2RA-related genes in mice leads to ulcerative colitis-like disease

Safety monitoring methodologies:
When blocking IL2RA in autoimmune models:

  • Monitor for infection susceptibility due to immune dysregulation

  • Assess for development of secondary autoimmune manifestations

  • Evaluate long-term effects on immune homeostasis

  • Document tissue-specific inflammatory changes

Combination therapy approaches:
Consider systematic testing of IL2RA antibodies with:

  • Standard-of-care immunosuppressants

  • Emerging biologics targeting complementary pathways

  • Cell-based therapies (e.g., adoptive Treg transfer)

By systematically applying these methodological considerations, researchers can generate more robust and translatable data on IL2RA monoclonal antibodies in autoimmune disease contexts.

How should researchers interpret conflicting data on IL2RA monoclonal antibody efficacy between different disease models?

Interpreting conflicting data on IL2RA monoclonal antibody efficacy between disease models requires systematic analysis of multiple variables that influence experimental outcomes:

Methodological framework for comparative analysis:

  • Model-specific factor assessment:
    Analyze fundamental differences between models:

    • Immune compartment composition: Treg/effector T cell ratios vary between models

    • Disease mechanisms: IL2RA may play central or peripheral roles

    • Kinetics: Acute vs. chronic disease progression alters intervention windows

    • Background strain influences: Genetic backgrounds modify immune responses

  • Antibody-specific variable analysis:
    Evaluate antibody characteristics that influence outcomes:

    • Epitope specificity: Different epitopes produce distinct functional effects

    • Isotype selection: Determines complement activation and Fc receptor engagement

    • Species cross-reactivity: Some antibodies may have different affinities across species

    • Pharmacokinetics: Half-life and tissue distribution vary between antibodies and models

  • Context-dependent mechanism evaluation:
    Research demonstrates that identical IL2RA antibody treatments can produce opposing effects in different contexts:

    • In normal mice: Anti-IL2Rα mAb abolishes vaccine-induced immune responses

    • During lymphodepletion: The same treatment augments therapeutic efficacy without impairing effector responses

Reconciliation strategies for conflicting data:

  • Direct comparative experimentation:

    • Test identical antibody clones across multiple models

    • Standardize dosing, timing, and assessment protocols

    • Include extensive controls and validation measures

  • Mechanism-focused investigation:

    • Trace IL-2 signaling pathway components across models

    • Compare Treg depletion efficiency and persistence

    • Examine compensatory mechanisms activated after IL2RA blockade

  • Parameter standardization:
    Define consistent parameters across studies:

    • Antibody concentration and exposure time

    • Readout timing and methodology

    • Analysis techniques and statistical approaches

Decision matrix for interpreting conflicting results:

VariableAssessment ApproachResolution Strategy
Disease stage differencesCompare intervention timing relative to disease courseStandardize to equivalent disease phases
Antibody clone variationsDirect comparison of multiple clonesIdentify most consistent performer across models
Dosing discrepanciesDose-response curves in each modelDetermine optimal dose for each specific model
Endpoint selectionComprehensive multiparameter assessmentEstablish hierarchical endpoint prioritization

Translational implications analysis:
When evaluating conflicting preclinical data for potential clinical translation:

  • Prioritize models that better recapitulate human disease pathophysiology

  • Consider human genetic data on IL2RA polymorphisms in the target disease

  • Evaluate whether contradictions reflect genuine biological differences or technical variables

  • Develop predictive biomarkers to identify responsive patient subsets

Systematic review methodology:
When conducting literature analysis of conflicting data:

  • Categorize studies by model, antibody characteristics, and experimental design

  • Weight evidence based on methodological rigor and reproducibility

  • Identify patterns that explain apparent contradictions

  • Generate testable hypotheses to resolve discrepancies

By systematically applying these analytical approaches, researchers can develop coherent interpretations of seemingly conflicting data on IL2RA monoclonal antibody efficacy across different disease models, leading to more effective translational strategies.

How can researchers leverage new IL2RA monoclonal antibody technologies for single-cell analysis of regulatory T cell heterogeneity?

Leveraging advanced IL2RA monoclonal antibody technologies for single-cell analysis of regulatory T cell heterogeneity requires integration of innovative methodological approaches:

Advanced antibody engineering applications:
Recent developments in antibody technology enable sophisticated single-cell analyses:

  • Site-specific conjugation: Precise attachment of fluorophores or metal tags at defined positions

  • Bispecific antibodies: Simultaneous targeting of IL2RA and other markers (e.g., Foxp3)

  • Recombinant antibody fragments: Better tissue penetration and reduced background

  • Photocleavable antibodies: Allow sequential staining and elution cycles

Multi-omics integration methodology:
Combine IL2RA antibody labeling with:

  • Single-cell RNA sequencing: Correlate IL2RA protein levels with transcriptomic profiles

  • CITE-seq: Simultaneously measure surface protein and gene expression

  • Single-cell ATAC-seq: Link chromatin accessibility with IL2RA expression

  • Spatial transcriptomics: Map IL2RA+ Tregs within tissue microenvironments

High-dimensional flow cytometry protocol:
Develop comprehensive IL2RA-focused cytometry panels:

  • Start with core Treg markers (CD4, IL2RA, Foxp3)

  • Add functional markers (CTLA-4, GITR, LAG-3, CD39)

  • Include markers of Treg subsets (Helios, Nrp1, CCR4, CCR7)

  • Incorporate transcription factors (BLIMP-1, c-MAF)

  • Add tissue-specific markers based on anatomical location

Mass cytometry optimization:
For CyTOF analysis of IL2RA+ cell heterogeneity:

  • Label IL2RA antibodies with rare earth metals

  • Develop panels with 30+ parameters

  • Implement dimensionality reduction (tSNE, UMAP)

  • Apply clustering algorithms to identify novel Treg subpopulations

Spatial analysis approaches:
For tissue-level understanding of IL2RA+ cell distribution:

  • Multiplex immunofluorescence: Simultaneously visualize 8+ markers including IL2RA

  • Imaging mass cytometry: Achieve subcellular resolution with 40+ markers

  • 4D analysis: Track IL2RA+ cells spatiotemporally using intravital microscopy

Functional correlation strategies:
Link IL2RA expression patterns to Treg functionality:

  • Single-cell cytokine secretion: Correlate IL2RA levels with suppressive cytokine production

  • TCR sequencing: Connect IL2RA expression with TCR specificity

  • Suppression assays: Sort IL2RA subpopulations for functional testing

  • Epigenetic analysis: Assess TSDR demethylation status in IL2RA subsets

IL2RA heterogeneity mapping framework:
Based on current research, IL2RA expression distinguishes multiple functionally distinct Treg populations:

  • IL2RA^high Foxp3^high: Stable, highly suppressive Tregs

  • IL2RA^int Foxp3^int: Potentially unstable or transitioning Tregs

  • IL2RA^high Foxp3^low: Activated conventional T cells

  • IL2RA^low Foxp3^high: Variant Treg population with distinct functionality

By systematically implementing these advanced methodological approaches, researchers can comprehensively characterize the heterogeneity of IL2RA-expressing regulatory T cell populations at unprecedented resolution, leading to new insights into immune regulation and therapeutic targeting strategies.

What are the methodological considerations for studying IL2RA-targeted approaches in combination with checkpoint inhibitors?

Studying IL2RA-targeted approaches in combination with checkpoint inhibitors requires systematic methodological consideration of multiple variables that influence experimental outcomes and translational relevance:

Mechanistic interaction assessment:
IL2RA blockade and checkpoint inhibition may interact through multiple mechanisms:

  • Treg modulation: IL2RA antibodies deplete Tregs while checkpoint inhibitors may reduce Treg suppressive function

  • Effector T cell priming: Both approaches may enhance effector responses through distinct pathways

  • Cytokine feedback loops: IL-2 signaling changes from IL2RA blockade may alter responsiveness to checkpoint inhibition

Sequential vs. concurrent administration protocol design:
Rigorous testing of administration schedules is critical:

  • IL2RA antibody first: May create favorable immune landscape by reducing Tregs before checkpoint blockade

  • Checkpoint inhibitor first: May activate T cells before altering IL-2 responsiveness

  • Concurrent administration: May provide synergistic enhancement but potentially increase toxicity

  • Intermittent scheduling: May optimize efficacy while reducing adverse events

Dose optimization strategy:
For combination approaches, standard monotherapy dosing may not be optimal:

  • Conduct full dose-ranging studies for both agents alone and in combination

  • Test fixed dose of one agent with variable doses of the other

  • Consider adaptive dosing based on pharmacodynamic markers (e.g., Treg levels)

Biomarker analysis methodology:
Comprehensive biomarker assessment should include:

Biomarker CategoryExamplesRelevance
CellularTreg/Teff ratio, CD8+ T cell activationDirect pharmacodynamic readouts
SolubleCytokines (IL-2, IFNγ), soluble IL2RASystemic immune activation
GeneticIL2RA polymorphisms, tumor mutational burdenResponse prediction
TissueImmune infiltration, spatial relationshipsLocal immune environment

Toxicity monitoring protocol:
Enhanced immune activation may increase adverse events:

  • Implement systematic toxicity grading across organ systems

  • Monitor for both overlapping and unique toxicities

  • Develop intervention algorithms for managing immune-related adverse events

  • Establish predictive biomarkers for toxicity risk

Model selection considerations:
Different models provide complementary insights:

  • Syngeneic models: Allow full assessment of immune components but lack human-specific interactions

  • Humanized models: Better recapitulate human IL2RA biology but have technical limitations

  • Ex vivo human systems: Enable testing on patient-derived samples but lack in vivo complexity

  • 3D organoid co-cultures: Bridge gap between 2D culture and in vivo models

Resistance mechanism evaluation:
Protocol for investigating adaptive resistance:

  • Compare tumors progressing on combination vs. single-agent therapy

  • Analyze changes in IL2RA expression patterns after treatment

  • Assess alternative checkpoint receptor upregulation

  • Evaluate changes in antigen presentation machinery

Translational pathway design:
When designing studies with clinical translation potential:

  • Prioritize clinically relevant endpoints and biomarkers

  • Consider practical administration schedules feasible in patients

  • Develop companion diagnostics to identify optimal responders

  • Establish robust safety monitoring protocols

By systematically implementing these methodological considerations, researchers can generate more rigorous and translatable data on IL2RA-targeted approaches in combination with checkpoint inhibitors, potentially leading to more effective immunotherapy strategies.

How should researchers evaluate and synthesize the current state of IL2RA monoclonal antibody research across different fields?

Evaluating and synthesizing the current state of IL2RA monoclonal antibody research requires a systematic methodological framework that integrates findings across diverse research domains:

Structured review methodology:
Implement a comprehensive approach to literature assessment:

  • Systematic search strategy: Use standardized terms (IL2RA, CD25, interleukin-2 receptor alpha) across multiple databases

  • Cross-disciplinary inclusion: Incorporate immunology, oncology, autoimmunity, and translational medicine

  • Quality assessment: Evaluate methodological rigor, reproducibility, and validation approaches

  • Temporal analysis: Track evolution of understanding and technological advances over time

Convergent findings identification:
Despite methodological differences, certain findings show consistency across fields:

  • IL2RA monoclonal antibodies effectively deplete CD4+CD25+Foxp3+ regulatory T cells

  • The efficacy and impact of IL2RA targeting is highly context-dependent

  • IL2RA plays significant roles in both autoimmunity and cancer immunobiology

  • Epitope specificity critically influences functional outcomes of antibody binding

Divergent results reconciliation:
When findings appear contradictory:

  • Analyze methodological differences that may explain discrepancies

  • Consider biological contexts (normal vs. lymphodepleted, cancer vs. autoimmunity)

  • Evaluate antibody characteristics (epitope, isotype, affinity)

  • Assess model-specific variables (species, strain, disease model)

Gap analysis framework:
Systematically identify knowledge gaps requiring further investigation:

  • Mechanistic understanding: Incomplete characterization of IL2RA signaling complexity

  • Biomarker development: Limited predictive markers for therapeutic response

  • Combination strategies: Insufficient systematic assessment of synergistic approaches

  • Long-term effects: Inadequate longitudinal studies on persistent immune changes

Translational bridge assessment:
Evaluate the strength of evidence supporting clinical application:

  • Analyze correlation between preclinical and clinical outcomes

  • Identify predictive biomarkers validated across species

  • Assess reproducibility of key findings in human systems

  • Evaluate risk-benefit profiles across different therapeutic contexts

Future direction prioritization:
Based on current state analysis, prioritize research avenues:

  • Technological innovation: Developing next-generation IL2RA-targeting approaches

  • Mechanistic refinement: Elucidating context-specific roles of IL2RA signaling

  • Combination optimization: Systematically mapping synergistic therapeutic strategies

  • Personalization approaches: Identifying patient-specific factors influencing response

Integration with broader immunology concepts:
Position IL2RA research within evolving immunological paradigms:

  • Connect with emerging understanding of Treg plasticity and heterogeneity

  • Incorporate findings on IL-2 signaling complexity and selectivity

  • Consider relationships with other immunomodulatory pathways

  • Evaluate in context of tissue-specific immune regulation

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 2025 TheBiotek. All Rights Reserved.