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 asthe therapeutic antibody Infliximab. This product is research use only. ABP-710 is an infliximab biosimilar targeting tumornecrosis factor alpha (TNFα). ABP-710 binding and effector functions are similar to infliximabin vitro. ABP-710 binds to both soluble and membrane-bound TNFα.
Background
TNFα is a 17.5 kD protein that mediates inflammation and immunity caused by the invasion of viruses, bacteria, and parasites by initiating a cascade of cytokines that increase vascular permeability, thus bringing macrophages and neutrophils to the site of infection. TNFα secreted
by macrophages cause the blood to clot which provides containment of an infection. TNFα is also associated with autoimmune diseases and its inactivation is important in downregulating inflammatory reactions associated with rheumatoid arthritis, psoriatic arthritis, ankylosing
spondylitis, Crohn's disease, moderate to severe chronic psoriasis, and juvenile idiopathic arthritis.
Infliximab is used in the clinical setting to treat immune-mediated inflammatory disorders and is a particularly efficacious treatment for inflammatory bowel disease1. Infliximab, and its biosimilar ABP-710, suppresses inflammation primarily by binding and neutralizing sTNFα2.
ABP-710 and infliximab are analytically similar regarding amino acid sequence, primary peptide structure, secondary structure, tertiary structure, conformation, thermal stability, and glycan mapping1,2.
ABP-710 and infliximab biological functions are also similar2. Like infliximab, ABP-710 inhibits sTNFα-induced apoptosis and mediates effector functions such as antibody-dependent cell-mediated cytotoxicity, antibody-dependent cellular phagocytosis, and complement-dependent cytotoxicity when interacting with membrane bound TNFα.
Antigen Distribution
TNFα is secreted by macrophages, monocytes, neutrophils, T cells, B
cells, NK cells, and LAK cells.
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 Infliximab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays to enable accurate and interchangeable measurement of infliximab concentrations in serum, regardless of the specific product (originator or biosimilar) administered.
In PK bridging ELISA for infliximab, the main steps involving biosimilars as standards are:
Calibration Curve Preparation: The biosimilar is serially diluted in a matrix (often serum or buffer that mimics matrix effects) to generate calibration standards at known concentrations. These standards establish the calibration curve against which unknown sample concentrations are assessed.
Reference Standard Interchangeability: Studies report that both originator infliximab (Remicade) and its biosimilars (e.g., Remsima) can be used interchangeably for assay calibration in ELISA and LC-MS/MS-based bioanalytical methods. Cross-verification shows nearly identical quantitation across products, supporting their use as reference controls in clinical PK assays. This interchangeability is essential for therapeutic drug monitoring in patients treated with either the originator or biosimilar.
Assay Design: In the bridging ELISA, the solid phase is typically coated with TNFα, which specifically binds infliximab (originator or biosimilar) from patient serum. Detection is achieved using anti-human IgG or other suitable antibody conjugates. The measured signal (e.g., absorbance) from patient samples is then compared to the calibration curve generated from the biosimilar-reference standards to quantify drug levels.
Control and Quality Verification: Biosimilars are used as positive controls to validate assay performance in each run. Their concentration and recovery help ensure robustness and reproducibility of results.
Key Supporting Insights
Robust Quantitation: Biosimilars show linear, accurate concentration-response in calibration curves (e.g., 0.293 to 300 µg/ml), with high precision in quantitation matching the originator performance.
Assay Harmonization: The use of biosimilar standards facilitates harmonization and mutual comparability between different assay platforms and labs, addressing differences in TNFα coating, immune complex formation, and potential for interference from anti-drug antibodies.
Clinical Relevance: Dose adjustment and therapy decisions typically rely on trough infliximab levels (often <3–5 µg/ml), which can be accurately determined using biosimilar calibration curves.
In summary, research-grade infliximab biosimilars are pivotal for preparing calibration standards and reference controls in PK bridging ELISA assays, ensuring reliable quantitation of drug concentration in serum samples and facilitating consistent clinical monitoring whether patients receive originator or biosimilar therapy.
The primary research models where a research-grade anti-TNF-α antibody is administered in vivo to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are mouse syngeneic tumor models and, less commonly, humanized mouse models.
Key context and details:
Syngeneic Mouse Models: These models use mouse tumor cell lines grafted into immunocompetent mice of matching strains, such as CT26 colon carcinoma in BALB/c mice. Anti-TNF-α antibodies are administered to evaluate tumor growth inhibition and the immune microenvironment, allowing analysis of TILs using immunohistochemistry and flow cytometry.
For example, anti-TNF-α mAb was administered in a CT26 orthotopic colorectal cancer model (BALB/c mice), with characterization of immune infiltrates, apoptosis, and stromal responses.
Other frequent tumor cell lines used include B16F10 (melanoma), RENCA (renal carcinoma), and 4T1 (breast cancer).
These models support detailed TIL characterization, benefiting from fully functioning mouse immune systems.
Humanized Mouse Models (less common specifically for anti-TNF-α studies): Humanized mice are immunodeficient strains engrafted with human immune cells and/or human tumors. While most anti-TNF-α studies use syngeneic models, humanized models are sometimes used to study the effects of anti-TNF-α antibody in a setting that more closely mimics the human immune-tumor interaction, including assessments of TILs.
Experimental Workflow in Syngeneic Models:
Tumor cells are implanted (subcutaneous or orthotopic) into inbred mice.
Anti-TNF-α antibody is administered systemically.
Tumor growth rates are measured, and tumors are harvested for immunohistochemical and flow cytometric analysis of TILs, such as CD8+ and CD4+ T cells, myeloid populations, and NK cells.
The effect on apoptosis, stromal response, and angiogenesis is often assessed in parallel.
Humanized Models: These models can allow the evaluation of human-specific antibodies and the study of human TILs but are less commonly used for anti-TNF-α preclinical research compared to syngeneic models. They are particularly relevant when the antibody does not cross-react with mouse TNF-α.
Summary Table:
Model Type
Immune System
Tumor Source
TIL Characterization
Use with Anti-TNF-α
Syngeneic
Mouse (intact)
Mouse cell lines
Mouse TILs
Most common; robust, well-characterized
Humanized
Humanized (reconstituted)
Human cell lines
Human TILs
Useful for human-specific antibodies; less common
Most studies, including those exploring anti-TNF-α effects on tumor immunity and TILs, rely on syngeneic mouse models such as CT26 in BALB/c or B16F10 in C57BL/6.
If you require protocols or detailed experimental setups, please specify the tumor type or antibody species (murine, human, chimeric).
Researchers use infliximab biosimilars in combination with checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) to investigate their potential synergistic effects in complex immune-oncology models, particularly by examining how TNF-α blockade can modulate both therapeutic efficacy and immune-mediated toxicity (irAEs).
Key points and supporting details:
Model Rationale: Infliximab (and its biosimilars) targets TNF-α, a central cytokine in inflammation that is also involved in immune therapy-associated adverse events (irAEs) and tumor biology. Preclinical and translational studies have shown that TNF-α blockade can improve the safety profile of immune checkpoint inhibitor (ICI) therapies (e.g., anti-CTLA-4, anti-PD-1/PD-L1), resolving irAEs without significantly compromising the antitumor immune response.
Combination and Mechanisms Studied: Researchers combine infliximab biosimilars with checkpoint inhibitors in animal models (like mouse melanoma or colon cancer models) to examine:
Whether TNF-α inhibition can reduce ICI-induced adverse effects (notably colitis and other autoimmune toxicity).
Whether TNF-α blockade permits or potentiates stronger antitumor T cell infiltration and improved tumor regression when combined with ICIs, enhancing overall efficacy.
The molecular and cellular mechanisms underlying any observed synergy or antagonism, such as changes in T cell activation, apoptosis, cytokine profiles, and tumor immune microenvironment composition.
Role of Biosimilars: Using biosimilars (like ABP 710) provides functionally equivalent substitutes for the originator infliximab, meeting rigorous comparability standards—making them suitable for both preclinical synergy studies and for expanding availability in translational research or clinical trials.
Checkpoint Inhibitor Combinations: Synergistic effects are of particular interest with anti-CTLA-4 or anti-LAG-3 biosimilars, as these enhance T cell responses but may also cause off-target inflammation. The combined administration helps distinguish mechanisms of tumor immunity from those of autoimmunity, allowing researchers to optimize dosing or sequencing to balance efficacy and safety.
Observed Outcomes: In mouse studies, the combination of TNF-α blockade (via infliximab or biosimilar) with anti-CTLA-4/anti-PD-1 therapy led to better tumor control and lower rates of severe immune adverse events. These results have prompted retrospective and prospective studies in humans to further delineate safety and potential augmentation of antitumor effects.
Immunogenicity Considerations: Combinatorial approaches may also affect the immunogenicity (anti-drug antibody formation) of protein therapeutics, an important parameter that is actively monitored in these studies.
In summary, infliximab biosimilars are combined with checkpoint inhibitors in immune-oncology models to study how TNF-α blockade modulates both therapeutic efficacy and toxicity, providing critical insights into the mechanisms of synergy, with significant translational potential for optimizing cancer immunotherapy regimens.
In a bridging anti-drug antibody (ADA) ELISA for monitoring a patient's immune response to infliximab, an infliximab biosimilar can serve as both the capture and detection reagent to identify anti-infliximab antibodies in patient samples.
How Infliximab Biosimilar Is Used:
Capture Reagent: The ELISA plate is coated with the infliximab biosimilar, which acts as the antigen to capture any anti-infliximab antibodies (ADA) present in the patient's serum or plasma.
Detection Reagent: After washing, a detection reagent—typically the same or a labeled form of infliximab biosimilar (e.g., biotinylated, enzyme-, or ruthenium-labeled)—is added. This tagged infliximab binds to another epitope on the ADA, forming a "bridge" between the plate-bound and the detection molecule, because each ADA is bivalent and can bind two infliximab molecules.
Assay Principle (Bridging ELISA Format):
Coating: Wells are coated with the infliximab biosimilar.
Sample Addition: Patient samples are added. If anti-infliximab antibodies are present, they will bind to the immobilized infliximab.
Detection: Labeled (e.g., biotinylated or enzyme-conjugated) infliximab biosimilar is added. This binds to the other arm of the ADA, creating a "bridge" structure.
Signal Generation: A secondary reagent (e.g., streptavidin-HRP if the detection antibody is biotinylated, or detection of enzyme activity) is added to generate a measurable signal proportional to the amount of ADA present.
Step
Purpose
Plate coating
Capture anti-infliximab antibodies
Patient sample
Potential source of anti-infliximab antibodies
Detection agent
Binds ADA, completes bridge, allows signal readout
Why Use a Biosimilar?
Biosimilars are structurally and functionally highly similar to the originator infliximab and have the same epitopes recognized by ADA, making them suitable for use as capture and detection reagents in the assay.
Application:
This method specifically detects antibodies that bind infliximab (including both originator and biosimilar versions) and is widely used to monitor immunogenicity, which can affect drug efficacy and safety in patients receiving infliximab therapy.
Important Considerations:
Bridging ELISAs require bivalent antibodies, so they may not detect monovalent anti-drug antibodies.
Free (circulating) infliximab in patient serum can interfere with ADA detection, so samples may need to be taken at trough drug levels or use drug-tolerant ADA assays.
Summary: An infliximab biosimilar, properly labeled, is used both to coat assay plates (capture) and as a detection reagent in a bridging ADA ELISA, enabling sensitive monitoring of anti-infliximab immune responses in patients.
References & Citations
1 Reinisch W, Cohen S, Ramchandani M, et al. Adv Ther. 39(1):44-57. 2022.
2 Saleem R, Cantin G, Wikström M, et al. Pharm Res. 37:114. 2020
3 Genovese MC, Sanchez-Burson J, Oh M, et al. Arthritis Res Ther. 22(1):60. 2020.
4 Chow V, Oh M, Gessner MA, et al. Clin Pharmacol Drug Dev. 9(2):246-255. 2020.
5 Lee YH, Song GG. Z Rheumatol. 82(2):114-122. English. 2023.