IL12B (Interleukin-12 subunit beta) is a 40 kDa protein that forms the heterodimeric cytokine interleukin-12 (IL-12) when paired with IL12A (p35 subunit) . This cytokine is critical for activating T cells and natural killer (NK) cells, promoting Th1 responses, and inducing interferon-gamma (IFN-γ) production . The IL12B recombinant monoclonal antibody is a genetically engineered antibody designed to bind specifically to the IL12B protein, enabling precise detection, neutralization, or modulation of its activity in research and therapeutic contexts .
Recombinant monoclonal antibodies are produced via in vitro cloning of heavy and light chain genes into expression vectors, followed by transfection into host cells (e.g., CHO, HEK) . Key features include:
PAT1D6AT (mouse IgG1): Targets human IL12B (aa 23–328), validated for WB and IHC .
OTI1A3 (mouse IgG): Recognizes human IL12B, suitable for WB and IHC .
C2 (mouse IgG2b): Cross-reactive with rat IL12B, used in WB, IHC, and IP .
Autoimmune Diseases: Overexpression of IL12B in multiple sclerosis (MS) and inflammatory bowel disease (IBD) highlights its role in Th1-driven inflammation . Antibodies like ustekinumab (anti-p40) neutralize IL-12/IL-23, reducing disease severity in IBD .
Cancer Immunology: IL12B antibodies block IL-12-mediated antitumor immunity in preclinical models, though targeted delivery (e.g., NHS-IL12) enhances efficacy while minimizing toxicity .
Clone | Host | Applications | Reactivity | Validated Species |
---|---|---|---|---|
PAT1D6AT | Mouse | WB, IHC | Human | - |
OTI1A3 | Mouse | WB, IHC | Human | - |
C2 | Mouse | WB, IHC, IP | Human, Rat | |
21 H19L14 | Rabbit | WB, FC | Human, predicted for NHP, mouse, rat |
Systemic IL-12 administration causes severe adverse effects (e.g., fever, thrombocytopenia) . Strategies to reduce toxicity include:
Targeted Delivery: Using nanoparticles or stromal cells to localize IL-12 to tumors .
Dose Optimization: NHS-IL12 achieves therapeutic efficacy at lower doses compared to recombinant IL-12 .
Preclinical studies suggest synergies with:
The IL12B recombinant monoclonal antibody is produced through a rigorous in vitro cloning process. The genes encoding both the heavy and light chains of the IL12B antibody are precisely integrated into expression vectors. These vectors are then introduced into host cells, enabling the recombinant antibody's expression within a controlled cell culture environment. Following expression, the antibody undergoes affinity chromatography purification. This antibody is recommended for the detection of human IL12B protein in ELISA and FC applications.
IL12B protein, also known as the p40 subunit, forms the biologically active IL-12 cytokine in conjunction with the p35 subunit. IL-12 plays a crucial role in immune responses, including the activation of immune cells, the differentiation of T cell subsets, and the regulation of immune functions essential for host defense against infections and cancer.
IL12B (p40) is the 40 kDa subunit of interleukin-12 that performs multiple crucial immunological functions. IL12B can form a heterodimer with either the p35 subunit (creating IL-12) or with the p19 subunit (creating IL-23) . As part of IL-12, it induces production of IFN-gamma and TNF by resting and activated T and NK cells, enhances cytotoxic activity of NK and T cells, and acts as a co-mitogen to stimulate T cell proliferation . The p40 subunit also exists as a monomer and homodimer, with the latter functioning as a potent IL-12 antagonist .
IL-12 represents a pivotal mediator of cell-mediated immunity through its actions on TH1 cell development, proliferation, and activity. It is primarily produced by macrophages, dendritic cells, activated phagocytes, and to a lesser extent, B cells in response to infectious agents .
IL12B recombinant monoclonal antibodies are produced through sophisticated in vitro cloning techniques. The general methodology involves:
Gene cloning and vector construction: The genes encoding both heavy and light chains of the IL12B antibody are isolated and integrated into expression vectors .
Host cell transfection: The vectors are introduced into appropriate host cells (commonly mammalian cell lines or insect cells like Spodoptera frugiperda) .
Cell culture expression: The transfected cells express the recombinant antibody in a controlled cell culture environment .
Purification: Following expression, the antibody undergoes affinity chromatography purification to isolate the target protein .
Quality control: The purified antibody is characterized through techniques such as SDS-PAGE and Size Exclusion Chromatography to verify purity and structural integrity .
This process allows for consistent production of high-quality monoclonal antibodies with specific binding characteristics and functional properties.
IL12B antibodies typically consist of standard immunoglobulin structures with two heavy chains and two light chains, forming antigen-binding Fab regions and an Fc region that can recruit immune effector functions . The specific binding regions are engineered to recognize distinct epitopes on the IL12B protein.
For example, the monoclonal antibody described in catalog number ABIN948031 targets amino acids 229-328 of human IL12B , while other antibodies may target different regions such as Val30~Ser320 . The choice of binding epitope significantly influences the antibody's functional properties, including:
Neutralization capability: Antibodies binding to crucial functional domains may block interactions with receptors or with the p35 subunit
Cross-reactivity: Some antibodies like the one described in search result cross-react with murine IL-12
Application suitability: Different epitope binding affects performance in specific applications like ELISA, flow cytometry, or Western blotting
The clonality, isotype (commonly IgG1, IgG2a), and species origin (mouse, rabbit, etc.) of the antibody further define its functional properties and experimental utility .
IL12B recombinant monoclonal antibodies have been validated for multiple research applications with specific optimization parameters:
For optimal detection of IL-12 p70 heterodimer by ELISA, researchers should consider combining complementary antibodies, such as using purified B-T21 antibody (targeting p35) for capture and biotinylated C8.6 antibody for detection .
For optimal flow cytometry results with IL12B antibodies, comprehensive optimization of experimental conditions is essential:
Cell stimulation protocols:
Fixation and permeabilization:
Antibody selection and staining procedure:
Primary antibody: Anti-IL12B at optimized concentration (typically 1:50-1:200 dilution)
Secondary antibody: Fluorophore-conjugated detection antibody (e.g., Allophycocyanin-conjugated Anti-species IgG)
Consider co-staining with cell surface markers (e.g., CD14 for monocytes) using PE-conjugated antibodies
Controls:
Include unstimulated cells as negative controls
Use isotype controls to confirm specificity
Consider fluorescence minus one (FMO) controls for multicolor panels
A properly optimized flow cytometry protocol allows for sensitive detection of IL12B in various cell populations while minimizing background and non-specific signals.
Validating IL12B antibody specificity requires a multi-faceted approach:
Subunit specificity determination:
Cross-species reactivity assessment:
Functional validation:
Application-specific controls:
Use knockout/knockdown systems as negative controls
Compare multiple antibodies targeting different epitopes
Include recombinant IL12B as positive control
These validation steps ensure experimental reliability and help researchers select the appropriate antibody for their specific research questions.
IL12B antibodies have been instrumental in developing novel immunotherapeutic approaches that leverage IL-12's potent antitumor properties while addressing toxicity concerns:
Antibody-cytokine fusion proteins:
Researchers have developed innovative IL12-antibody fusion constructs that selectively target the tumor microenvironment:
The IL12-F8-F8 fusion protein combines IL-12 with antibodies specific to the alternatively spliced EDA domain of fibronectin (a tumor neovasculature marker)
This construct demonstrated potent tumor growth inhibition in three different immunocompetent syngeneic models of cancer
Quantitative biodistribution analysis confirmed selective localization to tumor neovasculature in vivo
The approach allows production of both murine IL-12 (mIL12) and human IL-12 (hIL12) versions
FAP-targeted IL-12 delivery:
The mIL12-7NP2 fusion protein combines IL-12 with antibodies targeting human fibroblast activation protein (FAP)
Selective accumulation was observed in FAP-positive tumors but not in FAP-negative tumors or healthy organs
Immunofluorescence-based biodistribution analysis confirmed preferential accumulation in FAP-positive neoplastic lesions 24 hours after intravenous administration
These approaches demonstrate how IL12B antibodies can be engineered to overcome systemic toxicity limitations while preserving antitumor efficacy, providing crucial insights into tumor-immune interactions.
IL-12 pharmacokinetic desensitization represents a significant challenge in therapeutic applications, characterized by reduced IL-12 concentrations and biological effects following repeated exposure. Recent research has identified two potential mechanisms:
Increased clearance model:
Previously proposed to occur due to upregulation of IL-12 receptor on T-cells causing increased receptor-mediated clearance
Modeling studies demonstrated this mechanism failed to capture trends in clinical trial data
The model predicts changes in maximal time and half-life that weren't observed clinically
Reduced bioavailability model:
More accurately predicts IL-12 pharmacokinetics across clinical trials
The model suggests issues with IL-12 bioavailability and transport into blood rather than accelerated clearance
Mathematical modeling demonstrated this mechanism could fit clinical datasets accurately across a wide range of parameter values
Antibody-based approaches address these challenges through:
Targeted delivery: Fusion proteins like IL12-F8-F8 and mIL12-7NP2 reduce systemic exposure while maintaining local efficacy
Altered pharmacokinetics: Antibody components can extend half-life and modify distribution
Controlled release: Antibody-mediated binding to tissue components can create local reservoirs for sustained activity
These strategies represent promising avenues to overcome the pharmacokinetic limitations that have historically hindered IL-12's therapeutic potential.
IL12B antibodies significantly augment combination immunotherapy strategies through several mechanisms:
Rituximab combination therapy:
A phase I clinical study combining IL-12 with rituximab (anti-CD20 antibody) in B-cell lymphoma demonstrated:
Objective responses in 29 of 43 patients (69%)
8 of 11 complete responses at IL-12 doses ≥300 ng/kg
Optimal immunologic dose: 300 ng/kg subcutaneously twice weekly
20-fold increase in serum IFN-γ levels and 2.5-5 fold increase in IP-10 levels at IL-12 doses ≥100 ng/kg
The mechanism involves IL-12 enhancing rituximab efficacy by:
Paclitaxel combination:
Immune checkpoint inhibitor synergy:
IL-12's ability to stimulate Th1 responses can complement checkpoint inhibitor activity
Local IL-12 delivery may overcome resistance to checkpoint blockade
The success of these approaches highlights the potential of IL12B antibodies to enhance existing immunotherapies by promoting more robust and directed immune responses against malignancies.
Detecting low-abundance IL12B in complex samples requires specialized methodological approaches:
Enhanced stimulation protocols for cellular IL12B expression:
Advanced immunoassay techniques:
Signal amplification methods:
Tyramide signal amplification for immunohistochemistry
Polymer-based detection systems
Biotin-streptavidin amplification
Sample preparation optimization:
For cell lysates: Use appropriate detergents (RIPA buffer with protease inhibitors)
For tissue samples: Consider antigen retrieval methods
For serum/plasma: Pre-clearing with protein A/G can reduce background
Technical considerations for Western blotting:
Extended transfer times for optimal protein migration
Blocking with 5% BSA rather than milk to reduce background
Overnight primary antibody incubation at 4°C
These approaches significantly enhance detection sensitivity and reliability when analyzing IL12B in biological specimens where the target may be present at low concentrations.
Differentiating between free IL12B and its heterodimeric cytokine forms requires specific analytical approaches:
Antibody selection strategy:
Sequential immunoprecipitation approach:
First immunoprecipitation with anti-p35 antibodies captures IL-12 heterodimer
Second immunoprecipitation with anti-p19 antibodies captures IL-23 heterodimer
Final immunoprecipitation with anti-p40 antibodies captures remaining free IL12B
Size-based differential analysis:
Native PAGE or size exclusion chromatography to separate proteins based on size
Western blotting with anti-IL12B antibodies will detect different molecular weight bands:
~40 kDa: Free IL12B monomer
~70-80 kDa: IL-12 or IL-23 heterodimers
~80 kDa: IL12B homodimer
Functional bioassays:
IL-12-specific bioassays using cells expressing only IL-12 receptors (IL-12Rβ1/IL-12Rβ2)
IL-23-specific bioassays using cells expressing only IL-23 receptors (IL-12Rβ1/IL-23R)
Comparing results with these selective bioassays can distinguish which heterodimer is present
By employing these techniques, researchers can accurately quantify the distribution of IL12B between its free and heterodimeric forms, providing crucial insights into the biological significance of each form in various experimental systems.
Analyzing IL12B-mediated signaling requires tailored approaches for different cell populations:
Cell type-specific receptor expression profiling:
Phospho-protein analysis optimization:
For JAK-STAT pathway activation:
Optimal stimulation timepoints: pJAK2 (5-15 min), pSTAT4 (15-30 min)
Quick sample processing is essential (immediate fixation)
Use phosphatase inhibitors in all buffers
Recommended antibody panels:
For T cells: anti-pSTAT4 + anti-CD3/CD4/CD8
For NK cells: anti-pSTAT4 + anti-CD56/CD16
For monocytes: anti-pSTAT4 + anti-CD14
Downstream effect measurement:
IFN-γ production assays:
ELISA measurement at 24-48 hours post-stimulation
Intracellular cytokine staining at 6-12 hours (with protein transport inhibitors)
Cytotoxicity assays:
51Cr release assays for NK and T cell cytotoxicity
Flow cytometry-based killing assays
Pathway inhibition strategies:
JAK inhibitors (e.g., tofacitinib) to block STAT activation
Anti-IL12B neutralizing antibodies to block initial signaling
siRNA knockdown of receptor components
Cell-specific considerations:
T cells: Pre-activation with anti-CD3/CD28 enhances IL-12 responsiveness
NK cells: IL-15 priming increases receptor expression
Monocytes: LPS pre-treatment induces IL-12 production and alters signaling dynamics
These methodological considerations enable detailed characterization of IL12B-mediated signaling in different immune cell populations, providing insights into cytokine-specific effects on immune function.
Cross-reactivity issues present significant challenges in comparative immunology research. Researchers can implement several strategies to address these limitations:
Comprehensive cross-reactivity testing:
Species-specific antibody development approaches:
Target conserved epitopes when cross-reactivity is desired
Select species-unique regions when specificity is required
Consider developing species-specific detection panels
Alternative detection strategies when cross-reactivity is limited:
Species-specific PCR for mRNA detection
Mass spectrometry-based protein identification
Use of species-matched detection systems
Data interpretation with cross-reactivity limitations:
Adjust sensitivity expectations based on known affinity differences
Include appropriate controls for each species
Consider parallel validation with multiple antibody clones
Technical optimization for cross-reactive antibodies:
Adjust antibody concentrations based on species-specific affinity testing
Modify incubation conditions (time, temperature, buffer composition)
Evaluate different detection systems for optimal signal-to-noise ratios
When selecting antibodies for multi-species studies, researchers should prioritize clones with demonstrated cross-reactivity or develop species-specific detection strategies when necessary.
Proper storage and handling are critical for maintaining IL12B antibody functionality:
Storage Condition | Recommended Practices | Impact on Functionality |
---|---|---|
Temperature | Store at -20°C for long-term stability | Prevents protein denaturation and maintains binding capacity |
Buffer Composition | PBS with 0.02% sodium azide and 50% glycerol (pH 7.3) | Stabilizes antibody structure and prevents microbial growth |
Aliquoting | Unnecessary for -20°C storage with glycerol, but advisable for frequent use | Minimizes freeze-thaw cycles that can degrade antibody |
Protein Stabilizers | Some preparations contain 0.1% BSA | Prevents adsorption to surfaces and enhances stability |
Freeze-Thaw Cycles | Minimize; each cycle can reduce activity by 5-10% | Prevents formation of protein aggregates that reduce functionality |
Working Solution | Dilute in buffer with carrier protein (0.1-1% BSA) | Prevents non-specific binding and adsorption to plastics |
Short-term Storage | 2-8°C for up to one week when diluted | Allows convenient access while maintaining stability |
Recommendations from manufacturers indicate that properly stored antibodies remain stable for one year after shipment . Researchers should monitor antibody performance over time, particularly for critical applications like neutralization assays or therapeutic studies.
Inconsistent results with IL12B antibodies across experimental systems can be systematically addressed:
Sample preparation variability:
Standardize lysis buffers and protein extraction protocols
Control for post-translational modifications that might affect epitope recognition
Consider native vs. denatured conditions affecting epitope accessibility
Antibody validation across applications:
Recognize that antibodies optimized for one application may not perform equally in others
Validate each application independently (WB, ELISA, IHC, flow cytometry)
Example: The antibody described in ABIN948031 is validated for Western Blotting and ELISA but may not perform optimally in other applications
Technical optimization strategies:
Control integration:
Include positive controls (recombinant IL12B protein)
Utilize negative controls (IL12B knockout/knockdown samples)
Implement isotype controls for immunostaining applications
Data interpretation frameworks:
Triangulate results using multiple antibody clones targeting different epitopes
Compare results across complementary detection methods
Consider biological variability in IL12B expression across different experimental systems
By implementing these systematic approaches, researchers can identify the source of inconsistencies and develop standardized protocols that yield reproducible results across experimental systems.
Cutting-edge research is expanding the capabilities of IL12B antibodies through innovative engineering approaches:
Advanced tumor-targeting immunocytokine formats:
The IL12-F8-F8 format fuses IL-12 with two F8 antibodies in single-chain Fv (scFv) format targeting the EDA domain of fibronectin
The mIL12-7NP2 format targets fibroblast activation protein (FAP) in the tumor microenvironment
These designs demonstrate superior tumor selectivity compared to conventional IL-12 administration
Novel antibody engineering strategies:
Tandem diabody formats enhance tumor accumulation and retention
Sequential fusion of IL-12 as a single polypeptide with targeting antibodies
Integration of cleavable linkers for conditional activation in tumor microenvironments
Cell therapy integration approaches:
Combination therapy optimization:
These innovations represent promising avenues for enhancing the research and therapeutic utility of IL12B antibodies, especially in challenging areas like cancer immunotherapy.
Researchers are developing innovative antibody-based approaches to study the complex biology of IL12B heterodimeric interactions:
Bispecific antibody technologies:
Dual-targeting antibodies simultaneously binding p40 and p35 (for IL-12) or p40 and p19 (for IL-23)
These constructs allow specific isolation and functional analysis of intact heterodimers
Enable comparative studies between heterodimers sharing the common p40 subunit
Conformation-specific antibody development:
Antibodies recognizing unique epitopes formed only in the assembled heterodimer
These reagents distinguish between monomeric p40 and heterodimeric forms
Enable precise quantification of assembled cytokines vs. free subunits
Proximity-based detection systems:
Pairs of antibodies targeting different subunits coupled with proximity ligation assay (PLA) technology
FRET-based antibody pairs for real-time monitoring of heterodimer assembly
These approaches provide spatial and temporal information about heterodimer formation
Structural biology integration:
Antibodies that stabilize specific conformations for crystallography studies
Fragment antibodies facilitating cryo-EM analysis of heterodimeric complexes
These tools advance our understanding of the structural basis for IL12B interactions
These approaches are transforming our ability to study the complex biology of IL12B-containing heterodimers and their distinct roles in immune regulation and disease pathogenesis.
Emerging research is exploring the role of post-translational modifications (PTMs) in IL12B biology and the development of modification-specific antibodies:
Key IL12B post-translational modifications under investigation:
Glycosylation patterns affecting heterodimer assembly and stability
Phosphorylation events potentially regulating secretion and bioactivity
Proteolytic processing modifying biological functions
Development challenges for PTM-specific antibodies:
Generating antibodies with exquisite specificity for modified epitopes
Validating specificity across different experimental systems
Ensuring recognition of physiologically relevant modifications
Methodological innovations enabling PTM-specific detection:
Synthetic peptide antigens incorporating defined modifications
Recombinant expression systems with controlled modification patterns
Mass spectrometry validation of modification-specific antibody binding
Research applications:
Studying the kinetics of IL12B modification in different activation states
Analyzing tissue-specific modification patterns
Investigating the role of modifications in disease contexts
Therapeutic implications:
Targeting specifically modified forms of IL12B for enhanced selectivity
Developing antibodies that modulate modification-dependent protein-protein interactions
Engineering therapeutic antibodies that recognize disease-associated modification patterns
These emerging approaches promise to reveal new layers of IL12B regulation through post-translational modifications and provide novel tools for both basic research and therapeutic development.