Anti-Human CD4 (Ibalizumab) – Fc Muted™

Anti-Human CD4 (Ibalizumab) – Fc Muted™

Product No.: LT3205

- -
- -
Product No.LT3205
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
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
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 are used in pharmacokinetic (PK) bridging ELISAs as calibration standards or reference controls by serving as the analytical standard for quantitative measurement of Ibalizumab in serum samples, allowing direct comparison with the innovator/reference product. This supports method standardization, accuracy, and regulatory requirements.

Essential Context and Supporting Details:

  • Calibration Standard Role: In PK bridging ELISA, a single analytical standard—often the biosimilar—acts as the reference for establishing the assay's calibration curve. This enables quantification of both the biosimilar and reference (innovator) Ibalizumab across serum samples, ensuring direct comparability. The standard concentration range in commercial kits is typically specified (e.g., 0–2000 ng/ml).

  • Reference Controls: Research-grade biosimilar controls are used for quality control (QC) samples. These controls validate assay performance, enabling measurement consistency for both the biosimilar and the reference product. Such controls are crucial for demonstrating bioanalytical equivalence during regulatory submissions and method validation.

  • Assay Validation: PK bridging ELISA development mandates rigorous precision, accuracy, and comparability testing of the reference and biosimilar products within the assay. Validation steps often include:

    • Assessing recovery and matrix effects by spiking serum samples at various dilutions;
    • Conducting intra- and inter-assay variability studies (aiming for CV <10%);
    • Applying lyophilized standards for robust calibration and stability.
  • Bridging ELISA Rationale: Using a single PK assay calibrated to the biosimilar standard streamlines the analytical workflow, avoids inter-assay variability, and allows blinded clinical sample analysis. After scientific confirmation of bioanalytical comparability, the biosimilar is selected as the sole calibration standard, meaning all test samples (biosimilar and innovator) are quantified against it.

  • Reference and Control Usage:

    • Calibration Standard: Used to generate calibration curves for concentration quantification.
    • Reference Control: Benchmarks assay performance and supports QC sample quantification (prepared with both biosimilar and reference drug).

Additional Relevant Information:

  • Regulatory Compliance: This strategy aligns with best practices for ligand-binding assays (LBA) under FDA and EMA guidance. Rigorous method validation and comparability data are pivotal for regulatory approval of biosimilars.

  • Kit Construction: Commercial PK ELISA kits (e.g., for Ibalizumab) include lyophilized research-grade biosimilar standards to ensure reproducibility and stability, and are generally validated against international standards for enhanced accuracy.

  • Biosimilar Utility Beyond PK ELISA: Research-grade biosimilar antibodies serve not only as assay calibrators but also as capture/detection reagents in numerous assay designs.

Summary Table: Role of Ibalizumab Biosimilars in PK Bridging ELISA

UsagePurposeBenefit
Calibration StandardEstablish assay curve; quantify serum concentrationDirect comparison; streamlined quantification
Reference ControlEnsure accuracy; validate assay performanceReliable QC; supports regulatory submissions
Assay ReagentsCan serve as capture/detection antibody in sandwich ELISAExpanded utility in assay optimization

In conclusion, research-grade Ibalizumab biosimilars are central to assay calibration and reference control in PK bridging ELISA, supporting robust, precise, and regulatory-compliant measurement of drug concentrations in serum samples for both biosimilar and innovator products.

The primary models where a research-grade anti-CD4 antibody is administered in vivo to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic murine tumor models. These are widely used due to species compatibility, well-defined mouse strains, and established tumor lines.

Key Points and Supporting Details:

  • Syngeneic Murine Models:
    The administration of anti-CD4-depleting monoclonal antibodies in syngeneic mouse models (where tumor cells and host are from the same genetic background) is the established approach. Commonly used models include B16F10 (melanoma), Colon 26 (colon carcinoma), Lewis lung carcinoma, CT26 (colon), MC38 (colon), P815 (mastocytoma), Renca (renal), 4T1 (breast), and Sa1N. These models allow direct assessment of immune cell depletion on tumor growth within an intact, immunocompetent system.

  • Mechanistic Studies:
    Studies have demonstrated that CD4+ T cell depletion via anti-CD4 monoclonal antibodies in these models leads to pronounced inhibition of tumor growth, primarily through enhancement of CD8+ T cell-mediated responses. Tumor growth inhibition is frequently associated with increased infiltration and activity of tumor-specific CD8+ T cells, which can be directly assessed by analyzing TIL populations in excised tumors.

  • Characterization of TILs:
    Flow cytometry and immunohistochemistry are routinely used on tumors collected from these models to characterize CD4+ and CD8+ TILs, including markers of activation, exhaustion (e.g., PD-1, CD44, CD69), and functional status. These models support both direct in situ characterization and ex vivo expansion of TILs for functional assays.

  • Humanized Models:
    Although humanized mouse models (mice engrafted with components of the human immune system) are used in some research, the majority of published studies using research-grade anti-CD4 antibodies (usually rat anti-mouse CD4) for TIL analysis and tumor inhibition utilize syngeneic mouse models. Humanized models typically require antibodies specific for human CD4 and have complexities regarding engraftment efficiency and the fidelity of immune responses.

Summary Table: Major In Vivo Models Using Anti-CD4 Antibody for Tumor and TIL Studies

Model TypeSpeciesExample Tumor LinesTypical Antibody UsedTIL CharacterizationCitation
Syngeneic murineMouseB16F10, CT26, MC38Rat anti-mouse CD4 (e.g., GK1.5)Flow, IHC, ex vivo expansion
Humanized mouse (rare for anti-mouse CD4 studies)Mouse with human immune systemHuman tumor xenograftsHuman-specific anti-CD4Flow, IHC[mainly inferential]

Key References from Search Results:

  • Administration of anti-CD4 antibody in B16F10, Colon 26, Lewis lung carcinoma models with TIL analysis.
  • Expanded use across CT26, MC38, P815, Renca, 4T1, Sa1N tumor models; quantification of TILs post-therapy.
  • Characterization of TIL function and phenotype in orthotopic and subcutaneous tumor models following CD4 depletion.

To date, syngeneic murine models remain the primary and best-characterized system for such studies.

Researchers use Ibalizumab biosimilar—a non-depleting, Fc-muted anti-CD4 monoclonal antibody—for immune modulation in preclinical studies, often combining it with other checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 biosimilars to investigate synergistic effects in complex immune-oncology models.

Context and Mechanisms:

  • Ibalizumab biosimilar mirrors the variable region of the therapeutic antibody, but is formulated for research use and does not mediate cytotoxic depletion of CD4+ T cells due to its unique Fc modifications (IgG4 structure and Fc mutation).
  • Instead of inhibiting normal immune function (e.g., MHC-II interaction), Ibalizumab binds away from the MHC-II site on domain D1–D2 of CD4, avoiding global immunosuppression while modifying CD4 conformation and accessibility.
  • This property allows rodents or humanized immune models to retain antigen presentation and general T cell activation even during antibody exposure, providing a clean platform for combinatorial studies.

Combination Strategies in Immune-Oncology Models:

  • Combining checkpoint inhibitors with distinct mechanistic targets can enhance antitumor immune activity by simultaneously overcoming multiple immunoregulatory barriers. For example:
    • Anti-CTLA-4 acts mainly in lymph nodes to promote the priming and expansion of new T cells.
    • Anti-LAG-3 and similar inhibitors can synergize with anti-PD-1/PD-L1 blockade by restoring or enhancing T cell function both at the tumor site and in peripheral tissues.
    • Pairing these with a CD4-directed agent like Ibalizumab biosimilar allows for precise dissection of how helper T cell modulation impacts, or synergizes with, checkpoint inhibition.

Typical Research Approach:

  • Complex immune-oncology models (e.g., humanized mouse models or organoids) are treated with:
    • Ibalizumab biosimilar to alter helper T cell signaling or trafficking without causing CD4+ T cell depletion.
    • One or more checkpoint inhibitor biosimilars (anti-CTLA-4, anti-LAG-3) to release brakes on T cell activation and function.
  • Researchers monitor parameters such as tumor growth, T cell infiltration, cytokine production, and immune cell composition to detect synergy—i.e., antitumor effects greater than the sum of each monotherapy.

Utility of Biosimilars:

  • Using biosimilars ensures experimental consistency, cost-effectiveness, and access for exploratory research where clinical-grade agents are impractical.
  • Preclinical studies with biosimilars inform which combinations are most promising for advancing to clinical trials.

Summary Table: Ibalizumab vs. Other Checkpoint Inhibitor Biosimilars in Synergy Studies

AgentTargetMode of ActionUse in Combinations
Ibalizumab biosimilarCD4Non-depleting, blocks gp120, preserves MHC-II signalingModulates CD4+ signaling to study helper T cell–dependent synergy
Anti-CTLA-4 biosimilarCTLA-4Releases T cell priming in lymph nodesCombined to boost T cell expansion response
Anti-LAG-3 biosimilarLAG-3Restores exhausted T cells, boosts effector functionUsed to further augment antitumor immunity

Researchers leverage these combinations to identify novel therapeutic synergies and optimize checkpoint modulation strategies in cancer immunotherapy research.

A biosimilar of Ibalizumab can be employed as a capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor patient immune responses to the therapeutic drug by leveraging the drug’s antigenic properties to bind ADAs present in patient serum.

In a typical bridging ADA ELISA for monoclonal antibodies like Ibalizumab, the assay setup involves:

  • Capture Reagent: Ibalizumab biosimilar is immobilized on the ELISA plate.
  • Sample Incubation: Patient serum is added; if anti-Ibalizumab antibodies (ADAs) are present in the serum, they bind to the immobilized Ibalizumab.
  • Detection Reagent: The same Ibalizumab biosimilar, labeled (e.g., with HRP or biotin), is added as the detection reagent; it binds to the ADA, creating a “bridge” between the plate and detection layer via the bivalent ADA.

This format is called “bridging” because the ADA forms a bridge between two molecules of Ibalizumab biosimilar—one on the plate, one in solution—enabling detection only when ADA is present.

The advantages of using a biosimilar (rather than the reference product) as the reagent include:

  • Specificity: The biosimilar is structurally and antigenically almost identical to the reference drug, ensuring detection of ADA targeting the relevant epitopes.
  • Interchangeability: Regulatory guidelines often allow biosimilars for use in ADA assays due to their high degree of similarity to the reference product, making them suitable capture or detection reagents.
  • Practicality: Using a ready supply of biosimilar material may be more cost-effective and logistically feasible.

Detection is then performed via enzymatic or chemiluminescent readouts, with signal intensity corresponding to the amount of ADA captured. This method is well-established for monoclonal antibody immunogenicity testing across a range of products.

Essential context:

  • The highly specific interaction of the ADA with the therapeutic and detection antibody ensures assay selectivity.
  • Sensitivity depends on both the quality of the biosimilar used as reagent and the detection system employed.

Additional Notes:

  • Regulatory agencies recommend well-validated, comparative approaches for immunogenicity assessment of biosimilars versus reference products.
  • In the context of Ibalizumab, immunogenicity monitoring is critical given its therapeutic use in HIV, where immune response can impact efficacy and safety.

Summary Table: Ibalizumab (or Biosimilar) in Bridging ADA ELISA

ComponentRoleExplanation
BiosimilarCapture & Detection reagentBinds patient-derived ADA on solid phase and detects via labeled biosimilar
Patient serumProvides potential ADABridge forms if ADA is present
Readout (HRP/TMB)VisualizationQuantifies ADA via colorimetric/chemiluminescent signal

This approach ensures accurate monitoring of immunogenicity in patients treated with Ibalizumab or its biosimilars.

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.