Anti-human CD134 (OX40) (Vonlerolizumab)

Anti-human CD134 (OX40) (Vonlerolizumab)

Product No.: C3250

- -
- -
Product No.C3250
Clone
MOXR0916
Target
CD134
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
OX40, TNFRSF4, ACT35 antigen, OX40L receptor, TAX transcriptionally-activated glycoprotein 1 receptor
Isotype
Human IgG1κ
Applications
ELISA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
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.
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.
Regulatory Status
Research Use Only
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 Vonlerolizumab. Vonlerolizumab specifically targets and binds to CD134 (OX40).
Background
CD134, also known as OX40 or TNFRSF4, belongs to the tumor necrosis factor receptor (TNFR) superfamily. It is a protein found on the surface of activated T cells, natural killer (NK) cells, dendritic cells, and other immune cells. CD134 acts as a co-stimulatory molecule, boosting T cell proliferation, survival, and cytokine production when binding to its ligand, OX40L. This interaction plays a critical role in immune responses, rendering CD134 an appealing target for cancer immunotherapy1-4.

Vonlerolizumab, also known as MOXR0916, is a humanized monoclonal antibody of the IgG1 class, which specifically targets the CD134 (OX40) receptor in the body. Upon binding to this receptor, it acts as an agonist, thereby stimulating the activation and proliferation of T cells as well as other immune cells, and ultimately bolstering the body's antitumor response. Ongoing research focuses on exploring the potential of vonlerolizumab in treating a wide spectrum of solid tumors and hematologic malignancies5.

Antigen Distribution
CD134 is expressed on the surface of activated T cells, NK cells, dendritic cells, and other immune cells.
Ligand/Receptor
TNFSF4/OX40L/GP34, TRAF2, TRAF3, TRAF5, Human herpesvirus 6B/HHV-6B gQ1:gQ2 proteins
NCBI Gene Bank ID
UniProt.org
Research Area
Apoptosis
.
Biosimilars
.
Cancer
.
Cell Biology
.
Cell Death
.
Costimulatory Molecules
.
Immuno-Oncology
.
Immunology
.
Tumor Suppressors

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 Vonlerolizumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as the single analytical standard for quantifying both the biosimilar and reference (originator) drug concentrations in serum samples.

In a typical PK bridging ELISA for biosimilars:

  • A calibration curve is generated by spiking known amounts of the biosimilar or reference therapeutic into a relevant biological matrix (e.g., blank serum). These spiked samples cover a range of concentrations to enable accurate quantification across the expected concentration range in patient samples.
  • The biosimilar standard is routinely used as the assay calibrator, provided it is demonstrated to be bioanalytically similar to the reference product. This means that the biosimilar and reference must exhibit comparable concentration-response relationships in the assay.
  • Quality control (QC) samples are prepared using both the biosimilar and the reference product to further validate that the assay measures both entities with equivalent accuracy and precision.
  • This approach eliminates the need for separate calibration curves for the reference and biosimilar, minimizes variability, and streamlines bioanalytical comparability assessments.

Key steps in the use of biosimilars as standards in PK bridging ELISAs:

  • Establish bioanalytical similarity during the assay development phase. Comparative statistical analysis assesses whether the biosimilar and reference product yield overlapping calibration curves and similar QC recoveries in the matrix.
  • Formally validate the method, proving the ELISA can reliably quantify serum concentrations of both products using the biosimilar standard alone.
  • Ensure no significant matrix interference or non-specific binding, and comply with specificity, accuracy, linearity, and precision validation parameters as recommended by regulatory guidance and best practice.

Why biosimilars are used as standards:

  • When structural and functional comparability are established with chemistry, manufacturing, and control (CMC) data, it is generally accepted that the biosimilar will behave equivalently to the reference in ligand-binding assays like ELISA.
  • Using a single, well-characterized biosimilar standard reduces variability and simplifies data interpretation while maintaining regulatory compliance, as long as robust validation demonstrates equivalent assay performance for both molecules.

Thus, research-grade Vonlerolizumab biosimilars are employed as calibration standards in PK ELISA bridging studies after demonstrating bioanalytical similarity, enabling quantification of drug levels in clinical serum samples for both biosimilar and reference products using a single, validated protocol.

The primary in vivo models used to study the effects of research-grade anti-CD134 antibodies on tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) are syngeneic mouse tumor models. These models utilize immunocompetent mice with mouse-derived tumors, enabling robust evaluation of immune modulatory therapies such as anti-CD134 antibodies and detailed characterization of TILs.

Syngeneic Models:

  • Murine syngeneic models (e.g., BCL1 lymphoma in BALB/c mice, MC38 colon carcinoma, TC-1 lung epithelial tumor) are most commonly employed. These models provide a fully functional, intact immune system, making them suitable for studying the impact of CD134 targeting on tumor immunity and TIL composition.
  • Administration: In these studies, agonistic anti-CD134 antibodies are administered systemically (often intravenously or intraperitoneally) after tumor inoculation. Effects on tumor growth and survival are measured, and TILs are analyzed via flow cytometry or immunohistochemical methods.
  • Analysis of TILs: Syngeneic models allow detailed profiling of TIL populations—including CD8+ T cells, NK cells, and regulatory T cells—following treatment with anti-CD134 antibodies, thus characterizing immune changes within the tumor microenvironment.

Alternatives and Humanized Models:

  • While humanized mouse models (mice engrafted with human immune cells or genetically modified to express human CD134) are a theoretical option for evaluating anti-human CD134 antibodies, these are less routinely used due to complexity, higher cost, and the need for antibodies cross-reactive with human CD134.
  • Transgenic syngeneic models expressing human immune targets, or syngeneic tumors engineered to overexpress human CD134, can also be employed to bridge the species specificity issue for therapeutic antibodies targeting human CD134.
Model TypeExampleRelevance for Anti-CD134 StudiesTIL Analysis Feasibility
Syngeneic (mouse:mouse)BCL1 in BALB/c, MC38 in C57BL/6Highly relevant, widely usedComprehensive (mature immune system)
Humanized (mouse:human)NSG with human PBMCsLess common, human-specific onlyFeasible, technically demanding
Transgenic/ChimericMice expressing human CD134Useful for evaluating human mAbsFeasible, model availability varies

In the literature, the majority of preclinical studies with anti-CD134 mAbs use murine syngeneic models administered with murine-reactive agonistic anti-CD134 antibodies to evaluate tumor control and the composition and function of TILs. Enhanced findings from these models have contributed to justifying clinical development of OX40-targeting (CD134-targeting) immunotherapies.

Limitations: Cross-species reactivity is crucial—antibodies must recognize mouse CD134 for classic syngeneic studies, or the model must be adapted to express human targets for human-specific therapeutics.

Summary:
Syngeneic mouse tumor models remain the gold standard for in vivo studies of research-grade anti-CD134 antibodies aimed at assessing tumor inhibition and TIL characterization, with humanized or transgenic approaches reserved for specific antibody or mechanistic questions.

Researchers use Vonlerolizumab biosimilars—which target the PD-1/PD-L1 axis—in combination with other checkpoint inhibitor biosimilars (such as anti-CTLA-4 or anti-LAG-3) to assess potential synergistic effects in immune-oncology models by exploiting the complementary mechanisms of different immune checkpoint pathways. Such combinatorial approaches are designed to enhance anti-tumor immune responses beyond what is possible with monotherapies.

Key Methods and Rationale:

  • Mechanistic Complementarity: Anti-PD-1/PD-L1 agents such as Vonlerolizumab act primarily at the tumor site to reinvigorate exhausted T cells, while anti-CTLA-4 agents are thought to act predominantly in lymphoid organs, enhancing the priming and proliferation of T cells. Additionally, anti-LAG-3 or anti-TIM-3 agents target alternative inhibitory pathways that often co-exist with PD-1-mediated inhibition, further reversing T cell exhaustion in complex tumor environments.
  • Preclinical Models: Researchers evaluate combinations in mouse tumor models genetically engineered to express human checkpoints or using patient-derived xenografts. These studies measure tumor growth inhibition, survival outcomes, and comprehensive immune profiling (e.g., T cell activation, cytokine production, exhaustion markers).
  • Synergy Assessment: Synergistic effects are assessed by comparing tumor responses and immune activation from combination therapies to the effects of each agent alone. The aim is to demonstrate that dual or triple checkpoint blockade provides superior antitumor activity compared to monotherapies.
  • Biosimilar-Specific Studies: Biosimilar checkpoint inhibitors, including Vonlerolizumab analogs, are selected to improve access and reduce costs, but their equivalence to reference products is carefully validated in terms of pharmacodynamics, pharmacokinetics, safety, and immunogenicity. Studies are structured similarly to those with originator biologics, ensuring robust confirmation that observed effects derive from mechanistically relevant checkpoint interactions rather than biosimilar variability.

Experimental Readouts:

  • Tumor metrics: Tumor growth delay, regression, and overall survival.
  • Immune parameters: Levels of activated CD8+/CD4+ T cells, cytokine release, immune cell infiltration, and expression of exhaustion markers.
  • Toxicity assessment: Incidence and severity of immune-related adverse events, with attention to increased toxicities common in combination regimens.
  • Comparative analysis: Side-by-side evaluation of biosimilars and reference products in combination settings to confirm that biosimilars replicate reference drug synergy.

Limitations and Considerations:

  • Rigorous confirmation of biosimilarity is essential, as not all biosimilars undergo extensive head-to-head combination studies before regulatory approval.
  • Most preclinical synergy data are generated in animal models, which may not fully replicate human tumor immune microenvironments.
  • While promising, combination immunotherapy often increases the risk of immune-related adverse events, mandating careful dose optimization and monitoring in translational studies.

No literature directly describing "Vonlerolizumab" combination studies was identified in the current search. The provided answer is based on general checkpoint inhibition combinatorial models, supported by the cited reviews and biosimilar overview studies.

In immunogenicity testing, a Vonlerolizumab biosimilar can be used as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor patient immune responses against the therapeutic drug by leveraging the structural and functional similarity to the reference drug.

In a typical bridging ADA ELISA:

  • Biotinylated (labeled) biosimilar Vonlerolizumab is immobilized on a streptavidin-coated microplate, serving as the capture reagent.
  • Patient samples (e.g., serum containing potential anti-drug antibodies) are added, allowing any antibodies that recognize Vonlerolizumab to bind.
  • Subsequently, enzyme-labeled (e.g., HRP-conjugated) biosimilar Vonlerolizumab is added as the detection reagent. This detects bivalent anti-drug antibodies that can bridge between the immobilized and labeled drug molecules.
  • Upon the addition of the substrate, the resulting signal (color development) is proportional to the presence and amount of anti-drug antibodies in the patient’s sample.

Why use a biosimilar as reagent?

  • Biosimilars are designed to be highly similar, with no clinically meaningful differences from the reference drug in terms of safety, purity, and potency. Thus, the biosimilar can function identically to the original drug as a reagent in ADA assays, providing reliable immunogenicity detection.
  • Comparative studies confirm that biosimilars exhibit matching immunogenicity profiles with their reference drugs, validating their use in such sensitive assays.

Key points on the procedure:

  • Both the capture (usually biotinylated) and detection (usually labeled with HRP or similar) reagents can be the biosimilar form, so long as it possesses identical antibody-binding epitopes to the reference drug.
  • This assay detects bivalent antibodies present in patient samples, which are characteristic of an immune response to Vonlerolizumab.
  • High sensitivity allows detection even at low ADA concentrations but specificity must be closely managed, due to possible matrix interference from serum proteins or the therapeutic drug itself.

In summary, Vonlerolizumab biosimilar serves as a functional surrogate for the reference drug in a bridging ADA ELISA, effectively capturing and detecting anti-drug antibodies that develop in response to therapeutic exposure, thereby enabling immunogenicity monitoring.

References & Citations

1. Adler AJ, Vella AT. Oncoimmunology. 2013;2(1):e22837.
2. Li M, Zhang Y. Cell Mol Immunol. 2005;2(6):467-471.
3. Baumann R, Yousefi S, Simon D, Russmann S, Mueller C, Simon HU. Eur J Immunol. 2004;34(8):2268-2275.
4. Melero I, Hirschhorn-Cymerman D, Morales-Kastresana A, Sanmamed MF, Wolchok JD. Clin Cancer Res. 2013;19(5):1044-1053.
5. Kim TW, Burris HA, de Miguel Luken MJ, et al. Clin Cancer Res. 2022;28(16):3452-3463.
6. Vonlerolizumab Humanized Recombinant Monoclonal Antibody (MA5-42105). Accessed August 15, 2024. https://www.thermofisher.com/antibody/product/Vonlerolizumab-Humanized-Antibody-Recombinant-Monoclonal/MA5-42105
Indirect Elisa Protocol

Certificate of Analysis

Formats Available

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