CD4 Monoclonal Antibody,Purified

Shipped with Ice Packs
In Stock

Description

Structure and Specificity

CD4 mAbs are IgG-class immunoglobulins produced via hybridoma technology. Key characteristics include:

  • Molecular weight: ~55–59 kDa (antigen-specific)

  • Clones: Diverse clones (e.g., RPA-T4, GK1.5, MAX.16H5) recognize distinct epitopes on CD4’s extracellular domain .

  • Species reactivity: Primarily human and murine models, with some cross-reactivity in transgenic systems .

Table 1: Common CD4 mAb Clones and Properties

CloneIsotypeEpitope SpecificityKey Applications
RPA-T4IgG1Domain 1 of CD4Blocks HIV gp120 binding
GK1.5IgG2bMurine CD4T-cell depletion studies
MAX.16H5IgG1gp120-like epitopeAutoimmune disease therapy
KeliximabIgG1Human/chimpanzee CD4Asthma clinical trials

Purification and Quality Control

Purification protocols ensure high specificity and functionality:

  • Methods: Protein A/G affinity chromatography, ammonium sulfate precipitation, and size-exclusion chromatography .

  • Purity: >90% by SDS-PAGE, <10% aggregation via HPLC .

  • Endotoxin levels: <1.0 EU/mg .

Critical quality assessments include:

  • Binding kinetics: Affinity constants (K<sub>A</sub>) range from 10<sup>8</sup> to 10<sup>9</sup> M<sup>-1</sup> .

  • Stability: Stored at 2–8°C in buffered aqueous solutions .

Mechanisms of Action

CD4 mAbs modulate immune responses through:

  • Receptor blockade: Inhibit MHC class II interactions and T-cell activation .

  • Fc receptor-dependent effects:

    • Depletion: IgG1 variants (e.g., keliximab) induce CD4<sup>+</sup> T-cell loss via antibody-dependent cellular cytotoxicity (ADCC) .

    • Non-depleting: Engineered IgG4 variants (e.g., clenoliximab) minimize FcγR binding, reducing cytotoxicity while retaining immunomodulation .

Immunological Studies

  • Flow cytometry: Quantify CD4<sup>+</sup> T cells using 10 µL antibody per 1×10<sup>6</sup> cells .

  • T-cell functional assays: Assess cytotoxicity, cytokine production (e.g., TNF-α, IFN-γ), and proliferation .

Therapeutic Use

  • Autoimmune diseases: MAX.16H5 showed efficacy in rheumatoid arthritis and graft-versus-host disease (GVHD) by transiently depleting CD4<sup>+</sup> cells .

  • Asthma: Keliximab reduced allergen-induced T-cell proliferation in severe asthmatics .

  • HIV research: RPA-T4 blocks viral entry by competing with gp120 for CD4 binding .

Key Research Findings

  • Fc engineering: Clenoliximab’s modified IgG4 reduced ADCC by >10-fold compared to IgG1 counterparts .

  • Cytokine modulation: Anti-CD4 preincubation with RIB5/2 increased TNF-α secretion in rat adjuvant arthritis models .

  • Clinical outcomes: Keliximab trials demonstrated reduced CD4<sup>+</sup> counts (30–50%) and CD25/HLA-DR expression in asthma patients .

Product Specs

Form
Phosphate-buffered solution, pH 7.4, containing 0.09% sodium azide.
Lead Time
We typically dispatch orders for CD4 Monoclonal Antibody, Purified within 1-3 business days of receipt. Delivery timelines may vary depending on the order fulfillment method and destination. For specific delivery estimates, please contact your local distributor.

Q&A

What is CD4 and why is it an important target for monoclonal antibodies?

CD4 is a 55 kDa cell surface glycoprotein receptor that plays a crucial role in the immune system. It functions as a co-receptor for the T-cell receptor (TCR) complex and interacts with major histocompatibility complex (MHC) class II molecules on antigen-presenting cells. This interaction is fundamental for T cell development and optimal functioning of mature T cells. CD4 is primarily expressed on a subset of T lymphocytes (T helper cells), but is also found on monocytes, macrophages, and dendritic cells. At the tissue level, CD4 can be detected in thymus, lymph nodes, tonsils, spleen, and specific regions of the brain, gut, and other non-lymphoid tissues .

CD4 is also known to associate with protein tyrosine kinase p56lck through its cytoplasmic tail, which initiates or augments the early phase of T-cell activation. Additionally, CD4 serves as a co-receptor for HIV, making it a significant target for research in infectious diseases. The widespread distribution and critical immunological functions of CD4 make it an important target for monoclonal antibodies used in research, diagnostics, and potential therapeutic applications .

How do different clones of purified anti-CD4 monoclonal antibodies differ in their binding properties?

Different clones of purified anti-CD4 monoclonal antibodies recognize distinct epitopes on the CD4 molecule, resulting in varied binding properties and applications. For example, the RM4-5 clone reacts with mouse CD4 and has binding that can be blocked by another clone, GK1.5 . The OKT-4 clone specifically targets human CD4 , while RM4-4 binds to the CD4 (L3T4) differentiation antigen expressed on most thymocytes, subpopulations of mature T lymphocytes, and a subset of NK-T cells in various mouse strains .

These differences in epitope recognition impact several functional aspects of the antibodies. Some clones may preferentially bind to specific conformational states of CD4 or different domains of the protein. This can affect the antibody's ability to block or induce functional changes in CD4-expressing cells. For instance, an antibody that binds to the MHC class II interaction site might block T cell activation, while one binding to a different domain might not interfere with this function. Additionally, cross-reactivity with CD4 from different species varies between clones - some are highly species-specific, while others may recognize conserved epitopes across multiple species .

The binding affinity, avidity, and kinetics also differ between clones, affecting their suitability for particular applications. For optimal experimental results, researchers should carefully select clones based on the specific requirements of their research question and experimental system.

What are the standard purification techniques for CD4 monoclonal antibodies, and how do they affect antibody quality?

Several purification techniques are commonly employed for CD4 monoclonal antibodies, each with distinct impacts on the final product quality. A standard multi-step purification process typically begins with ammonium sulfate ((NH₄)₂SO₄) precipitation to initially separate antibodies from culture medium components. This is followed by chromatographic methods, including anion-exchange chromatography on columns such as MonoQ or Q-Sepharose, which separates proteins based on charge differences. Hydrophobic interaction chromatography, often using phenyl-Sepharose, further purifies antibodies based on hydrophobicity. Finally, gel filtration chromatography on columns like Superdex 200 achieves high-resolution separation based on molecular size .

Advanced manufacturing processes for high-grade antibodies may incorporate protein A or G affinity chromatography, which specifically binds the Fc region of antibodies with high selectivity. This approach is particularly valuable for functional grade preclinical antibodies manufactured in animal-free facilities using in vitro cell culture techniques .

Each purification step influences antibody quality in different ways. Precipitation methods may risk partial denaturation if conditions are not carefully controlled. Ion exchange chromatography can affect charge distribution on the antibody surface, potentially altering binding characteristics. Hydrophobic interaction chromatography might modify conformational aspects of the antibody structure. The final gel filtration step is crucial for removing aggregates, which can significantly impact functional properties and immunogenicity.

How can researchers assess the purity and quality of purified anti-CD4 monoclonal antibodies?

Researchers can implement multiple complementary analytical methods to comprehensively assess the purity and quality of purified anti-CD4 monoclonal antibodies. For protein purity assessment, analytical size exclusion chromatography (SEC) provides quantitative determination of monomer content, with high-quality antibodies typically showing ≥95% monomer by analytical SEC. SDS-PAGE analysis under both reducing and non-reducing conditions offers visual confirmation of purity (>95% standard) and can detect contaminating proteins or antibody fragmentation .

Functional integrity can be evaluated through binding assays such as ELISA, flow cytometry, or surface plasmon resonance to confirm target specificity and binding affinity. For antibodies intended for in vivo applications, endotoxin testing using the LAL method is essential, with acceptable levels typically below 1.0 EU/mg . Host cell protein and DNA contamination analyses are particularly important for antibodies produced in mammalian expression systems, as demonstrated in the analytical characterization of the MAX.16H5 antibody, which confirmed freedom from mouse DNA and other contaminants .

Physicochemical characterization should include assessment of aggregate formation through dynamic light scattering or analytical ultracentrifugation, charge heterogeneity through isoelectric focusing or ion-exchange chromatography, and glycosylation patterns through mass spectrometry or lectin binding assays. These parameters can significantly influence antibody stability, half-life, and effector functions.

For therapeutic-grade antibodies, additional testing for bioburden, sterility, viral contamination, pyrogenicity, and abnormal toxicity may be required, as was performed for the MAX.16H5 antibody preparation intended for human therapy . Stability studies under various storage conditions provide critical information on shelf-life and optimal storage parameters. Implementing this multi-faceted analytical approach ensures that researchers can confidently select antibody preparations that meet their specific experimental requirements.

What are the primary research applications for purified CD4 monoclonal antibodies?

Purified CD4 monoclonal antibodies serve numerous critical research applications across immunology, cell biology, and translational medicine. Flow cytometry represents one of the most common applications, where these antibodies enable precise identification, enumeration, and isolation of CD4-expressing cell populations from complex samples such as blood, lymphoid tissues, or in vitro cultures. Multiple clones, including RM4-5 for mouse samples and OKT-4 for human specimens, have been extensively validated for this purpose .

Immunohistochemistry and immunocytochemistry applications allow researchers to visualize CD4 expression patterns in tissue sections or cultured cells, respectively. Different formulations may be optimized for specific fixation methods, with some clones like H129.19 or RM4-5 specifically recommended for immunohistochemical staining of mouse tissues . When selecting antibodies for these applications, researchers should consider the compatibility with fixation protocols and tissue preparation methods.

In functional studies, purified CD4 antibodies can be employed to modulate T cell activation, either by blocking CD4-MHC class II interactions or by inducing signaling through CD4 itself. The functional characteristics can vary significantly between clones, with some serving as activating antibodies while others function primarily as blocking reagents. The keliximab (IDEC CE9.1) study exemplifies how anti-CD4 antibodies can be used therapeutically to modulate CD4+ T-cell populations in conditions like corticosteroid-dependent asthma .

Additional applications include western blotting for protein expression analysis, immunoprecipitation for studying protein-protein interactions, and in vivo administration for manipulating immune responses in animal models. The OKT-4 clone, for instance, has been formulated as an in vivo GOLD functional grade preparation specifically designed for such experimental therapeutic applications . When selecting an antibody for any application, researchers must consider the specific clone, species reactivity, isotype, and formulation properties to ensure optimal performance for their particular experimental system.

How should researchers optimize CD4 antibody concentrations for different experimental applications?

Optimizing CD4 antibody concentrations requires systematic titration approaches tailored to specific applications. For flow cytometry, the standard starting point is typically ≤0.125 μg per test (where a test represents 10^5 to 10^8 cells in a 100 μL volume), but researchers should perform titration experiments to determine the optimal signal-to-noise ratio for their specific cell system . This involves testing a range of antibody concentrations (usually 2-fold serial dilutions) against a fixed number of cells, then analyzing the staining index - the ratio of specific signal to background fluorescence.

For immunohistochemistry and immunocytochemistry, optimal concentrations vary considerably depending on tissue type, fixation method, and detection system. Initial titrations typically range from 1-10 μg/mL, with specific recommendations varying by clone. For instance, purified H129.19 or RM4-5 mAbs are specifically recommended for immunohistochemical staining of mouse tissues, suggesting that not all anti-CD4 clones perform equally in this application .

In functional assays where CD4 antibodies are used to stimulate or block T cell responses, concentration requirements can differ substantially from detection applications. Blocking experiments typically require higher concentrations (5-20 μg/mL) to ensure complete saturation of CD4 molecules. For in vivo applications, dose optimization should consider pharmacokinetic parameters - the keliximab study demonstrated that different dosing cohorts (0.5, 1.5, and 3.0 mg/kg) resulted in distinctly different pharmacokinetic profiles and clearance rates .

Critically, each new experimental system requires independent optimization, as antibody performance can vary with cell type, species, sample preparation, and detection methods. As noted in BD Biosciences' technical guidance, "Since applications vary, each investigator should titrate the reagent to obtain optimal results" . Documentation of optimization experiments creates valuable reference points for subsequent studies and contributes to experimental reproducibility.

What are the optimal storage conditions for maintaining long-term activity of purified CD4 antibodies?

For extended preservation of antibody activity, aseptically aliquoting in working volumes without dilution and storing at ≤ -70°C is recommended . This ultra-low temperature minimizes molecular motion and enzymatic processes that contribute to degradation. Creating multiple small aliquots rather than storing the entire antibody preparation in a single container is crucial, as it prevents repeated freeze-thaw cycles that significantly damage antibody structure and function. Each freeze-thaw event can lead to partial denaturation, aggregation, and precipitation of antibodies, progressively reducing binding capacity and specificity.

The formulation buffer significantly impacts storage stability. Most purified monoclonal antibodies are aseptically packaged in phosphate-buffered saline (typically 0.01 M PBS with 150 mM NaCl, pH 7.2-7.4) without carrier proteins or preservatives . While this minimizes potential interference in downstream applications, it may reduce stability compared to formulations containing stabilizers. Researchers should note that certain antibody preparations may be prone to precipitation over time due to inherent biochemical properties. If precipitation occurs, it can often be removed by aseptic centrifugation and/or filtration without significantly affecting antibody functionality .

For antibodies containing sodium azide as a preservative, special considerations apply. Sodium azide is a reversible inhibitor of oxidative metabolism and should not be used in cell cultures or injected into animals . For functional studies requiring azide-free preparations, researchers should consider using NA/LE (No Azide/Low Endotoxin) antibody formats or removing sodium azide by washing bound antibodies or dialyzing soluble antibodies into azide-free buffer .

How does the presence of preservatives affect CD4 antibody functionality in different applications?

The presence of preservatives in CD4 monoclonal antibody preparations can significantly impact functionality across different applications, with effects varying by preservative type and experimental context. Sodium azide, a commonly used antibody preservative, acts as a reversible inhibitor of oxidative metabolism by blocking cytochrome oxidase in the electron transport chain. This property makes azide-containing antibody preparations unsuitable for cell cultures or in vivo injections, as they can inhibit cellular metabolic processes critical for experimental outcomes .

For functional studies involving living cells, sodium azide can suppress T cell activation, proliferation, and cytokine production, potentially leading to false negative results when assessing CD4-mediated immune responses. In vitro functional assays particularly affected include T cell proliferation assays, cytotoxicity tests, and cellular activation studies. BD Biosciences specifically recommends NA/LE (No Azide/Low Endotoxin) antibody formats for such applications .

High-grade functional antibody preparations, such as in vivo GOLD formulations, are specifically manufactured without preservatives and are purified to ensure extremely low levels of endotoxins and leachable protein A . These preparations undergo aseptic processing and are formulated in PBS without carrier proteins, potassium, calcium, or preservatives to minimize interference in sensitive applications while maintaining stability. This absence of preservatives necessitates more stringent handling protocols to prevent microbial contamination, including aseptic technique when aliquoting and the potential for shorter shelf-life at refrigeration temperatures compared to preserved formulations.

What are common technical challenges when using CD4 antibodies and how can they be addressed?

Researchers frequently encounter several technical challenges when working with CD4 monoclonal antibodies. One common issue is inconsistent staining intensity in flow cytometry or immunohistochemistry applications. This variability can stem from epitope masking due to protein-protein interactions or conformational changes in the CD4 molecule. To address this, researchers should evaluate different antibody clones that recognize distinct CD4 epitopes. For example, the binding of RM4-5 clone is blocked by GK1.5, suggesting they recognize overlapping epitopes, whereas other clones may retain binding under conditions where these epitopes are masked .

Another significant challenge is high background signal, particularly in tissue sections or cell preparations with endogenous Fc receptor expression. This non-specific binding can be mitigated by including an Fc receptor blocking step using appropriate isotype controls. When using mouse-derived anti-CD4 antibodies like OKT-4, researchers should include mouse IgG2b isotype controls to account for non-specific binding .

Antibody internalization presents challenges in experiments requiring sustained CD4 labeling. CD4 receptors undergo dynamic internalization and recycling, particularly following activation. Extended incubation periods may result in decreased surface staining due to internalization rather than true reduction in CD4 expression. Time-course experiments and comparison with mRNA expression can help distinguish between these phenomena.

Sodium azide in antibody preparations can interfere with functional studies by inhibiting cellular metabolism. For functional assays examining T cell activation or proliferation, researchers should use azide-free formulations or remove azide through dialysis or washing procedures . Additionally, endotoxin contamination can trigger non-specific immune cell activation; for in vivo or sensitive in vitro applications, specially formulated low-endotoxin preparations (<1.0 EU/mg) should be employed .

For long-term storage, antibody aggregation and precipitation are common problems. These can be addressed by aseptically centrifuging preparations before use, avoiding repeated freeze-thaw cycles, and properly aliquoting antibodies for storage at recommended temperatures (≤-70°C for long-term storage) . When precipitation occurs despite proper handling, gentle filtration or centrifugation can often restore antibody functionality without significant loss of binding capacity.

How should researchers design validation experiments when using CD4 antibodies in new experimental systems?

Designing robust validation experiments for CD4 antibodies in new experimental systems requires a systematic, multi-parameter approach. Initially, researchers should perform antibody titration across a concentration gradient (typically 2-fold serial dilutions) to determine optimal signal-to-noise ratios for the specific application and cellular system. As noted in technical documentation, "each investigator should titrate the reagent to obtain optimal results" since applications vary significantly . This titration should be performed using positive control samples with known CD4 expression (e.g., isolated T helper cells) and negative control samples (e.g., CD8+ T cells or non-lymphoid cells).

Specificity validation is paramount and should include multiple complementary approaches. Isotype control antibodies matching the CD4 antibody class (e.g., mouse IgG2b κ for OKT-4 clone) should be tested in parallel to distinguish specific binding from background . Competitive binding assays using unlabeled CD4 antibodies or recombinant CD4 proteins can confirm epitope-specific interactions. For novel applications, validation should include comparative analysis of multiple anti-CD4 clones, as different clones may perform optimally in different experimental contexts.

Cross-platform validation strengthens confidence in antibody performance. For example, if establishing a new flow cytometry protocol, researchers should confirm CD4 expression patterns by orthogonal methods such as RT-PCR, western blotting, or immunohistochemistry. When analyzing tissue samples, co-localization with known T cell markers (e.g., CD3) provides contextual validation of staining patterns.

For functional studies or in vivo applications, dose-response relationships should be carefully established. The keliximab study exemplifies this approach, demonstrating distinct pharmacokinetic profiles and durations of detection in different dosing cohorts (0.5, 1.5, and 3.0 mg/kg) . Researchers should document not only the immediate effects of CD4 antibody administration but also the temporal dynamics of these effects, as clearance rates vary significantly between antibody preparations and experimental systems.

Finally, batch-to-batch variation should be assessed when transitioning to new antibody lots. This validation should include side-by-side comparisons of staining patterns, binding kinetics, and functional outcomes to ensure experimental continuity and reproducibility across studies.

How can CD4 antibodies be employed in therapeutic applications and what are the considerations for translation?

CD4 monoclonal antibodies have shown significant potential in therapeutic applications, particularly for immune-mediated disorders, though their translation to clinical use requires careful consideration of multiple parameters. The keliximab (IDEC CE9.1) study exemplifies this approach, demonstrating the effects of an anti-CD4 monoclonal antibody on peripheral blood CD4+ T-cells in corticosteroid-dependent asthmatics . This study illustrated important pharmacokinetic considerations, showing that different dosing cohorts (0.5, 1.5, and 3.0 mg/kg) resulted in distinctly different serum concentration profiles and clearance rates, with higher doses maintaining detectable antibody levels for up to 4 weeks .

For therapeutic translation, antibody purity and freedom from contaminants are paramount. The purification process developed for MAX.16H5 demonstrates the rigorous standards required, employing multiple chromatographic steps and extensive analytical characterization to confirm the absence of mouse DNA, viruses, pyrogens, and irritants before advancing to human therapy . This multi-faceted purification approach is essential for minimizing immunogenicity and adverse effects in clinical applications.

Humanization or fully human antibody development represents another critical consideration for reducing immunogenicity in therapeutic applications. While many research-grade CD4 antibodies are mouse-derived (e.g., OKT-4, RM4-5), therapeutic applications typically require humanized or fully human antibodies to minimize anti-drug antibody responses that can neutralize therapeutic efficacy and potentially cause hypersensitivity reactions.

Mechanism of action must be precisely defined for therapeutic development. CD4 antibodies may function through various mechanisms: depleting CD4+ cells, modulating CD4 receptor levels without depletion, blocking CD4-MHC class II interactions, or inducing regulatory T cell populations. Each mechanism has distinct implications for therapeutic applications and potential adverse effects. Understanding the precise epitope recognized by the antibody and its functional consequences is essential for predicting therapeutic outcomes.

Finally, formulation and delivery considerations significantly impact therapeutic potential. In vivo GOLD functional grade preparations specifically designed for preclinical research use aseptic manufacturing in animal-free facilities and employ multi-step purification processes to ensure extremely low levels of endotoxins (<1.0 EU/mg) and leachable protein A . These high standards for preclinical antibodies provide a foundation for subsequent GMP-grade production required for clinical translation.

What advanced imaging and analytical techniques benefit from highly purified CD4 antibodies?

Advanced imaging and analytical techniques increasingly rely on highly purified CD4 antibodies to achieve the sensitivity, specificity, and resolution required for sophisticated immunological investigations. Multiplexed imaging technologies such as PhenoCycler® (previously CODEX) utilize purified CD4 antibodies in conjunction with other immune markers to create detailed spatial maps of immune cell distributions and interactions within tissues . These approaches benefit from high antibody purity and defined epitope specificity to minimize cross-reactivity when simultaneously visualizing multiple targets.

Super-resolution microscopy techniques, including stimulated emission depletion (STED) microscopy, structured illumination microscopy (SIM), and single-molecule localization methods, push spatial resolution below the diffraction limit to examine nanoscale distribution of CD4 on cell membranes. These techniques demand exceptionally pure antibody preparations with minimal aggregation and precise target recognition to achieve accurate molecular localization. The monomer purity of ≥95% achieved through analytical size exclusion chromatography is particularly critical for these applications .

Mass cytometry (CyTOF) and spectral flow cytometry represent powerful high-dimensional single-cell analysis platforms that benefit from highly purified CD4 antibodies. In these systems, antibodies are conjugated to metal isotopes or fluorophores with distinct spectral signatures, allowing simultaneous measurement of dozens of cellular parameters. Antibody purity directly impacts signal-to-noise ratios and the accuracy of cellular phenotyping in these complex assays.

Single-cell RNA sequencing combined with protein detection (CITE-seq, REAP-seq) integrates transcriptomic and proteomic information at single-cell resolution. These methods often use oligonucleotide-labeled antibodies, including anti-CD4, to correlate surface protein expression with gene expression profiles. The specificity and purity of CD4 antibodies significantly influence the accuracy of protein quantification and subsequent multimodal data integration.

Live-cell imaging applications, including intravital microscopy for tracking CD4+ T cell dynamics in vivo, require antibody fragments or non-blocking antibodies that maintain target recognition without perturbing normal cellular functions. The specific epitope recognized by different CD4 antibody clones becomes particularly important in these applications, as some epitopes may induce signaling or alter cellular behavior upon antibody binding, potentially confounding experimental interpretations.

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.