Anti-Human LAG-3 (Relatlimab) – Fc Muted™

Anti-Human LAG-3 (Relatlimab) – Fc Muted™

Product No.: L345

- -
- -
Product No.L345
Clone
BMS-986016
Target
LAG-3
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Lymphocyte-activation gene 3. CD223
Isotype
Human IgG4κ
Applications
ELISA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Human LAG-3
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
Additional Applications Reported In Literature ?
ELISA
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 Relatlimab. This product is for research use only. Relatlimab activity is directed against human LAG-3. Relatlimab also binds cynomolgus monkey LAG-3 but with lower affinity relative to the human homolog.
Background
LAG-3 (CD223) is an immune inhibitory receptor in activated T cells that inhibits T cell activation and proliferation, immune function, cytokine secretion, effector functions, and T cell homeostasis1. LAG-3 functions by down-modulating TCR:CD3 intercellular signal transduction cascades and calcium fluxes in the immunological synapse. LAG-3 inhibitory activities are mediated by its ligands: major histocompatibility complex class II, galectin-3, liver secreted protein fibrinogen-like protein 1, and DC-specific intercellular adhesion molecule-3-grabbing non-integrin family member. Some of these LAG-3 ligand combinations are responsible for tumor immune evasion mechanisms1 and LAG-3 is considered an aggressive progression marker in several hematological and solid tumor malignancies2. As such, LAG-3 is a target of cancer immunotherapy.

Relatlimab is the first anti-LAG-3 monoclonal antibody to be clinically developed1. It was generated using proprietary transgenic mice having human immunoglobulin miniloci in an endogenous IgH and Igκ knockout background3. The mice were immunized with recombinant human LAG-3-Fc protein, consisting of the LAG-3 extracellular domain (Leu23-Leu450) fused to the Fc portion of human IgG1. Hybridomas were generated by fusing spleen cells with P3×63Ag8.653 myeloma cells and screened for reactivity to hLAG-3-hFc. Clone 25F7 was chosen for grafting onto human κ and IgG4 constant region sequences, expressed in Chinese hamster ovary cells, and sequence optimized. The S228P stabilizing hinge was incorporated into the resulting antibody to prevent Fab-arm exchange. The binding epitope was experimentally determined to be in the N-terminal D1 insertion loop domain of LAG-3 within the H63-W70 peptide sequence.

Relatlimab binds to the LAG-3 receptor, blocking interaction with its ligands3 and consequently promotes T cell proliferation and cytokine secretion3, 4. Relatlimab depletes leukemic cells and restores T cell and NK cell-mediated immune responses in vitro5. Relatlimab has also been developed as a combination therapy with an anti-PD-1 antibody for increased T cell activation and anti-tumor effects4.
Antigen Distribution
LAG-3 is a surface molecule expressed by many T cell subsets (CD4 T helper cells, cytotoxic CD8 T cells, activated T cells, NK T cells, effector CD4 T cells, regulatory T cells, CD8 tumor-infiltrating lymphocytes, and tumor infiltrating antigen specific CD8 T cells) as well as by natural killer cells, B cells, natural regulatory plasma cells, plasmacytoid dendritic cells, and neurons.
Ligand/Receptor
MHCII & Fibrinogen-like protein1 FGL1
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Immunology

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.

Use of Research-Grade Relatimab Biosimilars in PK Bridging ELISAs

Research-grade biosimilars such as Relatimab (a proposed biosimilar to rituximab or a similar monoclonal antibody) may be employed as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs for quantifying drug concentration in serum samples, but this use is governed by strict scientific and regulatory principles to ensure accurate and comparable measurements.

Calibration Standards in PK Assays

  • Single Standard Approach: For PK assays comparing a biosimilar and its reference product, the current industry best practice is to use a single analytical standard—often the biosimilar itself—as the assay calibrator for quantitating both the biosimilar and reference product in test samples. This minimizes variability that could arise from using separate calibration curves for each product, and eliminates the need for crossover analysis in blinded clinical studies.
  • Standard Preparation and Validation: The biosimilar standard is carefully prepared at known concentrations (e.g., 50–12,800 ng/mL) in human serum to generate a calibration curve. During method validation, both biosimilar and reference products are independently prepared at multiple concentrations and quantified against this standard curve to assess bioanalytical comparability. If the biosimilar and reference product demonstrate equivalent behavior (i.e., similar precision, accuracy, and robustness) within the assay, the single standard approach is validated.
  • Data Interpretation: The equivalence of the biosimilar and reference product within the assay is determined by comparing their performance against predefined equivalence criteria, such as 90% confidence intervals for bioanalytical equivalence. If equivalence is established, the biosimilar can be confidently used as the standard for PK measurements in both types of samples.

Reference Controls in Quality Assurance

  • Quality Controls: After establishing bioanalytical comparability, the biosimilar is also used to prepare quality control (QC) samples in human serum at defined concentrations. These QCs are included in each assay run to ensure ongoing assay performance and reliability when measuring both biosimilar and reference product concentrations.
  • Robustness and Harmonization: When a biosimilar is used as a standard, it is critical that the assay (in this case, a bridging ELISA) is fully validated according to regulatory guidelines, including assessments of precision, accuracy, specificity, and robustness. This ensures that concentration data are reliable and suitable for regulatory submission.

Regulatory and Scientific Considerations

  • Biosimilarity Assessment: The use of a biosimilar as a standard in PK assays is only appropriate after rigorous analytical and clinical demonstration of similarity to the reference product. This includes comparative analysis of critical quality attributes using advanced analytical techniques (e.g., mass spectrometry), ensuring that the biosimilar’s structural and functional characteristics closely match those of the reference.
  • Harmonization: Industry groups and regulatory agencies are harmonizing best practices for biosimilar standards and analytical methods to ensure consistency and reliability across laboratories.

Summary Table: Role of Biosimilar Standards in PK Bridging ELISA

ApplicationPurposeImplementation in PK Bridging ELISA
Calibration StandardGenerate standard curve for quantitationBiosimilar prepared at known concentrations; used to measure both biosimilar and reference in serum
Reference Control (QC)Monitor assay performance and consistencyBiosimilar QC samples included in each assay run; ensures reliability
Method ValidationConfirm assay suitability for both productsBoth products tested against biosimilar standard; equivalence established statistically
Regulatory ComplianceSupport PK similarity and biosimilarityAssay validated per regulatory guidelines; data used for approval

Key Points

  • Research-grade biosimilars can serve as calibration standards and reference controls in PK bridging ELISAs, provided they are analytically equivalent to the reference product.
  • A single calibration curve derived from the biosimilar is used to quantify both the biosimilar and reference product in serum samples, reducing assay variability.
  • Comprehensive validation and equivalence testing are essential to ensure the biosimilar standard yields accurate and comparable PK data for regulatory submissions.
  • Ongoing use of biosimilar-based QC samples ensures assay robustness and reliability.

This approach is now a best practice in biosimilar development, supported by regulatory guidance and harmonized industry standards.

Anti-LAG-3 antibodies are primarily tested for tumor growth inhibition and TIL characterization in two main in vivo model systems: syngeneic mouse models (including genetically engineered mice and transplanted tumor lines), and humanized/transgenic mouse models expressing human LAG-3.

Essential context and supporting details:

  • Syngeneic Mouse Models:

    • These use mouse-derived tumor cell lines transplanted into immunocompetent mice of the same genetic background, allowing a native mouse immune system interaction with tumors.
    • Anti-LAG-3 antibodies and combination therapies (commonly with anti-PD-1) have been thoroughly evaluated here for their impact on tumor growth and TIL function. Murine tumors with high LAG-3+ TIL populations ("LAG-3^hi") are especially used to study the efficacy and mechanisms of dual checkpoint blockade.
    • These models help characterize the activity and phenotype of CD8+ T cells, regulatory T cells (Tregs), and their plasticity in response to therapy. Specialized reporter mice (e.g., Foxp3 fate-tracking models) allow in-depth analysis of TIL subpopulations.
  • Humanized/Transgenic Models:

    • For research-grade antibodies that specifically recognize human LAG-3, transgenic mice expressing human LAG-3 are used so that the antibody will bind and exert its effect in vivo.
    • These models often combine human LAG-3 transgene with mouse tumors and anti-mouse checkpoint molecules, facilitating direct testing of fully human anti-LAG-3 antibodies (such as LBL-007).
    • These platforms are crucial for assessing the translational relevance of human-specific antibodies before clinical testing.

TIL Characterization Approaches:

  • After antibody administration, tumor-infiltrating lymphocytes are characterized by flow cytometry, mass cytometry, and single-cell RNA sequencing, enabling detailed profiles of CD8+ and CD4+ T cells, Tregs, and other immune subsets.
  • LAG-3 is typically co-expressed with PD-1 in dysfunctional or "exhausted" CD8+ T cells within tumors; therapies aim to reinvigorate these cells and reduce suppressive Treg functions.
  • The mechanistic studies focus on changes in TIL frequency, activation status, cytokine production, cytotoxicity, and phenotypic plasticity after treatment.

Model system summary:

Model TypeAntibody UsedTumor SystemPrimary Purpose
Syngeneic mouse (e.g., LAG-3^hi)anti-mouse LAG-3Murine tumorsTumor inhibition; TIL mechanism
Humanized/transgenic mouseanti-human LAG-3Murine tumors + human LAG-3Translational antibody testing; TIL characterization

In conclusion, both syngeneic and humanized (transgenic) models are foundational for in vivo testing of anti-LAG-3 antibodies, with syngeneic models broad for mechanistic studies and humanized models key for translational research using human-targeted reagents.

Use of Relatlimab Biosimilars in Synergistic Immune-Oncology Research

Relatlimab is a monoclonal antibody targeting the lymphocyte-activation gene 3 (LAG-3), an immune checkpoint receptor that suppresses T-cell activity within the tumor microenvironment. Biosimilars of Relatlimab, while not approved for clinical use, are valuable research tools for studying the mechanisms of LAG-3 inhibition and its interplay with other checkpoint pathways in complex immune-oncology models.

Mechanism of Synergy: LAG-3 and Other Checkpoint Inhibitors

LAG-3 inhibitors like Relatlimab (or its biosimilars) are often evaluated in combination with other checkpoint inhibitors—such as anti-PD-1 (e.g., nivolumab) or anti-CTLA-4 (e.g., ipilimumab)—to assess synergistic effects. The rationale is that blocking multiple immune checkpoints can more effectively reverse T-cell exhaustion and reactivate antitumor immunity, potentially overcoming resistance seen with single-agent therapies. Clinical studies have shown that dual inhibition of LAG-3 and PD-1, for example, leads to enhanced T-cell activation and improved clinical outcomes in melanoma compared to monotherapy.

Research Applications in Complex Models

Researchers use Relatlimab biosimilars in preclinical studies to:

  • Evaluate Combination Efficacy: Test the enhanced antitumor activity when LAG-3 inhibition is combined with anti-PD-1, anti-CTLA-4, or other immune checkpoint blockers in various cancer models (e.g., melanoma, non-small cell lung cancer, solid tumors).
  • Explore Resistance Mechanisms: Investigate how tumors develop resistance to single-agent checkpoint blockade and whether dual or triple checkpoint inhibition can overcome this resistance.
  • Optimize Treatment Strategies: Screen for optimal dosing, timing, and sequence of administration to maximize synergy and minimize toxicity in complex in vitro and in vivo models.
  • Expand Biomarker Discovery: Identify predictive biomarkers of response and resistance to guide patient selection for future clinical trials.
  • Cost-Effective Screening: Use biosimilars for high-throughput screening and mechanistic studies, enabling broader exploration of combination therapies without the expense of proprietary drugs.

Advantages of Biosimilars in Research

Relatlimab biosimilars offer several practical benefits for immune-oncology research:

  • Accessibility: Facilitate widespread use in academic and industry labs for mechanistic and combination therapy studies.
  • Scalability: Support large-scale preclinical evaluations and combination screening across diverse tumor models.
  • Cost Efficiency: Reduce research costs, allowing more resources to be allocated to exploring novel combinations and treatment regimens.

Challenges and Future Directions

While biosimilars enable robust preclinical research, they are not approved for clinical use and must be validated for equivalence to the reference biologic. Researchers must also consider the complexity of immune interactions, potential overlapping toxicities, and the need for careful patient selection when translating findings to clinical trials. Ongoing studies continue to explore the full potential of Relatlimab (and its biosimilars) in combination with other checkpoint inhibitors, aiming to refine treatment paradigms for advanced and refractory cancers.

Summary Table: Key Research Applications

Application AreaRelatlimab Biosimilar UseSynergy Target(s)Model Type
Combination EfficacyDual/triple checkpoint blockadeAnti-PD-1, Anti-CTLA-4, etc.In vitro, in vivo tumors
Resistance MechanismsOvercoming immune evasionMultiple checkpointsResistant tumor models
Treatment OptimizationDosing, timing, sequence studiesVarious ICIsPreclinical screens
Biomarker DiscoveryIdentifying response predictorsN/APatient-derived models
Cost-Effective ScreeningHigh-throughput assaysN/ACell lines, organoids

Conclusion

Relatlimab biosimilars are instrumental in preclinical immune-oncology research, enabling detailed investigation of synergistic effects when combined with other checkpoint inhibitors. These studies aim to enhance antitumor immunity, overcome resistance, and optimize combination strategies—ultimately informing the design of next-generation cancer immunotherapies.

A Relatimab biosimilar can be used as the capture or detection reagent in a bridging ADA ELISA to monitor a patient's immune response against the therapeutic drug by leveraging its identical protein sequence to the original relatlimab, enabling specific detection of anti-relatimab antibodies in patient samples.

In a bridging ADA ELISA for immunogenicity testing:

  • Capture reagent: The relatimab biosimilar (matching the original therapeutic antibody’s sequence and binding characteristics) is immobilized on a solid phase, such as a microtiter plate or via biotinylation onto streptavidin-coated wells.
  • Patient sample addition: Serum or plasma from the patient, potentially containing anti-drug antibodies (ADAs) formed as an immune response against relatimab, is incubated with the immobilized biosimilar.
  • Detection reagent: A second, labeled form of the relatimab biosimilar (e.g., HRP-conjugated or biotinylated) is introduced. If bivalent ADAs are present, they will bridge between the immobilized and detection relatimab biosimilar, forming a detectable complex.
  • Signal generation: The detection antibody's label (usually HRP) is developed with a substrate (e.g., TMB), resulting in a measurable color change proportional to ADA concentration.

Key technical considerations:

  • The use of a biosimilar version ensures the assay detects immune responses against all epitopes presented by the therapeutic relatimab.
  • The bridging format is sensitive and requires ADAs to be bivalent (able to bind two copies of the drug simultaneously), which minimizes false positives from monovalent interactions.
  • High-purity, research-grade relatimab biosimilar (e.g., human IgG4(S228P)-Kappa, >95% purity) is required for consistent assay performance.
  • Matrix effects from serum components, or soluble LAG-3, can reduce specificity and may require blocking steps or assay optimization.

Additional context:

  • The same approach and principles are widely used for monitoring immunogenicity of other mAb-based biologics.
  • Assay customization may be necessary depending on the biosimilar preparation, patient population, and clinical requirements.

In summary, a relatimab biosimilar serves as both the capture and detection molecule in the bridging ADA ELISA, allowing sensitive and specific measurement of immune responses to the therapeutic, which is crucial for evaluating clinical safety and efficacy.

References & Citations

1. Chocarro L, Bocanegra A, Blanco E, et al. Cells. 11(15):2351. 2022.
2. Long L, Zhang X, Chen F, et al. Genes Cancer. 9(5-6):176-189. 2018.
3. Thudium K, Selby M, Zorn JA, et al. Cancer Immunol Res. 10(10):1175-1189. 2022.
4. Paik J. Drugs. 82(8):925-931. 2022.
5. Sordo-Bahamonde C, Lorenzo-Herrero S, González-Rodríguez AP, et al. Cancers (Basel). 13(9):2112. 2021.
Indirect Elisa Protocol

Certificate of Analysis

Formats Available

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