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 Evinacumab. Evinacumab is designed to specifically target
Angiopoietin-like protein 3 (ANGPTL3).
Background
Angiopoietin-like protein 3 (ANGPTL3) is a glycoprotein primarily produced in the
liver and plays a crucial role in lipid metabolism. It hinders the function of lipoprotein
lipase (LPL) and endothelial lipase (EL) which are essential for breaking down
triglycerides and phospholipids. By blocking these enzymes ANGPTL3 increases the
levels of triglycerides LDL cholesterol and HDL cholesterol in the blood. Research
has shown that mutations in ANGPTL3 that result in reduced function are associated
with lower lipid levels and a reduced risk of coronary artery disease1-3.
The monoclonal antibody REGN 1500, also known as evinacumab, targets
ANGPTL3 to treat hypercholesterolemia (HoFH) a rare genetic condition
characterized by extremely high cholesterol levels. Evinacumab works by inhibiting
ANGPTL3, which helps break down fats and leads to a decrease in low-density
lipoprotein cholesterol (LDL-C). Clinical trials have proven that evinacumab can
reduce LDL-C levels by around 47% in patients, with HoFH4-7.
Antigen Distribution
ANGPTL3 is found primarily in the liver; however, it is also expressed in
other tissues, such as adipose tissue and podocytes in the kidney.
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 Evinacumab biosimilars are used as analytical standards or reference controls in pharmacokinetic (PK) bridging ELISAs to ensure accurate and comparable quantitation of drug concentrations in serum samples.
In a PK bridging ELISA designed to measure concentrations of Evinacumab (a monoclonal antibody) in serum, it is critical to use a well-characterized standard to generate the calibration curve against which unknown samples are quantified. Here’s how research-grade Evinacumab biosimilars are employed in this context:
Calibration Standard: A single lot of Evinacumab biosimilar is often selected as the analytical standard for constructing the standard curve in the ELISA. This standard is serially diluted in serum to cover the relevant concentration range (for example, 50 to 12,800 ng/mL, depending on assay sensitivity and therapeutic window).
Reference Control/Quality Controls (QCs): Separate aliquots of the biosimilar, and often of the reference (originator) product, are prepared at specific concentrations (e.g., low, medium, high QCs) and included in each plate or run. These controls verify assay accuracy, precision, and consistency throughout the study.
Assay Equivalency: Before adopting a biosimilar as the sole standard, the laboratory establishes bioanalytical comparability, confirming the biosimilar and the reference product are measured equivalently by the ELISA (i.e., the assay is not biased toward one molecular form over another). This is typically done by spiking known amounts of each into serum, quantifying them against the chosen standard, and showing comparable recovery and precision across products.
PK Bridging Function: Using the biosimilar as a single standard eliminates variability that could be introduced if separate calibration curves were constructed for biosimilar versus originator drug, meeting regulatory and scientific expectations for robust PK comparability assessments.
Result Expression: Serum samples from preclinical or clinical PK studies are quantitated with this calibration curve and reported as concentrations of Evinacumab-equivalent (often in units such as ng/mL), facilitating cross-product bridging analysis and regulatory acceptance.
Key steps in practice:
Prepare a standard curve from the biosimilar Evinacumab, diluted in blank serum.
Prepare QC samples from both biosimilar and, if required, reference (originator) product at several points across the dynamic range.
Assess recovery, accuracy, and precision of both products using the biosimilar standard curve.
If bioanalytical equivalency is demonstrated, proceed to use the biosimilar as the single calibration standard for all PK samples.
This strategy is widely recommended to reduce analytical variability, avoid cross-over studies, and assure regulatory compliance in biosimilar development PK programs.
Note: There may be minor, product-specific protocol optimizations depending on the drug’s matrix effects, immunogenicity, or ELISA configuration, but the above process reflects current best practices as described in recent regulatory and scientific literature.
If you require a specific validation protocol or technical SOP for Evinacumab, those typically follow the general schematic above but may be customized for the molecule’s biochemical specifics.
The primary preclinical models used to study in vivo administration of a research-grade anti-ANGPTL3 antibody for tumor growth inhibition and characterization of tumor-infiltrating lymphocytes (TILs) are murine syngeneic tumor models. There is little evidence from the literature that fully humanized models or patient-derived xenografts have been systematically used for this purpose.
Essential context and supporting details:
Syngeneic mouse tumor models—where mouse tumor cells are implanted into immunocompetent mice of the same genetic background—are the platform of choice for immunotherapy studies that assess both tumor growth and changes in the immune cell populations within the tumor microenvironment, including TILs.
These models allow detailed immunophenotyping (e.g., via flow cytometry or single-cell sequencing) of TIL populations after in vivo treatment with an antibody, and proper exploration of the mechanism of action for candidate immunotherapeutics in an immune-competent setting.
Commonly used syngeneic tumor models include:
MC38 (colon carcinoma)
RENCA (renal cell carcinoma)
CT26 (colon carcinoma)
B16F10 (melanoma)
4T1 (breast cancer)
These models represent a spectrum from highly immune-infiltrated (e.g., RENCA) to poorly infiltrated (e.g., B16F10).
Studies have reported the characterization of TILs such as CD8+ T cells, Tregs, NK cells, and myeloid subsets post-treatment with immunotherapies in these syngeneic settings.
Humanized mouse models, where mice are engrafted with a human immune system, are useful for testing human-specific reagents or antibodies. However, there is no direct evidence that anti-ANGPTL3 antibodies have been systematically tested in humanized models for tumor immunology to date, likely due to technical complexity and the more mature immune context available in syngeneic models for mechanistic studies.
Studies of ANGPTL3 blockade in vivo, including those using anti-ANGPTL3 or anti-ANGPTL3/8 antibodies, have primarily been conducted for metabolic disease in hypertriglyceridemic mice and not to evaluate tumor growth or TILs explicitly.
When anti-ANGPTL3/IL22 bifunctional fusion proteins are used in mice, these are tested for biological effects relevant to inflammation and tissue injury (e.g., diabetic nephropathy models), but assessment of tumor growth or TIL composition is not described in these studies.
Summary Table: Model Types Used for In Vivo Anti-ANGPTL3 Tumor Studies
Model Type
Immune Competence
TIL Profiling Feasibility
Reports with Anti-ANGPTL3/Tumor/TILs
Murine syngeneic tumor models
Yes
High
Preferred/standard (general)
Humanized mouse tumor models
Partial
Medium
No direct evidence
Patient-derived xenograft (PDX)
No (immunodeficient)
Low/absent
Not applicable
Genetically engineered mouse tumor models (GEMM)
Yes
High
Rare/No evidence for ANGPTL3
Key Points:
Syngeneic models are the most widely used and well-characterized systems for evaluating in vivo tumor inhibition and TILs with experimental antibodies targeting tumor-associated or immune-related proteins.
There is no published evidence of humanized models being systematically used for anti-ANGPTL3 antibody studies in the tumor immunology setting.
Published studies of anti-ANGPTL3 antibodies focus on metabolic and vascular endpoints in normal mice, not tumor inhibition or TILs.
If your goal is to study anti-ANGPTL3 effects in cancer immunology, syngeneic models (e.g., MC38, RENCA, B16F10, CT26) will provide the best-established system for examining both tumor growth and the immune microenvironment, including TIL dynamics.
Researchers studying synergistic effects in complex immune-oncology models often combine biosimilars of immune checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 with other agents, including angiogenesis inhibitors like bevacizumab biosimilars, though specific literature on evinacumab biosimilars in such combinations is limited. The general strategy involves evaluating whether dual or multiple checkpoint blockade enhances antitumor immune responses beyond what is achieved with single agents.
Key context and supporting details:
Checkpoint inhibitor combinations: Research has shown that combining drugs targeting PD-1/PD-L1 with those targeting CTLA-4 or LAG-3 increases immune activation and antitumor effects in advanced cancers such as melanoma. Synergy is assessed by comparing combination regimens to monotherapy in terms of objective response rates (ORR), progression-free survival (PFS), and overall survival (OS).
Mechanisms of synergy: Combinations of checkpoint inhibitors work via distinct but complementary mechanisms. For example, anti-CTLA-4 inhibits regulatory T-cell suppression at the priming phase, while anti-LAG-3 may boost effector T-cell activity or target different cell populations. These mechanistic differences can be leveraged to overcome resistance and achieve broader, more durable tumor control.
Role of biosimilars: Biosimilars are used to replicate the clinical and immunological profiles of original monoclonal antibodies, facilitating comparative and combination studies while potentially lowering costs. For instance, bevacizumab biosimilars are commonly combined with other immunotherapies and chemotherapies to evaluate additive or synergistic effects on immunogenicity and efficacy. Trials with LAG-3 antagonists have combined them with pembrolizumab to potentiate immune activation in patients with melanoma who are refractory to PD-1 therapy.
Experimental design in immune-oncology models:
Researchers use in vitro co-culture assays, murine tumor models, and humanized mouse models to test checkpoint and angiogenesis inhibitor combinations.
Endpoints include tumor size reduction, immune cell infiltration, cytokine production, and survival.
Efficacy and safety assessments involve not only tumor response but also analysis of immunogenicity, adverse events (such as injection site reactions or immune-related toxicity), and pharmacokinetics.
Additional relevant information:
There is robust clinical precedent for combining checkpoint inhibitors, but published combinations with evinacumab or true biosimilars of this agent are scarce. The principles and methods for evaluating synergy remain consistent across monoclonal antibodies addressing different targets.
Optimization of outcomes depends on selecting combinations that address distinct immune escape mechanisms and testing in relevant tumor models.
If you require details specific to evinacumab biosimilars (an ANGPTL3 inhibitor typically used in lipid disorders) within immune-oncology studies, such literature is currently not represented in the search results. Most combination studies focus on checkpoint blockade agents and angiogenesis inhibitors rather than lipid-modulating antibodies like evinacumab.
A Evinacumab biosimilar can be used as either the capture or detection reagent in a bridging ADA ELISA to monitor a patient's immune response by specifically binding anti-drug antibodies (ADA) that a patient may develop against the therapeutic drug (evinacumab).
How the bridging ADA ELISA works with a Evinacumab biosimilar:
Format: In a typical bridging assay, the same molecule (here, the Evinacumab biosimilar) is used in two forms: one immobilized (capture reagent), and one labeled—for example, with a detection tag such as horseradish peroxidase (HRP), biotin, or another appropriate label (detection reagent).
Sample incubation: Patient serum (which may contain ADA) is incubated with both the labeled and unlabeled Evinacumab biosimilar.
Immune bridging: If ADA is present, it bridges (binds) between the capture Evinacumab biosimilar on the plate and the labeled Evinacumab biosimilar, forming a "sandwich" complex via its bivalent binding.
Detection: After washing steps to remove unbound components, a signal is generated from the detection label, which is proportional to the amount of ADA present in the patient sample.
Why use a biosimilar?
Specificity: The Evinacumab biosimilar has the same antigenic epitopes as the therapeutic drug, ensuring it can capture the specific ADA that would react with the clinical product.
Reproducibility: Biosimilars are used when the original drug is not available, for research, or to standardize immunogenicity assays without using precious clinical-grade material.
Example bridging ELISA setup with Evinacumab biosimilar:
Coating: Immobilize biotinylated Evinacumab biosimilar on a streptavidin-coated ELISA plate.
Incubation: Add patient serum and HRP-conjugated Evinacumab biosimilar to the wells.
Bridging: If ADA is present, it will bind both the immobilized and HRP-labeled Evinacumab biosimilar, forming a bridge.
Detection: Develop with appropriate substrate and measure signal (e.g., colorimetric or chemiluminescent).
Key technical points:
Choose high-purity, well-characterized biosimilars for both capture and detection to reduce background and improve specificity.
Controls are essential (negative, positive, assay cut-point) to set sensitivity and specificity.
This format mainly detects bivalent, “bridging” ADA, reflecting the majority of clinically relevant anti-drug antibodies, but may not detect low-affinity or monovalent ADA.
In summary: A Evinacumab biosimilar used as both capture and detection reagent in a bridging ADA ELISA allows for highly specific, sensitive detection of anti-evinacumab antibodies in human serum to monitor immunogenicity during therapy.
References & Citations
1. Lang W, Frishman WH. Cardiol Rev. 2019;27(4):211-217.
2. Jiang S, Qiu GH, Zhu N, Hu ZY, Liao DF, Qin L. J Drug Target. 2019;27(8):876-884.
3. Lai M, Jiang X, Wang B, Cheng Y, Su X. Curr Mol Med. 2024;24(6):771-779.
4. Banerjee P, Chan KC, Tarabocchia M, et al. Arterioscler Thromb Vasc Biol. 2019;39(11):2248-2260.
5. Gao Y, Zhang B, Yang J. Expert Rev Clin Pharmacol. 2022;15(2):139-145.
6. Raal FJ, Rosenson RS, Reeskamp LF, et al. N Engl J Med. 2020;383(8):711-720.
7. Stefanutti C, Chan DC, Di Giacomo S, Morozzi C, Watts GF. Pharmaceuticals (Basel). 2022;15(11):1389.