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.
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 Epcoritamab. Epcoritamab is a bispecific antibody
that targets CD3 on T-cells and CD20 on B-cells.
Background
Epcoritamab is a CD20xCD3 Bispecific T cell Engager (BiTE) antibody developed as a
cancer immunotherapeutic drug 1 . Simultaneous targeting of CD20 B cell on
lymphomas and CD3 on T cells, leads to T cell activation, the release of perforin and
granzymes, and ultimately the depletion of malignant B cells through lysis and cell
death 1 . This approach is potent in treating B-cell malignancies like non-Hodgkin lymphoma.
The distribution of Anti-Human CD3 x CD20 includes blood and lymphatic tissues, enabling
effective engagement with T and B-cells, enhancing its therapeutic efficacy2-4.
Epcoritamab (GEN3013) is a bispecific antibody that targets CD3 on T-cells and CD20 on B-
cells, inducing potent T-cell-mediated cytotoxicity against CD20-expressing B-cells.
Administered subcutaneously, Epcoritamab has demonstrated significant efficacy in clinical
trials for relapsed or refractory B-cell lymphomas. In laboratory settings, Epcoritamab is
utilized to study bispecific antibody mechanisms and develop therapeutic strategies for B-cell
malignancies5-7.
Antigen Distribution
CD20 is widely expressed on normal B cells during all stages of
development, as well as by most B cell malignancies. CD3 is a T cell surface
glycoprotein.
Ligand/Receptor
CD3ε: TCR
CD20: Src family tyrosine kinases, MHC class I, II, CD53, CD81, CD82
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 Epcoritamab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays to generate standard curves for quantifying drug concentration in serum samples. The following explains their roles and the scientific rationale in detail:
Calibration Standards: In a PK bridging ELISA, a known concentration series of Epcoritamab biosimilar is added to drug-free serum or plasma, generating a standard curve against which the concentration of Epcoritamab in test samples is interpolated. Using biosimilar (non-clinical grade but structurally and functionally equivalent) Epcoritamab allows for consistent standardization, especially when the reference (clinical or commercial) Epcoritamab is limited, costly, or unavailable for routine assay calibration.
Reference Controls: Along with calibration standards, at least one reference control (a quality control sample spiked with a known amount of the biosimilar antibody) is run on each ELISA plate to ensure that the assay performs as expected and to monitor intra- and inter-assay variability. This helps validate the accuracy and reproducibility of the quantified concentrations in actual patient serum samples.
ELISA Workflow in PK Studies:
Plates are coated with anti-idiotype or anti-human IgG capture antibodies, followed by addition of standards (biosimilar-spiked matrix), controls, and unknown samples.
Detection is typically via a secondary antibody conjugated to an enzyme, which produces a measurable signal proportional to the Epcoritamab concentration.
The measured absorbance of patient serum samples is compared to the standard curve generated with biosimilar standards, allowing for accurate quantitation of circulating drug levels in those samples.
Choice of Biosimilar Standard: Research-grade Epcoritamab biosimilars used for these purposes must be sourced with attention to purity, stability, and buffer compatibility, as these characteristics determine assay performance and reliability.
Reason for Use in Bridging ELISA: PK bridging ELISAs are often used when bioanalytical methods from different clinical phases or manufacturing campaigns must be correlated. The use of a consistent, characterized biosimilar as a calibration/reference bridges results across studies and manufacturing lots even when the originator drug is in limited supply.
Restrictions: Research-grade Epcoritamab biosimilars are for research use only and not for diagnostic or therapeutic use in humans.
In summary, biosimilars serve as critical assay standards and reference controls to provide accurate and reproducible quantitation of Epcoritamab concentrations in PK studies, underpinning data quality and comparability across trials and batches.
The primary in vivo models for administering a research-grade anti-CD3 x CD20 bispecific antibody (bsAb) to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models engineered to express human CD20 and/or humanized CD3, and to a lesser extent, humanized mouse models.
Key Models Used:
Syngeneic models with human CD3/CD20 knock-in: These are immunocompetent mice (often transgenic for human CD3 and/or CD20) implanted with murine tumor cells engineered to express human CD20. Such models allow the study of bsAbs requiring human CD3/CD20 recognition, providing an environment to analyze tumor growth inhibition and the composition/function of TILs. For example, studies using an A20-human CD20 mouse lymphoma model evaluate the efficacy of CD20-TDB (T-cell dependent bispecific) antibody and its ability to drive T-cell activation, B-cell depletion, and modulate TILs. Additionally, these models reveal pharmacodynamic immune responses, such as T-cell activation/interactions and the characteristic dynamics of TILs post-therapy.
Humanized mouse models: These involve immunodeficient mice engrafted with human immune cells (such as PBMCs, HSCs) and transplanted with human tumor xenografts expressing CD20. Humanized mice are particularly suited for studying TIL phenotype and function, as they provide a partially human immune microenvironment and support the evaluation of human-specific immune responses induced by anti-CD3 x CD20 bsAbs.
Model Characteristics and Rationale:
Syngeneic human CD20+ tumor models allow studying:
Tumor growth inhibition after bsAb administration.
Detailed analyses of TILs using flow cytometry and immunohistochemistry.
Immune checkpoint interactions by combining bsAb with agents such as PD-1/PD-L1 inhibitors, revealing synergistic effects on tumor growth and TIL modulation.
Humanized models are necessary when precise characterization of human T-cell activation, trafficking, and function is required, especially for dissecting mechanisms that may not fully recapitulate in syngeneic (mouse-origin) models.
These models are central for evaluating immune dynamics not easily observable in the peripheral blood, such as temporary T-cell sequestration in lymphoid organs, cytokine release syndrome, and direct anti-tumor efficacy driven by T-cell engagement.
Summary Table:
Model Type
Host Immune System
Tumor Origin
Utility for Anti-CD3 x CD20 bsAb Studies
Syngeneic (humanized CD3/CD20)
Mouse (genetically engineered)
Murine, expressing human CD20
Tumor inhibition, TIL characterization, immune kinetics
Humanized (PBMC/HSC engraftment)
Human-mouse hybrid
Human CD20+ xenografts
Human T-cell driven tumor rejection, TIL phenotype
Conclusion: Syngeneic mouse models expressing human CD20 (and/or humanized CD3) are the predominant and most thoroughly characterized system for in vivo administration of anti-CD3 x CD20 bsAbs to study both tumor inhibition and TIL composition; humanized mouse models are also used for mechanistic and translational insight into human immune responses.
Researchers investigate the synergistic effects of Epcoritamab (a CD20xCD3 bispecific antibody, often referred to as a "biosimilar" in this experimental context) with other checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 by designing combination studies, often in advanced preclinical models or early-phase clinical trials.
Research Strategies
In vitro assays: Scientists combine Epcoritamab (or its biosimilars) with checkpoint inhibitors in cell culture systems, typically using tumor cell lines and co-cultured immune cells, to assess cytotoxicity, T cell activation, and cytokine release. For example, CD20xCD3 bispecific antibodies like Epcoritamab show significant single-agent activity against lymphoma cell lines, and researchers compare multiple biosimilars to identify the most potent molecules and understand interactive mechanisms.
Animal (mouse) models: Immune-competent or humanized mice bearing complex tumors (e.g., melanoma or lymphoma) are treated with combinations of bispecific antibodies and checkpoint inhibitors. These models reveal how each agent modulates the immune system. In studies with other checkpoint inhibitor combinations (e.g., anti-PD-1 + anti-CTLA-4 or anti-PD-1 + anti-LAG-3), detailed immune profiling (e.g., T cell subset analysis, depletion experiments) was used to identify which immune cells are critical to therapy response and map out mechanism-driven synergy.
Immune monitoring: Researchers track changes in immune cell phenotypes, activation markers, cytokine production, and tumor microenvironment remodeling. For combinations involving CTLA-4 or LAG-3 inhibitors, mechanistic differences have been noted:
Anti-CTLA-4 combinations mainly bolster priming and expansion of new T cells in lymph nodes.
Anti-LAG-3 combinations tend to reduce regulatory T cells (Tregs) and enhance CD4 helper T cell functions, thereby boosting CD8 cytotoxic T cell responses.
Clinical translation: Early-phase trials combine bispecific antibodies with checkpoint inhibitors in patients to assess safety, tolerability, and evidence of synergy (e.g., improved response rates, durable remissions). For instance, CD20xCD3 antibodies like Epcoritamab are studied alone and, in principle, with other immunotherapies in relapsed/refractory B-cell lymphomas. The chief challenge is balancing enhanced anti-tumor activity with the increased risk of immune-related toxicities observed with combination regimens.
Key Insights from Related Combination Science
Distinct Mechanisms: Anti-CTLA-4 versus anti-LAG-3 combinations with other checkpoint inhibitors yield differing immune responses due to non-overlapping effects on immune cell subsets. These distinctions are important when formulating hypotheses about how a bispecific T-cell engager (like Epcoritamab) might synergize with specific checkpoint blockers in vivo.
Toxicity management: Synergy in immune activation often comes with increased immune-related adverse events; thus, preclinical synergy must be balanced with clinical safety and dosing strategies.
Translational models: Researchers often validate mechanistic findings in murine models before moving to in-human combination studies. Epcoritamab biosimilars, when available, would be expected to be deployed in this staged research fashion.
Limitations
No direct clinical or robust preclinical reports were found showing published results from Epcoritamab specifically combined with anti-CTLA-4 or anti-LAG-3 biosimilars. The synthesis here relies on the well-characterized approaches used in the field for similar immunotherapy combinations and mechanistic studies in immune-oncology.
In summary, researchers use in vitro, animal, and emerging clinical models to combine Epcoritamab biosimilars with checkpoint inhibitors, focusing on immune activation, mechanistic synergy, and toxicity, as established for other immune-oncology combinations.
A Epcoritamab biosimilar can be used as the capture or detection reagent in a bridging ADA (anti-drug antibody) ELISA assay to monitor a patient's immune response against the therapeutic drug by exploiting its structural similarity to the original therapeutic, allowing specific binding to ADAs directed against Epcoritamab.
In a bridging ADA ELISA, the key principle is that patient serum containing ADAs forms a bridge between two Epcoritamab molecules: one immobilized (capture) on the plate and one labeled (detection) for visualization. The general workflow is:
Capture reagent: The Epcoritamab biosimilar is coated onto the microtiter plate. When patient serum is added, any ADA present will bind to the immobilized biosimilar.
Detection reagent: After washing, a second, differently labeled Epcoritamab biosimilar is added. If the serum contains ADA, it acts as a bridge, binding both the capture and detection biosimilar molecules, enabling detection (usually via HRP- or biotin-labeling for chemiluminescent or colorimetric readout).
This "bridging" format:
Ensures specificity: Since both arms of the IgG ADA must bind to Epcoritamab, results are less influenced by non-specific interactions.
Detects all Ig classes: The method can detect any ADA isotype (IgG, IgM), as long as both antigen-binding sites are free to bind Epcoritamab.
Reduces interference from drug: Bridging assays are less sensitive to interference from circulating drug compared to competitive formats, though high levels of drug may still cause false negatives.
Why use a biosimilar as reagent?
Cost and availability: Biosimilars are more accessible for large-scale manufacturing and reagent supply than the originator.
Structural/epitope equivalence: Biosimilars closely match the originator antibody’s epitopes, so they efficiently capture and detect relevant ADAs.
Monitoring immunogenicity:
Using such an assay, clinicians can detect and quantify ADAs that may neutralize the therapeutic effect, anticipate loss of efficacy, guide drug switching, or adjust dosing.
Assay formats and advances:
Alternative affinity capture elution bridging assays can offer improved sensitivity and specificity as screening tools for patient ADA responses.
In summary, a Epcoritamab biosimilar functions as the antigenic bait in a bridging ELISA, enabling monitoring of immunogenicity by quantifying patient ADAs that recognize the therapeutic’s structure and may influence treatment outcomes.
References & Citations
1 Kang C. Drugs. 82(11):1229-1234. 2022.
2. Xiong D, Xu Y, Liu H, et al. Cancer Lett. 2002;177(1):29-39.
3. Chen Q, Yuan S, Sun H, Peng L. Hum Immunol. 2019;80(3):191-194.
4. Liu Y, Ao K, Bao F, et al. Vaccines (Basel). 2022;10(8):1335.
5. Izutsu K, Kumode T, Yuda J, et al. Cancer Sci. 2023;114(12):4643-4653.
6. Thieblemont C, Phillips T, Ghesquieres H, et al. J Clin Oncol. 2023;41(12):2238-2247.
7. Hutchings M, Mous R, Clausen MR, et al. Lancet. 2021;398(10306):1157-1169.