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.
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.
Pathogen Testing
To protect mouse colonies from infection by pathogens and to assure that experimental preclinical data is not affected by such pathogens, all of Leinco’s recombinant biosimilar antibodies are tested and guaranteed to be negative for all pathogens in the IDEXX IMPACT I Mouse Profile.
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.
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 Mogamulizumab. Clone KW-0761 recognizes human CD194 (CCR4). This product is for research use only.
Background
Clone KW-0761 (Mogamulizumab) is a research-grade, humanized monoclonal antibody generated from mouse anti-CCR4 mAb7 that targets human CCR4.1 CC chemokine receptor type 4 (CCR4) is a protein that belongs to the G protein-coupled receptor family and is a receptor for a variety of CC chemokines including MCP-1, MIP-1, RANTES, TARC, and Macrophage-derived chemokine. Chemokines are involved in the development, homeostasis, and function of the immune system and are known to regulate cell trafficking of various types of leukocytes. In a 2018 Phase I clinical trial, Mogamulizumab was found to decrease the number of HTLV-1–infected cells and the levels of inflammatory markers related to HTLV-1–Associated Myelopathy.3
Antigen Distribution
CCR4 is expressed on a variety of cell types: T lymphocytes (Th2, Th17, and regulatory T cells), platelets, NK cells, monocytes, macrophages, dendritic cells, neurons, microglia, and astroglia.1 Expression of CCR4 is increased on leukemic cells in cutaneous T-cell lymphoma (CTCL).2
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 Mogamulizumab biosimilars are commonly used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays to measure drug concentrations in serum samples, providing a standardized approach for quantifying both biosimilar and innovator drug in clinical and preclinical studies.
Key points about their use:
Single Analytical Standard: Regulatory and industry guidance recommends developing a single PK ELISA method that uses one well-characterized analytical standard, typically the biosimilar, to create the calibration curve for all measurements. This approach reduces assay variability and supports robust, cross-study comparison.
Calibration Curve Construction: The ELISA kit includes lyophilized calibration standards (often the biosimilar) at defined concentrations. These standards are diluted and run in parallel with test serum samples to generate a standard curve, which quantitatively relates optical density (OD) signals to Mogamulizumab concentration in the samples.
Assay Validation and Equivalence: A critical step is the bioanalytical comparability assessment: testing both the innovator and biosimilar drugs in the assay during method validation. Analytical equivalence is established if both products deliver similar quantitative results when measured against the biosimilar calibration standard. This ensures the reliability of the assay for both reference and biosimilar drug detection.
Bridging ELISA Format: In the bridging ELISA, antibodies specific to Mogamulizumab’s idiotype (anti-idiotypic antibodies) capture the drug from serum samples, enabling specific detection even in complex biological matrices. This design enables the assay to measure drug concentrations regardless of slight physicochemical differences between biosimilars and innovator.
Use as Controls: Aside from calibration, research-grade biosimilars may be included as external or quality control samples at known concentrations, enabling the monitoring of assay accuracy and precision across runs and batches.
Summary of application workflow:
Calibration standards (biosimilar) are serially diluted in serum/plasma and included on each plate.
Patient or preclinical serum samples are assayed in parallel.
Drug concentration in each sample is determined by referencing the standard curve.
The assay is validated to ensure equivalent detection and quantification of both the biosimilar and innovator products, typically following FDA, EMA, and ICH guidelines.
This approach ensures consistent, accurate measurement of Mogamulizumab drug levels in serum—critical for PK and bioequivalence studies—by minimizing cross-assay variation and supporting regulatory compliance.
Primary Models for Anti-CD194 Antibody Administration in Vivo
Syngeneic mouse models and humanized mouse models are the principal platforms used in research to evaluate novel immunotherapies such as anti-CD194 (CCR4) antibodies for tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization. While the provided search results do not directly describe studies with an anti-CD194 antibody, they offer foundational insights into the standard approaches and rationale for using these models in similar contexts.
Syngeneic Mouse Models
Mechanism and Rationale: Syngeneic models involve implanting mouse-derived tumor cells into immunocompetent mice of the same genetic background. These models retain a fully functional immune system, enabling researchers to study the dynamic interplay between tumor cells and host immunity—critical for characterizing TILs and evaluating immune-mediated tumor inhibition.
Model Selection: Commonly used syngeneic lines (e.g., MC38, 4T1, CT26, EMT6, RENCA, B16F10) each have distinct immune infiltrate profiles, ranging from highly immunogenic (RENCA, CT26) to poorly infiltrated (B16F10). The choice of model depends on the desired immunological context and tumor type under investigation.
Application: These models are widely used to demonstrate the activity of novel immunotherapies, including checkpoint inhibitors and other antibody-based approaches, and to profile changes in TIL populations (e.g., CD8⁺, CD4⁺ T cells, regulatory T cells, myeloid-derived suppressor cells). For example, studies have shown that drug combinations can increase cytotoxic CD8⁺ T cell infiltration and that depletion of these cells abolishes anti-tumor effects, highlighting the importance of a functional immune system for therapeutic efficacy.
Advantages: Syngeneic models are predictive for immunotherapy efficacy, allow detailed immune profiling, and are relatively easy to establish and maintain. They are especially valuable for screening and mechanistic studies before advancing to more complex or human-specific models.
Humanized Mouse Models
Mechanism and Rationale: Humanized mouse models involve engrafting human tumors or immune cells into immunodeficient mice (e.g., NSG, NOG strains), sometimes with additional transgenic components to support human immune cell function. These models are essential when the target (e.g., human CD194/CCR4) or the therapeutic agent (e.g., a human-specific antibody) is not cross-reactive with the mouse immune system.
Application: While the search results do not specifically reference anti-CD194 studies in humanized models, humanized mice are typically used when the compound under investigation is a human-specific antibody or when the interaction must occur within a human tumor microenvironment. This is common in later-stage preclinical development, especially for antibodies targeting human-specific antigens or pathways.
Advantages: These models enable evaluation of human antibody–human target interactions in vivo, provide a platform for studying human TIL dynamics, and can bridge the gap between preclinical and clinical studies for drugs that do not cross-react with murine targets.
Profiling TILs: Both syngeneic and humanized models support flow cytometry, immunohistochemistry, and single-cell sequencing to quantify and characterize TIL populations (e.g., CD8⁺, CD4⁺ T cells, Tregs, myeloid cells). For example, combination therapies in syngeneic models have been shown to increase CD8⁺ T cell infiltration and induce memory T cell responses.
Model-Specific Dynamics: The composition and dynamics of TILs vary by model. For instance, CT26 tumors show increasing CD8⁺ T cell density with tumor progression, while RENCA tumors exhibit a decline in T cell populations and enrichment of immunosuppressive myeloid cells as they grow.
Relevance to Human Tumors: The immune profiles of syngeneic models can be mapped to human tumor subtypes to identify those most likely to respond to specific immunotherapies, aiding translational research.
Conclusion
Syngeneic mouse models are the standard workhorse for initial in vivo evaluation of research-grade immunotherapies—including anti-chemokine receptor antibodies—where a functional murine immune system is required to study tumor growth inhibition and TIL dynamics. Humanized mouse models are employed when the therapeutic agent or target is human-specific, enabling studies of human antibody–human target interactions in a more clinically relevant context.
For anti-CD194 (CCR4) antibodies specifically, the choice between these models depends on the species specificity of the antibody and the scientific question. However, syngeneic models remain the primary platform for immune-oncology mechanism-of-action studies in immunocompetent hosts.
Researchers utilize Mogamulizumab biosimilars in combination with other checkpoint inhibitors—such as anti-CTLA-4 or anti-LAG-3 biosimilars—to investigate synergistic effects in complex immune-oncology models by targeting different immune regulatory pathways and evaluating both immunologic and clinical responses.
Key approaches and findings include:
Mechanistic Rationale: Mogamulizumab targets CCR4 on regulatory T cells and some malignant T cells, inhibiting their trafficking and promoting antitumor immune activity via antibody-dependent cellular cytotoxicity (ADCC). When combined with checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3), which act by reversing immune exhaustion or suppression, the hypothesis is that the combined blockade could result in more robust T cell activation and tumor regression.
Experimental Designs:
In preclinical models and early-phase clinical trials, combinations often involve biosimilar antibodies against CCR4 (Mogamulizumab) and checkpoint molecules (PD-1, CTLA-4, or LAG-3).
Researchers monitor parameters such as regulatory T cell (Treg) depletion, increase in effector CD8(^+) T cells, immune contexture of the tumor microenvironment, and changes in tumor burden.
Enhanced effects—such as increased lymphocyte infiltration or improved pathological response—may indicate synergy, especially if seen beyond what either agent achieves alone.
Clinical Evidence and Complexity:
Some clinical trials have evaluated combination therapies (e.g., Mogamulizumab with anti-CTLA-4 or anti-PD-1 monoclonal antibodies). While these studies have consistently shown depletion of Tregs in the blood and tumor tissue and sometimes increased CD8(^+) T cell infiltration, the overall improvement in therapeutic effect has been inconsistent. In some scenarios, no additional benefit was seen over monotherapy, possibly due to unintended depletion of CCR4(^+) effector T cells or emergence of alternative suppressive immune mechanisms.
Timing and clinical setting matter: Preoperative use in early-stage tumors has shown more promising results—such as increased intratumoral lymphocytes and improved pathological response—than use in advanced or bulky diseases.
Combination
Experimental Outcome
Potential Limitation/Insight
Mogamulizumab + anti-CTLA-4/PD-1/LAG-3
Treg depletion, occasionally increased CD8(^+) T cell infiltration; sometimes improved histological response
No consistent add-on clinical effect; off-target depletion of CCR4(^+) effectors; induction of suppressive myeloid populations
Checkpoint Inhibitor Monotherapy
Restores T cell function; variable response by tumor microenvironment
May be insufficient in context of high Treg burden
Biosimilars in Research: Biosimilars are increasingly used in preclinical models due to cost, accessibility, and reproducibility, allowing high-throughput evaluation of different combination regimens and immune microenvironmental effects before clinical translation.
In summary, the use of Mogamulizumab biosimilars alongside biosimilar checkpoint inhibitors is a promising research strategy to dissect and optimize synergistic immunotherapy regimens. Results suggest that efficacy depends on both the tumor milieu and the detailed immunologic consequences of dual targeting—most notably, depleting both suppressive and sometimes effector CCR4(^+) populations, which may blunt synergy under certain conditions.
A Mogamulizumab biosimilar can be used in a bridging anti-drug antibody (ADA) ELISA as either the capture reagent (immobilized on the plate) or detection reagent (labeled and soluble) because it is structurally and functionally equivalent to the reference antibody, allowing it to specifically bind anti-drug antibodies that developed in the patient's blood.
In a bridging ADA ELISA:
The drug (e.g., Mogamulizumab biosimilar) is immobilized on the microtiter plate to capture any ADAs present in patient serum.
After incubation and washing, labeled Mogamulizumab biosimilar (e.g., biotinylated or HRP-conjugated form) is added to detect bound ADAs; the ADA "bridges" between the capture and detection drug molecules since it has two binding sites (Fab arms) specific to the drug.
This antibody-antigen-antibody “bridge” enables sensitive detection using the label on the detection reagent.
The use of a biosimilar as the assay reagent is valid because it presents the same epitopes and structure as the reference product, ensuring that ADAs raised against the therapeutic (whether innovator or biosimilar) are equally detected in the assay.
The FDA and general immunogenicity methodology allow for the use of biosimilar reagents when they are analytically highly similar to the reference product.
Key steps in summary:
Coat the plate with Mogamulizumab biosimilar (capture).
Add patient sample; allow ADAs to bind if present.
Add labeled Mogamulizumab biosimilar (detection); ADAs, if present, bind both coated and labeled drug.
Detect the signal (e.g., colorimetric or chemiluminescent readout).
This format monitors immune response against the therapeutic by measuring the presence and titer of ADAs, which can affect the efficacy and safety of Mogamulizumab treatment. Biosimilar usage is standard practice when analytic comparability is established, ensuring assay reliability for both biosimilar and originator drugs.
References & Citations
1. Nicolay, J. et al. (2021) Eur J Immunol. 51(7):1660-1671.
2. Bogacka, J. et al. (2022) Int J Mol Sci.. 23(24):15638.
3. Yamamoto, K. et al. (2010) J Clin Oncol. 28(9):1591-8.
4. Mimura, Y. et al. (2018) Protein Cell 9(1):47-62.
5. Yamano, Y. et al. (2018) N Engl J Med 378 (6), 529-538.