IL18 Antibody refers to monoclonal antibodies (mAbs) designed to neutralize Interleukin-18 (IL-18), a proinflammatory cytokine critical in innate and adaptive immune responses. IL-18 is structurally related to the IL-1 family, exists as a 193-amino acid precursor (24 kDa) that is cleaved into an 18 kDa active form by caspase-1 during inflammasome activation . These antibodies target IL-18 to inhibit its binding to IL-18 receptors (IL-18Rα and IL-18Rβ), thereby blocking downstream signaling pathways involved in interferon-gamma (IFN-γ) production, macrophage activation, and Th1/Tc1 cell differentiation .
IL18 antibodies bind to epitopes on IL-18, preventing its interaction with receptors. This inhibition suppresses:
IL18 antibodies are investigated for diseases driven by IL-18 overproduction:
Autoimmune/Inflammatory Diseases
Very Early-Onset Inflammatory Bowel Disease (VEOIBD): Anti-IL-18 mAb (GSK1070806) induced sustained remission in a pediatric patient with IL-18opathy, enabling discontinuation of parenteral nutrition .
Crohn’s Disease/Sepsis: Elevated IL-18 levels correlate with disease severity, making it a biomarker for targeted therapy .
Cancer Immunotherapy
IL-18 Regulation Complexity: IL-18 activity is tightly regulated by IL-18BP, which binds IL-18 with high affinity. Neutralizing IL-18 directly or blocking IL-18BP requires precise dosing to avoid excessive immune activation .
Off-Target Effects: Anti-IL-18 antibodies may suppress beneficial IL-18 functions, such as anti-tumor immunity in certain contexts .
Biomarker Validation: IL-18 levels vary widely among patients, necessitating biomarker-driven trials to identify responders .
Combination Therapies: Pairing anti-IL-18 antibodies with checkpoint inhibitors (e.g., anti-PD-1) to modulate tumor immunity .
Precision Medicine: Stratifying patients based on IL-18/IL-18BP ratios to optimize therapeutic outcomes .
Novel Antibody Formats: Developing bispecific antibodies or engineered variants with enhanced stability or half-life.
IL-18 (Interleukin-18, also known as IL-1F4) is a proinflammatory cytokine belonging to the IL-1 family that acts as a potent inducer of T helper 1 and cytotoxic responses. It is expressed as a 24 kDa precursor by various cell types including endothelial cells, epithelial cells, keratinocytes, gamma δ T cells, and phagocytes. This precursor is activated intracellularly through Caspase-1 mediated proteolysis, releasing the 17 kDa mature cytokine .
IL-18 exerts distinct immune effects depending on the local cytokine environment. Its primary functions include stimulating IFN-gamma production by T and NK cells and inducing CD8+ T cell and NK-mediated cytotoxicity, making it central to host defense mechanisms . In experimental settings, IL-18 has been shown to stimulate IFN-gamma secretion in certain cell lines (such as KG-1 human acute myelogenous leukemia cells) in a dose-dependent manner, particularly in the presence of other cytokines like TNF-alpha .
IL-18 antibodies are utilized across multiple experimental applications in immunological research. Based on published data, the most frequently employed techniques include:
When designing experiments, researchers should note that optimal dilutions are often sample-dependent and should be determined empirically for each experimental system .
Validating antibody specificity is critical for ensuring reliable research outcomes. For IL-18 antibodies, a comprehensive validation approach should include:
Positive and negative controls: Test the antibody against tissues or cell lines known to express IL-18 (such as U2OS, HeLa, HepG2, or A549 cells) versus those with low or no expression .
Knockout/knockdown validation: Compare antibody reactivity in wild-type versus IL-18 knockout or knockdown samples. Published studies have used this approach to confirm specificity .
Peptide competition assays: Pre-incubate the antibody with recombinant IL-18 protein before application to demonstrate binding specificity.
Multiple detection methods: Confirm findings using different techniques (e.g., if using WB, validate with IHC or IF).
Cross-reactivity testing: If working across species, verify whether the antibody recognizes IL-18 from different organisms (human, mouse, rat) as specified in product information .
When interpreting validation results, researchers should carefully examine the molecular weight of detected proteins (typically 22-24 kDa for IL-18) and compare this with expected values from literature .
Designing a robust IL-18 neutralization bioassay requires careful consideration of cell types, stimulation conditions, and readout parameters. A methodologically sound approach would include:
Cell selection: KG-1 human acute myelogenous leukemia cells are commonly used for IL-18 bioassays due to their responsiveness to IL-18 stimulation .
Stimulation conditions:
Experimental setup:
Pre-incubate IL-18 with various concentrations of the test antibody
Add this mixture to cells (with TNF-alpha)
Include appropriate controls: unstimulated cells, IL-18+TNF-alpha without antibody, isotype control antibody
Readout: Measure IFN-gamma secretion in cell supernatants via ELISA as the primary functional readout
Data analysis: Calculate the neutralization dose (ND50), defined as the antibody concentration that inhibits 50% of the cytokine activity. For anti-IL-18 antibodies, ND50 values typically range from 0.05-0.3 μg/mL .
This approach has been validated in published research, where antibodies like h18-108 demonstrated inhibition of IFN-gamma production in KG-1 cells, confirming their neutralizing activity .
Successful immunohistochemistry (IHC) with IL-18 antibodies requires optimization of several critical parameters:
Antigen retrieval: For formalin-fixed paraffin-embedded tissues, published protocols recommend:
Antibody dilution: Starting with a range of 1:500-1:2000 is recommended, followed by optimization for specific tissue types .
Incubation conditions:
Temperature: Room temperature to 4°C
Duration: Typically 1-24 hours (overnight incubation often produces optimal results)
Detection system: HRP/DAB systems are commonly used, with amplification steps recommended for low-abundance targets.
Positive controls: Human tonsillitis tissue and human stomach cancer tissue have been validated as positive controls for IL-18 IHC .
Background reduction:
Use appropriate blocking sera based on secondary antibody host
Include washing steps with Tris-buffered saline containing 0.1% Tween-20
Consider specific blockers for endogenous peroxidase activity
Signal interpretation: IL-18 typically shows cytoplasmic localization, with potential nuclear positivity in some cell types.
Optimizing these parameters for each tissue type and experimental question will yield more reliable and reproducible results.
Differentiating between the precursor (pro-IL-18) and mature forms of IL-18 in Western blot analysis requires careful experimental design and interpretation:
Sample preparation considerations:
Use protease inhibitors to prevent artifactual processing
Include positive controls for both forms (recombinant pro-IL-18 and mature IL-18)
Consider cell fractionation to separate cytoplasmic (containing mature IL-18) from whole cell extracts
Gel resolution optimization:
Antibody selection:
Choose antibodies that recognize epitopes present in both forms
Alternatively, use antibodies specific to the pro-domain for selective detection
Interpretation guidelines:
Pro-IL-18 typically appears at ~24 kDa
Mature IL-18 appears at ~17-18 kDa
Confirm identities with recombinant protein standards
Verify using caspase-1 inhibitors (which should increase pro-IL-18 and decrease mature IL-18)
Quantification:
Calculate the ratio of mature to precursor forms as an indicator of IL-18 processing/activation
Normalize to appropriate loading controls
This methodological approach allows researchers to assess IL-18 processing in various experimental conditions, providing insights into the regulation of this important cytokine.
The epitope specificity of anti-IL-18 antibodies critically determines their neutralizing capacity through several mechanisms:
Receptor binding site targeting: Antibodies that recognize epitopes involved in receptor binding show superior neutralizing activity. For example, the human anti-IL-18 antibody h18-108 binds to site 3 of IL-18, which is associated with IL-18 receptor beta binding, effectively blocking the interaction and downstream signaling .
Conformational versus linear epitopes: Antibodies recognizing conformational epitopes often demonstrate better neutralizing capacity than those binding linear epitopes, as they can interfere with the three-dimensional structure necessary for receptor engagement.
Binding affinity considerations: Even among antibodies targeting the same functional domain, those with higher binding affinity (lower KD values in the nanomolar range) typically demonstrate superior neutralizing capacity .
Epitope mapping techniques: To characterize epitope specificity, researchers can employ:
Functional implications: Different epitope specificities may result in:
This explains why antibodies binding to IL-18 with similar affinity can display drastically different neutralizing capacities, as observed with antibodies 441 and 445 in anti-IL-18BP studies .
Developing antibodies capable of disrupting preformed IL-18/IL-18BP complexes represents an advanced research objective with therapeutic potential. Several strategies have proven effective:
Epitope-focused selection strategy:
Target epitopes at the IL-18/IL-18BP interface rather than distal binding sites
Screen antibody libraries specifically for clones that compete with IL-18BP binding
Characterize binding to IL-18BP in the presence of IL-18 to identify disruptive candidates
Time-course disruption assays:
Form IL-18/IL-18BP complexes prior to antibody addition
Add candidate antibodies at various timepoints (0h, 0.5h, 2h) after complex formation
Measure free IL-18 levels to assess complex disruption capacity
This approach successfully identified antibody 445, which could release IL-18 from preformed complexes even when added hours after complex formation, while antibody 441 with similar affinity could not .
Free IL-18 detection methodology:
Binding kinetics optimization:
In vivo validation:
These strategies have translational potential for enhancing anti-tumor immune responses by liberating endogenous IL-18 from inhibitory complexes with IL-18BP .
Addressing cross-reactivity challenges in multi-species IL-18 research requires systematic methodological approaches:
Comprehensive antibody validation across species:
Sequence homology analysis:
Identify conserved and variable regions between species
Select antibodies targeting highly conserved epitopes for cross-species applications
For species-specific detection, choose antibodies recognizing divergent regions
Experimental design accommodations:
When possible, use species-matched systems (human antibodies for human samples)
For xenograft models, employ antibodies validated for both host and graft species
Include appropriate isotype controls from the same species as the primary antibody
Technical optimization by application:
For IHC/IF: Optimize antigen retrieval and antibody concentration separately for each species
For WB: Adjust protein loading and exposure times to account for affinity differences
For functional assays: Determine species-specific neutralization potencies and adjust dosing accordingly
Reporting standards:
Clearly document species reactivity in publications
Specify which antibody clones work across species versus those with restricted reactivity
Include validation data demonstrating specific reactivity in supplementary materials
Following these methodological guidelines will enhance data quality and reproducibility in comparative IL-18 studies spanning multiple species models.
The limited clinical efficacy of IL-18-based cancer immunotherapies despite encouraging preclinical data can be attributed to several mechanisms:
IL-18 binding protein interference:
Tumor microenvironment adaptation:
Cancer cells may downregulate IL-18 receptors
Immunosuppressive factors in tumor microenvironments can counteract IL-18's pro-inflammatory effects
Regulatory T cells can be induced that dampen IL-18-mediated responses
Pharmacokinetic limitations:
Short half-life of recombinant IL-18 in circulation
Poor tumor penetration of administered cytokines
Suboptimal dosing schedules in early clinical trials
Alternative strategies with improved potential:
Administration of mutated IL-18 forms that don't bind IL-18BP but retain receptor binding capability showed promising effects in mouse cancer models
Anti-IL-18BP antibodies demonstrated marked benefits in cancer models without significant safety concerns, likely because effects remained localized to the tumor microenvironment
Combination approaches with checkpoint inhibitors may overcome resistance mechanisms
These insights suggest that strategies targeting IL-18BP rather than directly administering IL-18 may prove more effective for enhancing anti-tumor immune responses .
Standardizing IL-18/IL-18BP ratio measurements for biomarker applications in inflammatory conditions requires addressing several methodological challenges:
Assay selection and optimization:
Sample processing standardization:
Establish consistent collection protocols (timing, anticoagulants)
Define optimal storage conditions (-70°C with minimal freeze-thaw cycles)
Document pre-analytical variables that affect measurements
Calibration and reference standards:
Use international reference preparations when available
Include multi-point calibration curves with recombinant proteins
Develop disease-specific reference ranges from appropriate control populations
Quality control measures:
Include internal controls spanning low, medium, and high concentrations
Participate in external quality assessment programs
Document inter-assay and intra-assay coefficients of variation
Reporting format standardization:
Express results as molar ratios rather than concentration ratios
Account for the 1:1 binding stoichiometry between IL-18 and IL-18BP
Calculate free IL-18 index based on binding kinetics models
Validation across disease states:
Establish reference ranges in healthy controls
Determine diagnostic performance characteristics (sensitivity, specificity)
Evaluate prognostic value through longitudinal studies
This standardized approach would facilitate the clinical utility of IL-18/IL-18BP measurements as biomarkers for inflammatory disease activity and treatment response monitoring.
Engineering IL-18 antibodies with enhanced therapeutic properties represents an active research frontier with several promising approaches:
Epitope-focused optimization:
Antibody format engineering:
Progress from single-chain (scFv) to Fab and complete IgG formats for enhanced affinity and half-life, as demonstrated with h18-108
Explore bispecific antibodies targeting both IL-18 and complementary inflammatory mediators
Develop smaller formats (nanobodies, affibodies) for enhanced tissue penetration
Fc engineering strategies:
Modify Fc regions to extend half-life (YTE or LS mutations)
Enhance or eliminate effector functions (ADCC, CDC) depending on therapeutic objectives
Engineer pH-dependent binding for improved intracellular trafficking
Tissue-targeted approaches:
Develop antibody-drug conjugates targeting IL-18 in specific tissue microenvironments
Create bispecific constructs linking anti-IL-18 domains with tissue-homing domains
Design antibodies with conditional activation in disease microenvironments
Functional screening innovations:
Employ cell-based reporter systems for high-throughput functional screening
Develop in vitro assays that better predict in vivo efficacy
Utilize machine learning to predict antibody properties from sequence data
These engineering approaches aim to overcome the limitations of earlier IL-18-targeted therapeutics while minimizing potential safety concerns associated with systemic IL-18 neutralization .
Anti-IL-18BP antibodies represent a distinct approach from conventional anti-IL-18 antibodies, with important mechanistic and application differences:
Mechanism of action:
Functional classification:
Anti-IL-18BP antibodies can be further categorized based on their specific activity:
Experimental applications:
Anti-IL-18BP antibodies are valuable for:
Anti-IL-18 antibodies are useful for:
Investigating IL-18's role in inflammatory pathology
Therapeutic approaches to autoimmune conditions
Neutralizing excessive IL-18 in models of hyperinflammation
Technical considerations:
Binding affinity: Both antibody types show comparable IL-18BP binding affinity (KD) in the low nanomolar range
Specificity validation: Anti-IL-18BP antibodies require testing in IL-18BP knockout models
Complex formation assessment: Anti-IL-18BP antibodies may require specialized assays to detect free versus complexed IL-18
These differences explain why researchers must carefully select antibodies based on whether they aim to enhance or inhibit IL-18 signaling in their experimental systems.
Cutting-edge technologies are revolutionizing research into IL-18 signaling complexes and antibody interactions:
Advanced structural biology approaches:
Cryo-electron microscopy (cryo-EM) for visualizing IL-18/receptor/antibody complexes at near-atomic resolution
Single-particle reconstruction techniques revealing conformational changes upon binding
Molecular dynamics simulations predicting binding energetics and conformational changes
Biointerface characterization technologies:
Advanced immunoprecipitation approaches:
Proximity-dependent biotin identification (BioID) for mapping protein interaction networks
Cross-linking mass spectrometry (XL-MS) for identifying interaction interfaces
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) for mapping conformational changes
Live-cell imaging innovations:
Fluorescence resonance energy transfer (FRET) sensors for monitoring IL-18/receptor interactions
Single-molecule tracking of IL-18 signaling components
Lattice light-sheet microscopy for visualizing signaling complex formation
Functional genomics integration:
CRISPR-Cas9 screening for identifying novel components of IL-18 signaling
Proteomics of isolated complexes (antibody pull-downs followed by mass spectrometry)
Phosphoproteomics for mapping downstream signaling events
These technologies are enabling researchers to move beyond simple binding studies toward understanding the structural and dynamic aspects of IL-18 signaling complexes and how therapeutic antibodies modulate these interactions.
Designing IL-18 antibody derivatives with pathway-selective modulation capabilities represents a frontier in cytokine engineering:
Receptor complex-specific targeting:
IL-18 signals through a heterodimeric receptor complex (IL-18Rα and IL-18Rβ)
Developing antibodies that selectively disrupt interaction with only one receptor component could create biased signaling
Targeting site 3 of IL-18 (IL-18Rβ binding site) versus other epitopes provides different functional outcomes
Context-dependent activation strategies:
Engineer antibody fragments that become activating only in specific tissue microenvironments
Develop pH-sensitive antibodies that change conformation or binding properties in acidic tumor environments
Create protease-activated antibodies that unmask activity only in inflammatory settings
Bispecific and multispecific formats:
Link anti-IL-18 binding domains with domains targeting tissue-specific markers
Create constructs that simultaneously engage IL-18 and its receptor to enhance signaling in specific contexts
Develop antibodies that simultaneously block IL-18BP and stabilize IL-18/receptor interactions
Intracellular delivery approaches:
Design cell-penetrating antibody derivatives to modulate intracellular IL-18 processing
Target precursor versus mature IL-18 forms with different specificity profiles
Develop antibody-small molecule conjugates for selective pathway modulation
Engineered binding kinetics:
Tune antibody binding kinetics (kon and koff rates) to alter signaling duration
Create antibodies with temperature-dependent binding properties for spatiotemporal control
Develop antibodies with cooperative binding properties for threshold-dependent effects
These innovative approaches move beyond simple blockade toward precise modulation of IL-18 signaling networks, potentially addressing the limitations of current IL-18-targeted therapies in cancer and inflammatory diseases .
Interleukin-18 (IL-18) is a proinflammatory cytokine that plays a crucial role in both innate and adaptive immune responses. It was originally identified as an interferon-gamma (IFN-γ) inducing factor and is produced by various cell types, including macrophages, dendritic cells, and epithelial cells . IL-18 is involved in the regulation of immune responses, inflammation, and tissue repair.
IL-18 is synthesized as an inactive precursor and requires cleavage by the enzyme caspase-1 to become biologically active. The mature form of IL-18 binds to its receptor, IL-18R, which consists of two subunits: IL-18Rα and IL-18Rβ. This binding triggers a signaling cascade that leads to the production of IFN-γ and other proinflammatory cytokines .
IL-18 plays a significant role in the host defense against infections, particularly viral and intracellular bacterial infections. It enhances the cytotoxic activity of natural killer (NK) cells and T cells, promoting the production of IFN-γ, which is essential for the activation of macrophages and the clearance of pathogens .
Mouse anti-human IL-18 antibodies are monoclonal or polyclonal antibodies developed in mice to specifically target and bind to human IL-18. These antibodies are widely used in research and clinical applications to study the role of IL-18 in various diseases and to develop potential therapeutic strategies.
Mouse anti-human IL-18 antibodies are utilized in several scientific applications, including:
Research on IL-18 and its antibodies has provided valuable insights into the pathogenesis of various inflammatory and autoimmune diseases, such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis. Additionally, IL-18 has been implicated in the progression of certain cancers and viral infections .
The therapeutic potential of targeting IL-18 has been explored in several studies. Neutralizing IL-18 activity using specific antibodies or IL-18 binding proteins (IL-18BP) has shown promise in reducing inflammation and disease severity in preclinical models . These findings suggest that IL-18 could be a potential therapeutic target for treating inflammatory and autoimmune diseases .
In conclusion, IL-18 is a critical cytokine involved in immune regulation and inflammation. Mouse anti-human IL-18 antibodies are valuable tools in research and have potential therapeutic applications in treating various diseases.