Anti-Human CD4 (Ibalizumab) (Clone TNX-355)

Anti-Human CD4 (Ibalizumab) (Clone TNX-355)

Product No.: LT3200

- -
- -
Product No.LT3200
Clone
TNX-355
Target
CD4
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
CD4, CD4mut, CD4 molecule, OKT4D, IMD79
Isotype
Human IgG4κ
Applications
ELISA
,
FA
,
FC
,
IHC
,
N
,
WB

- -
- -
Select Product Size

Data

Western Blot Data for Anti-Human CD4 Clone TNX-355 Leinco Product No. LT3200Western Blot
Western blot shows Recombinant human CD4. Membrane was probed with 15ug/mL of Human Anti-Human CD4 Ibalizumab (Leinco Prod. No.: LT3200) followed by HRP conjugated Goat Anti-Human IgG secondary antibody (Leinco Prod. No.: H215).
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Recommended Isotype Controls
Immunogen
Recombinant Human CD4
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
< 1.0 EU/mg as determined by the LAL method
Purity
≥95% by SDS Page
≥95% monomer by analytical SEC
Formulation
This biosimilar antibody is aseptically packaged and formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.2 - 7.4 with no carrier protein, potassium, calcium or preservatives added. Due to inherent biochemical properties of antibodies, certain products may be prone to precipitation over time. Precipitation may be removed by aseptic centrifugation and/or filtration.
State of Matter
Liquid
Product Preparation
Recombinant biosimilar antibodies are manufactured in an animal free facility using only in vitro protein free cell culture techniques and are purified by a multi-step process including the use of protein A or G to assure extremely low levels of endotoxins, leachable protein A or aggregates.
Storage and Handling
Functional grade preclinical antibodies may be stored sterile as received at 2-8°C for up to one month. For longer term storage, aseptically aliquot in working volumes without diluting and store at ≤ -70°C. Avoid Repeated Freeze Thaw Cycles.
Regulatory Status
Research Use Only (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Applications and Recommended Usage?
Quality Tested by Leinco
ELISA
WB
Additional Applications Reported In Literature ?
FA
IHC-FF
FC
N
Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change.

Description

Description

Specificity
This non-therapeutic biosimilar antibody uses the same variable region sequence as the therapeutic antibody Ibalizumab. This product is for research use only. Ibalizumab binds to domain 2 of CD4 T cell receptors, on the protein surface opposite where the major histocompatibility complex-class II and HIV-1 gp120 binding sites are located. Ibalizumab binds to both human and monkey CD4.
Background
CD4 is a cell surface glycoprotein essential for both T cell activation and human immunodeficiency virus type-1 (HIV-1) infection 1,2. CD4 consists of an extracellular segment composed of four tandem immunoglobulin-like domains (D1 to D4), a single transmembrane span, and a short C-terminal cytoplasmic tail 1. HIV-1 entry into host CD4 cells is a complex process that occurs through the interaction of HIV-1 glycoprotein 120 (gp120) with extracellular CD4 D1 3,4. When gp120 binds to CD4, a conformational shift occurs that allows co-receptors to bind to the gp120/receptor complex, leading to viral fusion and entry. Ibalizumab is the first CD4-directed post-attachment HIV-1 entry inhibitor and prevents entry of a broad spectrum of HIV-1 isolates 1,5,6,7,8.

Ibalizumab selectively binds to an epitope on CD4 D2 (residues 121-124 and 127-134 9 and especially L96, P121, P122, and Q163 1,7) as well as residues E77 and S79 on D1 at the interface between D1 and D2 4,7. Ibalizumab primarily contacts the BC-loop in D2 at the D1-D2 junction on the opposite side to the gp120 and major histocompatibility complex II (MHC-II) binding sites 2. Ibalizumab does not inhibit HIV-1 gp120 binding to D1, but instead induces conformational changes that via steric hindrance block gp120 and HIV co-receptors from interacting, thereby preventing viral fusion and entry 3,4,7,10,11. Additionally, because the cellular epitope is distant from the D1 MHC-II binding site 4, MHC-II mediated immunosuppression is prevented 3. Furthermore, as a humanized IgG4 antibody, ibalizumab displays low affinity for C1q and FcɣRI receptors of natural killer cells and consequently has low cellular cytotoxic dependent activity and no Fc-mediated CD4+ T cell depletion 3.

Ibalizumab was derived from mu5A8 by grafting the mouse complementary-determining region onto a human IgG4 construct 1,3,12. The chemical name is immunoglobulin G4, anti-(human CD4 (antigen)) (human-mouse monoclonal 5A8 γ4-chain), disulphide with human-mouse monoclonal 5A8 κ-chain, dimer 5.
Antigen Distribution
CD4 is primarily found on T lymphocytes.
Ligand/Receptor
CD4/CD4 receptor
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
HIV
.
Immunology
.
Pathology

Leinco Antibody Advisor

Powered by AI: AI is experimental and still learning how to provide the best assistance. It may occasionally generate incorrect or incomplete responses. Please do not rely solely on its recommendations when making purchasing decisions or designing experiments.

Research-grade Ibalizumab biosimilars can be used as calibration standards or reference controls in a pharmacokinetic (PK) bridging ELISA to measure drug concentration in serum samples by employing a precise and robust analytical strategy. Here's how they are utilized:

Calibration and Reference Controls

  1. Calibration: Biosimilars are calibrated against international standards, such as those from the National Institute of Biologicals and Control (NIBSC), to ensure accuracy and consistency in the measurement of drug concentrations.
  2. Reference Controls: These biosimilars serve as reference controls by providing a standard against which unknown samples can be compared. This is crucial in PK bridging ELISAs, where the goal is to assess the bioequivalence of the biosimilar to the reference product.

PK Bridging ELISA

  1. Biosimilar Use: In a PK bridging ELISA, biosimilars are used within a single assay to measure both the biosimilar and the reference product. This approach helps reduce variability and streamlines the process.
  2. ELISA Format: ELISAs are typically performed in a sandwich format, using anti-idiotypic monoclonal antibodies to ensure specificity and sensitivity in detecting Ibalizumab levels in serum.

Pharmacokinetic Studies

  1. PK Studies: The assays are critical for demonstrating the pharmacokinetic profiles of the biosimilar and reference products. They help in assessing bioequivalence by comparing their concentration profiles in patients.
  2. Regulatory Compliance: Data from these assays are essential for regulatory submissions, providing evidence of biosimilarity in terms of safety, efficacy, and pharmacokinetics.

ELISA Performance

  1. Sensitivity and Specificity: ELISAs are optimized for high sensitivity and specificity to detect low concentrations of Ibalizumab, ensuring reliable pharmacokinetic data.
  2. Validation: Assays are validated according to stringent guidelines (e.g., FDA and EMEA requirements) to ensure precision and accuracy. This involves assessing intra- and inter-assay variability and ensuring that there is no matrix interference from serum or plasma.

By using biosimilar Ibalizumab as calibration standards or reference controls in PK bridging ELISAs, researchers can accurately measure drug concentrations, assess pharmacokinetics, and support regulatory submissions for biosimilar approval.

The primary models where research-grade anti-CD4 antibodies are administered in vivo to study tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse tumor models. Humanized mouse models are less commonly used for anti-CD4 studies due to the species specificity of available antibodies and differences in immune repertoires.

Details and context:

  • Syngeneic Models:
    Syngeneic mouse tumor models are the most common systems for in vivo administration of anti-mouse CD4 antibodies. In these studies, murine tumors (e.g., B16F10, CT26, MC38, Renca, 4T1, Sa1N) are implanted in immunocompetent mice of the same genetic background. Research-grade anti-mouse CD4 antibodies are used for depletion or functional blockade of CD4+ T cells.

    • Example: Mice are given anti-CD4 antibody (such as clone GK1.5) intraperitoneally before or after tumor challenge to deplete CD4+ T cells. Tumor growth inhibition can then be monitored, and TILs—specifically CD8+ T cells—are characterized via flow cytometry or immunohistochemistry. These models enable direct study of how CD4+ T cell depletion alters both tumor progression and the phenotype of TIL populations (e.g., expansion of tumor-specific CD8+ T cells).

    • Imaging-labeled or depleting antibodies specific for mouse CD4 are available for use in these models, supporting robust in vivo and ex vivo characterization of tumor-infiltrating immune cells.

  • Humanized Mouse Models:
    There are limited reports of in vivo anti-CD4 antibody administration in humanized mouse models for tumor/TIL studies. This is due to:

    • The requirement for anti-human CD4 antibodies that cross-react with the human immune system engrafted in the mouse.
    • The complexity and cost of establishing these models.
    • Lack of widespread use in standard studies of tumor growth inhibition relative to the extensive literature and tools available for syngeneic models.

    Most research using anti-CD4 antibodies for in vivo depletion and TIL characterization focuses on syngeneic mouse models due to antibody compatibility and the ability to observe robust immune responses in a fully immunocompetent host.

Additional Notes:

  • Studies using anti-CD4 antibodies often analyze effects on CD8+ T cell expansion, Treg numbers, TIL phenotype, and overall tumor control.
  • Tumor growth inhibition and immune profiling (TIL quantification and functional assays) are key endpoints in these studies.
  • Anti-CD4 depletion is often combined with other interventions—such as checkpoint inhibitors or agonistic antibodies—to study combination effects on the tumor microenvironment.

In summary, syngeneic mouse tumor models are the main systems in which research-grade anti-CD4 antibodies are administered in vivo to study tumor growth inhibition and TIL dynamics, while humanized models are less frequently used due to logistical and technical restrictions.

Researchers use Ibalizumab biosimilars—non-therapeutic antibodies mirroring the clinical molecule—to study immune modulation in advanced immune-oncology models, often in parallel or combination with other checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 biosimilars to elucidate possible synergistic effects.

Research Approach and Methodology

  • Model Systems: Ibalizumab biosimilars are used in preclinical models (cell lines, humanized mouse models, organoids) to investigate their specific effects on immune cell function, particularly through the modulation of human CD4+ T cell populations.
  • Mechanism Exploration: Unlike classical checkpoint inhibitors, ibalizumab binds the CD4 receptor on a unique epitope (D1-D2 junction of CD4), blocking crucial viral or pathological cell interactions without depleting CD4+ T cells or suppressing MHC-II-mediated immunity. This makes it valuable for examining immune modulation without overt immunosuppression.
  • Combination Studies: Researchers combine Ibalizumab biosimilars with other checkpoint inhibitor biosimilars (e.g., anti-CTLA-4, anti-LAG-3) to:
    • Interrogate distinct immune regulatory pathways. For example, CTLA-4 and LAG-3 act via different checkpoints and cellular compartments than CD4, so their combined inhibition may unlock additive or synergistic anti-tumor activity not visible with monotherapy.
    • Study potential enhanced T cell activation, tumor infiltration, and tumor regression when multiple inhibitory axes are blocked.
    • Assess the effect on toxicity profiles and immune-related adverse events, comparing outcomes to monotherapy and establishing therapeutic windows.
  • Outcome Measures: Researchers evaluate:
    • Cytokine production, T cell proliferation, and functional assays.
    • Tumor growth and survival in animal models.
    • Single-cell RNA sequencing and flow cytometry to map repertoire and phenotypic changes in immune populations.

Example Workflow

  1. Treat immune-oncology models (often humanized mouse models with tumor xenografts) with:
    • Ibalizumab biosimilar alone.
    • Anti-CTLA-4 or anti-LAG-3 biosimilar alone.
    • Combinations of both or additional checkpoint inhibitors.
  2. Assess end points such as tumor volume, immune infiltration, T cell function, and expression of inhibitory markers.

Rationale

  • Multi-checkpoint blockade: Given that different immune checkpoints regulate T cell activity at different levels and contexts (priming versus effector function, lymphoid versus tumor microenvironment), combining inhibitors may overcome adaptive resistance to single agent therapy and produce more robust anti-tumor responses.
  • The biosimilar format provides a cost-effective and scalable way for early research, ensuring that pharmaceutical-grade molecules are reserved for later-stage translational and clinical studies.

Limitations and Special Considerations

  • Toxicity and adverse events: Multi-checkpoint combinations can amplify immune-related adverse effects, seen in clinical studies of CTLA-4/PD-1 dual blockades; such phenomena must be carefully modeled preclinically.
  • Translational gaps: While biosimilar use in preclinical models is invaluable, results may not always fully predict clinical efficacy or safety without further validation.

In summary, use of ibalizumab biosimilars in conjunction with other checkpoint inhibitor biosimilars in research settings allows detailed dissection of complex immune interactions, aids in identifying promising multi-target therapy strategies, and supports the optimization of next-generation immune-oncology combinatorial regimens.

In a bridging anti-drug antibody (ADA) ELISA for immunogenicity testing, an Ibalizumab biosimilar can be used either as the capture or detection reagent because it mimics the structure of the therapeutic drug, enabling sensitive and specific detection of antibodies generated in patients against Ibalizumab.

Key steps and rationale:

  • In a bridging ELISA, the assay detects ADA that can bind two Ibalizumab molecules simultaneously: one immobilized on the plate (capture) and one labeled with a detection marker (e.g., HRP or biotin).
  • The Ibalizumab biosimilar serves as the test reagent rather than the reference drug to ensure supply, cost-effectiveness, and to avoid interference with the clinical therapeutic.
  • Typically:
    • The microtiter plate is coated with the Ibalizumab biosimilar to capture ADA present in the patient serum.
    • After washing, a labeled form of the same biosimilar is added; if ADA is present, it forms a “bridge” between the immobilized and labeled biosimilar, enabling detection.
  • The bridging format is advantageous because it only detects bivalent antibodies (generally immunoglobulins), making it specific for true ADA rather than unrelated binding components.
  • This methodology has broad precedent in immunogenicity assays for monoclonal antibodies and is adaptable to new biopharmaceuticals—including Ibalizumab and its biosimilars.

Summary table: Bridging ADA ELISA components

RoleReagentFunction
Capture reagentIbalizumab biosimilar (unlabeled)Binds ADA from patient sample
Detection reagentLabeled Ibalizumab biosimilarBinds to the other arm of ADA and allows detection (e.g., via HRP substrate)
SamplePatient serumSource of potential anti-Ibalizumab antibodies

This setup enables monitoring of a patient’s immune response to the drug during therapy, fulfilling regulatory and clinical requirements for immunogenicity surveillance.

References & Citations

1. Song R, Franco D, Kao CY, et al. J Virol. 84(14):6935–6942. 2010.
2. Freeman MM, Seaman MS, Rits-Volloch S, et al. Structure. 18(12):1632–1641. 2010.
3. Iacob SA, Iacob DG. Front Microbiol. 8:2323. 2017.
4. Chahine EB, Durham SH. Ann Pharmacother. 55(2):230-239. 2021.
5. Markham A. Drugs. 78(7):781-785. 2018.
6. Reimann KA, Lin W, Bixler S, et al. AIDS Res Hum Retroviruses. 13(11):933-943. 1997.
7. Beccari MV, Mogle BT, Sidman EF, et al. Antimicrob Agents Chemother. 63(6):e00110-19. 2019.
8. Blair HA. Drugs. 80(2):189-196. 2020.
9. Burkly LC, Olson D, Shapiro R, et al. J Immunol. 149:1779–1787. 1992.
10. Moore JP, Sattentau QJ, Klasse PJ, et al. J Virol. 66(8):4784-4793. 1992.
11. Bettiker RL, Koren DE, Jacobson JM. Curr Opin HIV AIDS. 13(4):354-358. 2018.
12. Boon L, Holland B, Gordon W, et al. Toxicology. 172(3):191-203. 2002.
13. Reimann KA, Khunkhun R, Lin W, et al. AIDS Res Hum Retroviruses. 18(11):747-755. 2002.
14. Kuritzkes DR, Jacobson J, Powderly WG, et al. J Infect Dis. 189(2):286-291. 2004.
15. Jacobson JM, Kuritzkes DR, Godofsky E, et al. Antimicrob Agents Chemother. 53(2):450-457. 2009.
16. Toma J, Weinheimer SP, Stawiski E, et al. J Virol. 85(8):3872–3880. 2011.
17. Pace CS, Fordyce MW, Franco D, et al. J Acquir Immune Defic Syndr. 62(1):1–9. 2013.
Indirect Elisa Protocol
FA
Flow Cytometry
IHC
N
General Western Blot Protocol

Certificate of Analysis

- -
- -

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.