Anti-Human CTLA-4 (Tremelimumab)

Product No.: C980

- -
- -
Product No.C980
Clone
CP-675
Target
CTLA-4
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Cytotoxic T-lymphocyte associated protein 4
Isotype
Human IgG2κ
Applications
B
,
ELISA
,
FA
,
FC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Recommended Isotype Controls
Immunogen
Original antibody generated by immunizing mice with cells expressing Human CTLA-4 recombinantly.
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
B
FA
FC
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 Tremelimumab. This product is for research use only. Tremelimumab activity is directed against human and cynomolgus monkey CTLA-4.
Background
Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is an activation induced, type I transmembrane protein of the Ig superfamily that is expressed as a covalent homodimer 1. CTLA-4 functions as an inhibitory receptor for the costimulatory molecules B7.1 (CD80) and B7.2 (CD86), inhibiting T cell activation and proliferation as well as IL-2 gene transcription by directly inhibiting TCR signal transduction.

Immune checkpoint blockade of CTLA-4 is a well-established treatment for cancer 2. Since CTLA-4 inhibits T cell activation, blocking CTLA-4 function enhances T cell activation as well as the immune response. Additionally, tremelimumab activity enhances the production of interleukin-2 and interferon-γ in human T cell blasts stimulated with B7-positive Raji cells. Tremelimumab also stimulates upregulation of the Th1/Th2 pathway, activates the Th17 pathway, and reduces expression of genes involved in epithelial-mesenchymal transition, angiogenesis, and cancer stemness. The mechanism of action includes antibody-dependent cell cytotoxicity.

Tremelimumab was generated by recombinant DNA technology using engineered XenoMice 1. Tremelimumab binds to CTLA-4 and blocks interaction with its ligands B7.1 (CD80) and B7.2 (CD86), thereby activating an enhanced T cell response against tumors 2. Additionally, tremelimumab inhibits binding of CTLA-4-Ig to immobilized B7.1 and B7.2. In vitro, binding of tremelimumab to CTLA-4 is >500 fold more selective than for human CD28-Ig, B7.2-Ig and IgG1. Tremelimumab does not initiate a nonspecific cytokine release or bind to Fc receptors 1. Additionally, tremelimumab activity is mainly mediated by direct activation of T effector cells rather than by affecting T regulatory cells 3.

Tremelimumab has been tested in a variety of therapeutic trials, including for hepatocellular, non-small cell lung, small cell lung, urothelial, biliary tract, thyroid, renal, gastrointestinal, and cervical cancers 2. Tremelimumab has been approved for use in the treatment of unresectable hepatocellular carcinoma and some metastatic non-small cell lung cancers.
Antigen Distribution
CTLA-4 is expressed by T lymphocytes and monocytes.
Ligand/Receptor
CD80 (B7-1) & CD86 (B7-2)
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Immunology
.
Inhibitory Molecules

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.

Research-grade Tremelimumab biosimilars are used as calibration standards or reference controls in PK bridging ELISAs by serving as the single analytical standard against which both biosimilar and reference product concentrations are measured in serum samples.

This approach is grounded in the current industry consensus favoring the use of a single PK assay with one analytical standard for both biosimilar and reference products during quantitative assessment. Here’s how the process works and why it is essential for PK bridging studies:

  • Single Analytical Standard: After confirming that the biosimilar and reference Tremelimumab are bioanalytically equivalent in the assay (i.e., the assay measures both with comparable accuracy and precision), the research-grade biosimilar is selected as the calibration standard for the entire method. This means all sample concentrations—whether from biosimilar or reference product—are interpolated from a standard curve generated with the biosimilar.

  • Preparation of Standards and QCs: During method validation and actual sample analysis, calibration standards (multiple defined concentrations of the biosimilar) are spiked into serum to generate a standard curve. Quality controls (QCs), prepared with both the biosimilar and the reference product at different concentrations, are assayed to confirm that both are quantified accurately against the single biosimilar-based standard curve.

  • PK Bridging ELISA Principle: A sandwich ELISA format is typically used, where Tremelimumab in serum is captured by plate-bound antibodies and detected by a secondary labeled antibody. The readout (commonly colorimetric) is proportional to the Tremelimumab concentration, which is then determined using the biosimilar-derived standard curve.

  • Ensuring Assay Suitability: Comprehensive method validation includes demonstrating that the assay’s accuracy, precision, and sensitivity are acceptable for both biosimilar and reference preparations, supporting the argument that PK comparability is measured without analytical bias.

  • Why Research-grade Biosimilars?: These are used for standards and controls rather than clinical-grade product because they are readily available in research quantities, can be characterized extensively, and are representative of the test article used in studies.

  • Purpose in PK Bridging: Using a biosimilar standard enables robust measurement of relative drug concentrations in clinical or preclinical serum samples, facilitating unbiased head-to-head PK comparison between biosimilar and originator products—a regulatory requirement for biosimilar development.

Key Points:

  • Research-grade Tremelimumab biosimilars act as the sole calibration standard in the bridging ELISA.
  • The assay provides direct quantitation of both biosimilar and reference drug in serum samples using this single standard.
  • Method validation ensures the assay is accurate and unbiased for both products across the anticipated concentration range.

Typical workflow:

  1. Generate a standard curve in serum using defined concentrations of the biosimilar.
  2. Prepare and analyze QC samples with both biosimilar and reference Tremelimumab.
  3. Analyze unknown serum samples from PK studies.
  4. Quantify all samples using the biosimilar-based standard curve, ensuring equivalence was established in validation.

This best-practice approach is endorsed by regulatory guidance and scientific literature as the optimal standard for PK bridging assays in biosimilar development.

Research-grade anti-CTLA-4 antibodies are commonly used in various in vivo models to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs). These models include:

Syngeneic Models

  1. Breast Cancer Models (4T1, EMT6): These models allow researchers to study the effects of anti-CTLA-4 blockade in breast cancer, where some cell lines respond partially while others do not respond well to the treatment.
  2. Colon Cancer Models (CT26, MC38): These models provide insights into differential sensitivity to anti-CTLA-4 monotherapy, with CT26 showing partial responses and MC38 being insensitive.
  3. Glioblastoma (GL261): A model used to study brain tumors and the effects of CTLA-4 blockade.
  4. Lung Cancer (LLC1): Useful for studying lung cancer responses to anti-CTLA-4 treatment.
  5. Pancreatic Cancer (Pan02): Utilized to explore pancreatic cancer treatment outcomes.
  6. Renal Cancer (Renca): Allows for the investigation of renal cancer responses.
  7. Sarcoma (MCA205): Used for studying sarcoma treatment effects.

Humanized Models

While syngeneic models are more commonly mentioned, humanized models can also be used to study human tumor biology and immune responses in a more relevant context. However, specific details about commonly used humanized models for anti-CTLA-4 studies are less prevalent in the literature.

Key Features of These Models

  • Immune System: Syngeneic models have an intact immune system, which is crucial for studying immune checkpoint blockade therapies like anti-CTLA-4.
  • Tumor Implantation: Tumors can be implanted subcutaneously or orthotopically, allowing for diverse experimental setups.
  • Treatment Protocols: Robust protocols are in place, aligned with published literature, to ensure consistent results.
  • Readouts: Common readouts include body weight, tumor size, and survival.

Characterization of TILs

In these models, TILs can be characterized by their ability to infiltrate tumors and their functionality, such as cytokine production and cytotoxic activity. For example, anti-CTLA-4 treatment often enhances CD8+ T cell infiltration and IFNγ production, contributing to antitumor activity.

Use of Tremelimumab Biosimilar with Other Checkpoint Inhibitors in Immune-Oncology Research

Researchers leverage Tremelimumab biosimilars—designed to mimic the effects of tremelimumab, an anti-CTLA-4 monoclonal antibody—primarily to study immune checkpoint blockade and explore synergistic effects in complex immune-oncology models. Here’s how they employ these biosimilars in combination with other checkpoint inhibitors:

Mechanism of Action

  • Tremelimumab targets CTLA-4, a key immune checkpoint protein expressed on T cells, blocking its inhibitory signal and thereby promoting T cell activation and tumor cell killing.
  • Anti-PD-1/PD-L1 and anti-LAG-3 antibodies block other inhibitory pathways, each with distinct mechanisms, potentially allowing for complementary or synergistic anti-tumor immune responses.
  • Combination therapy rationale: Since CTLA-4, PD-1, and LAG-3 inhibitors act on different regulatory axes, their simultaneous blockade can theoretically overcome compensatory immune suppression in the tumor microenvironment, leading to enhanced anti-tumor activity.

Laboratory and Preclinical Strategies

  • In vitro immune cell assays: Researchers incubate human T cells or PBMCs with tumor cells in the presence of Tremelimumab biosimilar alone or in combination with anti-PD-1, anti-PD-L1, or anti-LAG-3 biosimilars. They measure T cell proliferation, cytokine release (e.g., IL-2), and tumor cell killing as readouts of immune activation and synergy.
  • Co-culture models: Tumor organoids or 3D tumor spheroids are co-cultured with autologous immune cells to study the penetration, activity, and potential off-target effects of combination checkpoint blockade.
  • Preclinical animal models: Humanized mouse models engrafted with human immune cells and patient-derived xenografts (PDX) are treated with combinatorial regimens to assess tumor regression, immune infiltration, and biomarker changes.
  • Biomarker analysis: Flow cytometry and RNA sequencing are used to track changes in immune cell populations (e.g., Tregs, CD8+ T cells) and checkpoint receptor expression (CTLA-4, PD-1, LAG-3) following combination therapy.
  • Toxicity profiling: Given the increased risk of immune-related adverse events (irAEs) with combination therapy, researchers closely monitor for cytokine release syndrome, organ toxicity, and autoimmunity in these models.

Clinical and Translational Research

  • Phase I/II clinical trials: Tremelimumab has been tested in combination with anti-PD-L1 antibodies (e.g., durvalumab) and immune agonists (e.g., anti-CD40) in various solid tumors, including melanoma and non-small cell lung cancer. These trials assess safety, efficacy, and the potential for durable responses.
  • Synergy evaluation: Early clinical data suggest that combining CTLA-4 and PD-1/PD-L1 blockade can yield higher response rates than monotherapy, though with increased toxicity. Similar strategies are now being explored with LAG-3 inhibitors, based on preclinical evidence of complementary mechanisms.
  • Mechanistic studies: Blood and tumor biopsies from clinical trial participants are analyzed to identify predictive biomarkers of response, resistance mechanisms, and immune correlates of synergy or toxicity.

Key Challenges and Considerations

  • Toxicity management: Combination regimens often lead to higher rates of severe irAEs, necessitating careful dose optimization and supportive care strategies in both preclinical and clinical settings.
  • Model complexity: The use of biosimilars in immune-competent models is essential to recapitulate human immune responses, but can be limited by species differences in checkpoint protein biology.
  • Translational relevance: Findings from biosimilar-based studies must be validated in clinical trials, as the immune microenvironment and pharmacokinetic profiles can differ between models and patients.

Conclusion

Researchers use Tremelimumab biosimilars in conjunction with other checkpoint inhibitors (e.g., anti-PD-1, anti-PD-L1, anti-LAG-3) to dissect the mechanisms of immune checkpoint synergy, optimize combination regimens, and identify biomarkers of response and toxicity in complex immune-oncology models. These studies bridge preclinical discovery and clinical translation, informing the development of next-generation immunotherapies for cancer.

A Tremelimumab biosimilar can be used as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response against the therapeutic drug. In this assay format, the biosimilar molecule serves as a stand-in for the original Tremelimumab, allowing detection of patient antibodies directed against any formulation of the drug.

Context and Detail:

  • In a bridging ELISA, patient serum (which may contain anti-Tremelimumab antibodies) is incubated with two forms of Tremelimumab (or its biosimilar):
    • One form is immobilized on the ELISA plate to capture any anti-Tremelimumab antibodies present in the sample.
    • The other form is usually labeled (e.g., with biotin or horseradish peroxidase) and acts as the detection reagent.
  • If a patient’s sample contains ADA, these antibodies will “bridge” between the plate-bound and labeled Tremelimumab biosimilar, allowing detection through the labeled conjugate.
  • This format detects antibodies that bind to Tremelimumab regardless of whether they are produced in response to the original or the biosimilar, thus monitoring immunogenicity equivalently for both the reference and biosimilar drugs.

Supporting Information:

  • The biosimilar can be used interchangeably with the reference product in immunogenicity assays as long as it is analytically proven to be highly similar, which is the basis for regulatory acceptance.
  • Bridging ELISAs are the gold standard for the sensitive detection of ADAs for monoclonal antibody therapeutics and are commonly employed in the development and monitoring of biosimilars.
  • Use of a biosimilar as the assay reagent does not impact detection sensitivity if it is structurally and functionally equivalent to the reference.

Summary Table: Example Bridging ADA ELISA Using Tremelimumab Biosimilar

StepReagent UsedPurpose
Plate CoatingTremelimumab biosimilarCapture ADA from patient serum
SamplePatient serumMay contain anti-Tremelimumab antibodies (ADA)
DetectionLabeled Tremelimumab biosimilarBinds to ADA, completing the bridge
ReadoutEnzyme/fluorescence developmentQuantifies ADA in the sample

This assay measures the immunogenicity of Tremelimumab treatment by detecting antibodies patients develop against the drug, using the biosimilar interchangeably as a reagent in the assay.

References & Citations

1. Ribas A, Hanson DC, Noe DA, et al. Oncologist. 12(7):873-883. 2007.
2. Keam SJ. Drugs. 83(1):93-102. 2023.
3. Khan S, Burt DJ, Ralph C, et al. Clin Immunol. 138(1):85-96. 2011.
4. Hanson DC, Canniff PC, Primiano MJ, et al. Cancer Res. 64(7 Suppl):877. 2004.
5. Ribas A, Camacho LH, Lopez-Berestein G, et al. J Clin Oncol. 23(35):8968-8977. 2005.
6. Comin-Anduix B, Lee Y, Jalil J, et al. J Transl Med. 6:22. 2008.
7. von Euw E, Chodon T, Attar N, et al. J Transl Med. 7:35. 2009.
8. Ribas A, Benz MR, Allen-Auerbach MS, et al. J Nucl Med. 51(3):340-346. 2010.
9. Suarez N, Alfaro C, Dubrot J, et al. Int J Cancer. 129(2):374-386. 2011.
10. Cheng L, Creasy T, Pilataxi F, et al. Cancer Immunol Immunother. 71(5):1167–1181. 2022.
B
Indirect Elisa Protocol
FA
Flow Cytometry

Certificate of Analysis

- -
- -

Formats Available

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