Anti-Human PDGFR (Olaratumab) – Fc Muted™

Anti-Human PDGFR (Olaratumab) – Fc Muted™

Product No.: LT2605

- -
- -
Product No.LT2605
Clone
IMC-3G3
Target
PDGF Rα
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Anti PDGFR, DB06043, IMC-3G3
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC
,
IP
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Human PDGFRA
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 (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Additional Applications Reported In Literature ?
ELISA,
WB,
IP,
FC,
FA
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 Olaratumab. This product is for research use only. Olaratumab activity is directed against human PDGFRα.
Background
Platelet-derived growth factor receptor (PDGFR) is a class III receptor tyrosine kinase that upon binding to PDGF molecules, dimerizes and activates, triggering an intracellular signaling cascade essential to normal embryogenesis, development, migration, survival signaling, proliferation, cellular chemotaxis, and intracellular calcium metabolism1. Hyperactive signaling along the PDGF/PDGFR axis drives pathogenesis in nonmalignant disorders (e.g., atherosclerosis, pulmonary fibrosis) and can promote tumor growth1 or neurodegenerative disease2. PDGFR is a target for anticancer therapeutic development1. PDGF/PDGFR signaling influences cancer biology via autocrine growth stimulation of cancer cells, regulation of stromal-derived fibroblasts, and regulation of angiogenesis.

Olaratumab was developed as an anticancer therapeutic agent1. Human IgG transgenic mice were immunized with PAE Rα cells and boosted with human PDGFRα extracellular domain (ECD)3. Splenocytes with high serum PDGFRα binding activity and high blocking titers against the PDGF/PDGFRα ligand receptor interaction were isolated, fused with myeloma cells, subcloned, and purified. Antibodies were further tested for binding to PDGFRα by direct binding ELISA and surface plasmon resonance, for blocking activity in solid-phase and cell-based ligand binding assays, and for receptor/ligand activation. Olaratumab was found to inhibit PDGF stimulated mitogenesis, PDGF-AA and PDGF-BB induced receptor phosphorylation, activation of the MAPK proliferation and Akt survival pathways, and in mouse xenograft models inhibits tumor growth and PDGFRα stimulation. Additionally, olaratumab inhibits cell proliferation and survival in mouse and human hepatoma cell lines 4 as well as PDGF-AA induced receptor phosphorylation and cell proliferation in ovarian cancer cells5. Clinical trials were initiated on the basis of these results6, 7, 8, 9.

Olaratumab shows no cross reactivity with PDGFRβ in solid phase ELISA or cell-based phosphorylation assays, nor to mouse PDGFRα as determined by ELISA, mitogenic, and phosphorylation assays3.

Olaratumab clone AL10, a non-therapeutic biosimilar antibody for research use only was developed recombinantly and has the same variable regions as the original therapeutic.
Antigen Distribution
PDGFRα is expressed on platelets, megakaryocytes, fibroblasts, pericytes, vascular smooth muscle cells, neurons, and myoblasts. Malignant cells from several types of cancer (ovary, prostate, breast, lung, brain, skin, bone, gastrointestinal, kidney) can also express PDGFRα.
Ligand/Receptor
PDGFRA, CD140b
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.

Research-grade Olaratumab biosimilars serve as critical analytical tools in pharmacokinetic bridging ELISA assays, providing standardized reference points for accurate drug concentration measurements in serum samples. The integration of biosimilars as calibration standards represents a sophisticated bioanalytical approach that addresses the unique challenges of measuring therapeutic antibodies across different product formulations.

Biosimilar Integration in PK Assay Development

The most effective strategy involves developing a single PK assay using a single analytical standard for quantitative measurement of both biosimilar and reference Olaratumab products. This approach offers significant advantages by decreasing inherent variability associated with running multiple methods and eliminating the need for crossover analysis during blinded clinical studies.

The bioanalytical strategy begins with a comprehensive method qualification study that generates precision and accuracy data sets for both biosimilar and reference products, followed by statistical analysis to determine if the test products are bioanalytically equivalent within the method. When bioanalytical comparability is established, the biosimilar is typically selected as the analytical standard for the single method.

ELISA Assay Configuration and Standards

Olaratumab ELISA kits utilize either sandwich ELISA or competitive enzyme immunoassay formats for drug quantification. In the competitive format, recombinant Human CD140a (PDGFR) is pre-coated onto microplates, and standards or samples are premixed with biotin-labeled antibody before being pipetted into wells. The Olaratumab in samples competitively binds to the pre-coated protein with biotin-labeled Olaratumab, creating an inverse relationship between color development and drug concentration.

Standard Preparation and Range:

  • Biosimilar standards are typically prepared at concentrations ranging from 50 to 12,800 ng/mL in human serum
  • Commercial ELISA kits offer detection ranges of 156.25 to 10,000 ng/mL
  • Multiple independent standard sets (typically nine) are analyzed during method validation

Quality Control and Validation Framework

The validation process incorporates multiple product sources to ensure method robustness. Validation samples are prepared using biosimilar, FDA-licensed, and EU-authorized products at specific concentrations including 50, 150, 1,250, 9,600, and 12,800 ng/mL. These samples are quantified against the biosimilar standard curve to demonstrate analytical equivalence across different product sources.

Performance Characteristics:

  • Sensitivity: Detection limits range from 0.01 ng/mL to 112.17 ng/mL depending on the specific assay format
  • Precision: Both intra-assay and inter-assay precision maintained at <20%
  • Recovery: Typically 80-120% for serum and plasma samples

Bioanalytical Equivalence Assessment

Statistical evaluation of bioanalytical comparability involves comparing 90% confidence intervals to pre-defined equivalence intervals of [0.8, 1.25]. This stringent criterion ensures that measurement variability is minimized when supporting PK similarity studies. The approach combines totality of evidence to conclude bioanalytical equivalence, providing scientific rigor necessary for regulatory submissions.

The validated method enables accurate quantification of Olaratumab concentrations in both serum and plasma samples, supporting pharmacokinetic studies that demonstrate biosimilar equivalence to the reference product Lartruvo. This bioanalytical framework ensures that concentration data serves as a reliable foundation for PK bioequivalence assessment and dose-response profile characterization in clinical development programs.

The primary models for research-grade anti-PDGF Rα antibody in vivo studies to assess tumor growth inhibition and profile tumor-infiltrating lymphocytes (TILs) are syngeneic mouse tumor models; humanized and xenograft models are rarely used due to species specificity of available antibodies and immune context dependencies.

Supporting context and core details:

  • Syngeneic Models:
    These involve transplanting mouse tumor cells into immunocompetent mice of the same genetic background. This setup allows for meaningful study of tumor-immune interactions because the mouse’s immune system remains intact, enabling robust characterization of how anti-PDGF Rα antibodies modulate the immune environment—including effects on TILs.

    • Common syngeneic tumor lines include MC38 (colon carcinoma), TC-1, and B16 (melanoma).
    • These models are critical for immunotherapy evaluation since they allow assessment of lymphocyte infiltration, immune activation, and checkpoint effects.
  • Antibody Specificity and Use
    Most research-grade anti-PDGF Rα antibodies (e.g., R&D Systems AF1062) are raised against mouse PDGFRα and validated in mouse tumors or immunological studies, making syngeneic mouse models the preferred setting.

    • Neutralization and in vivo inhibition of PDGFRα signaling have been demonstrated in murine models, linking to altered tumor growth and immune infiltration.
  • Humanized and Xenograft Models:
    Few research-grade anti-PDGF Rα antibodies have cross-reactivity to both human and mouse; most "humanized" or xenograft models instead use antibodies or receptor-targeting strategies for PDGFRβ (not α), as in several published in vivo studies. While xenografts are widely used for evaluating tumor growth inhibition, their immunodeficient hosts prevent meaningful TIL characterization.

  • Example Tumor Characterization
    Studies using syngeneic systems often measure:

    • Tumor size/growth in response to antibody treatment.
    • Flow cytometry or immunohistochemistry for TIL profiling—assessing CD8(^+) T cells, Tregs, macrophages, and activation markers.

Summary:
Syngeneic mouse tumor models are the primary and most functionally relevant system for in vivo evaluation of anti-PDGF Rα antibodies aimed at studying both tumor control and TIL changes, due to compatibility with both immunological readouts and available research antibody specificity. Humanized or xenograft models are not typically used for this purpose.

Use of Olaratumab Biosimilar with Checkpoint Inhibitors in Immune-Oncology Research

Olaratumab is a fully human monoclonal antibody targeting platelet-derived growth factor receptor alpha (PDGFR-α), blocking the binding of its ligands and inhibiting downstream signaling crucial for tumor growth and survival. While primarily approved for soft tissue sarcoma, interest in its broader application—including combination with immune checkpoint inhibitors—has grown, though clinical and preclinical evidence in this specific context remains limited.

Mechanisms and Potential Synergies

  • Olaratumab's Mechanism: Olaratumab inhibits PDGFR-α, a receptor implicated in tumor proliferation, angiogenesis, and microenvironment remodeling, which can indirectly affect immune cell infiltration and function within tumors. This indirect immune modulation is distinct from the direct T-cell activation facilitated by checkpoint inhibitors such as anti-CTLA-4, anti-PD-1/PD-L1, or anti-LAG-3 agents, which act by releasing immune suppression and enabling cytotoxic T-cell attack on cancer cells.
  • Checkpoint Inhibitor Mechanisms: Agents like anti-CTLA-4 and anti-LAG-3 have different molecular targets and immune effects. For example, anti-PD-1/CTLA-4 combinations directly activate cytotoxic CD8+ T cells, while anti-PD-1/LAG-3 regimens rely more on CD4+ helper T cells and reduce regulatory T-cell (Treg) activity. These differences highlight the potential for varied synergistic effects when combined with pathway inhibitors like olaratumab.

Research Strategies

  • Preclinical Combination Studies: Researchers can employ olaratumab biosimilars alongside checkpoint inhibitor biosimilars (e.g., anti-CTLA-4, anti-PD-1, anti-LAG-3) in complex immune-oncology models to investigate whether dual pathway blockade enhances anti-tumor immunity. The hypothesis is that olaratumab’s modulation of the tumor microenvironment (e.g., reducing fibrosis, improving vascularization, altering immune cell recruitment) may make tumors more susceptible to immune attack by checkpoint inhibitors.
  • Model Systems: In vitro and in vivo models (e.g., mouse xenografts, organoids, or humanized immune system models) can be used to assess tumor growth inhibition, immune cell infiltration, cytokine profiles, and survival outcomes. For instance, olaratumab has demonstrated efficacy in PDGFR-α expressing tumor models, and similar studies could be expanded to include immune checkpoint blockade.
  • Biosimilars in Research: The use of biosimilars allows for cost-effective, reproducible preclinical studies. Biosimilars faithfully replicate the properties of reference biologics, enabling researchers to explore combination therapies without the cost barriers of originator drugs.

Challenges and Considerations

  • Mechanistic Overlap and Distinction: While checkpoint inhibitors target immune cell regulation, olaratumab targets tumor and stromal cell signaling. The two may act synergistically if olaratumab “primes” the tumor for immune attack or additively if their mechanisms are independent.
  • Biomarker Development: Identifying tumors with high PDGFR-α expression and specific immune cell infiltrates may help predict which patients could benefit most from such combinations.
  • Safety and Toxicity: Combining agents with distinct side-effect profiles (e.g., immune-related adverse events from checkpoint inhibitors and potential cardiovascular effects from olaratumab) requires careful preclinical evaluation.

Current Evidence and Gaps

There is strong preclinical rationale for studying olaratumab biosimilars in combination with checkpoint inhibitors, but published studies specifically addressing this combination in immune-oncology models are not detailed in the available literature. Most research on combination checkpoint inhibitors focuses on combining multiple immune-targeting agents (e.g., anti-CTLA-4 plus anti-PD-1). The unique contribution of a PDGFR-α inhibitor like olaratumab to such regimens remains an area ripe for investigation.

Summary Table: Potential Research Approaches

CombinationMechanism HypothesisModel SystemsKey Readouts
Olaratumab + anti-CTLA-4Tumor microenvironment modulation + direct T-cell activationMouse xenografts, organoidsTumor growth, immune infiltration
Olaratumab + anti-PD-1Stromal signaling inhibition + immune checkpoint releaseHumanized mice, 3D culturesSurvival, cytokine profiles
Olaratumab + anti-LAG-3Reduced fibrosis + enhanced CD4+/CD8+ T-cell activityMetastatic modelsTreg suppression, tumor regression

Conclusion

Researchers can leverage olaratumab biosimilars with checkpoint inhibitor biosimilars to probe synergistic anti-tumor effects in complex immune-oncology models, hypothesizing that stromal and immune pathway dual blockade may overcome resistance and enhance efficacy. Preclinical studies should focus on mechanistic interactions, optimal dosing, biomarker development, and safety, using advanced model systems and biosimilars to enable robust, reproducible science. While the field awaits more direct evidence, the rationale for such combinations is compelling and aligns with the trend toward multimodal immunotherapy in cancer research.

In immunogenicity testing for biologics like Olaratumab and its biosimilars, a bridging anti-drug antibody (ADA) ELISA is commonly used to monitor whether a patient’s immune system has generated antibodies against the therapeutic drug. Here’s how a biosimilar version of Olaratumab can be employed in this assay, either as the capture or detection reagent:

Role of Biosimilar in Bridging ADA ELISA

Bridging ELISA is designed to detect bivalent antibodies (such as IgGs) that can simultaneously bind two drug molecules—one molecule for capture and another for detection. In this context, the biosimilar serves as a critical reagent, replacing the reference Olaratumab, to assess immune responses that may cross-react with both the reference and biosimilar products.

As Capture Reagent

  • Immobilization: The biosimilar is immobilized onto the assay plate (e.g., via biotinylation and binding to streptavidin-coated wells). Patient serum or plasma is added; if anti-Olaratumab antibodies (ADAs) are present, they bind to the captured biosimilar molecules.
  • After washing, a detection reagent is added—typically the same biosimilar (or reference drug) conjugated to an enzyme such as horseradish peroxidase (HRP). If ADAs are present, they will “bridge” the captured and detection drug molecules.
  • Signal generation: Subsequent addition of a chromogenic substrate (e.g., TMB) yields a detectable signal if ADA is present.

As Detection Reagent

  • Capture and detection roles can be reversed: In some assay designs, the reference drug is immobilized, and the biosimilar is enzyme-labeled for detection (or vice versa).
  • Same principle applies: ADAs that recognize epitopes common to both the biosimilar and reference will bind both molecules, enabling signal generation.

Advantages and Considerations

  • Cross-reactivity: Using a biosimilar as a reagent allows detection of ADAs that recognize shared epitopes between the biosimilar and reference, ensuring the assay is relevant for biosimilar immunogenicity assessment.
  • Highly sensitive: Bridging ELISA is sensitive and high-throughput, but specificity can be affected by serum components, requiring robust optimization.
  • Comparative immunogenicity: If the biosimilar is compared directly against the reference product, a “two-assay” approach may be used, where each drug is used in separate, parallel assays to assess any differences in immunogenicity, though this introduces complexity.

Regulatory and Clinical Relevance

  • FDA Guidelines: Immunogenicity testing for biosimilars typically employs a tiered approach (screening, confirmation, titration, and neutralization). ADA assays are usually semi-quantitative and designed to detect antibodies that may affect drug safety and efficacy.
  • Neutralizing antibodies (NAbs): If ADAs are detected, further cell-based or ligand-binding assays may be used to determine if these antibodies neutralize the drug’s function.

Summary Table

StepCapture ReagentDetection ReagentPurpose
ImmobilizationBiosimilar OlaratumabN/ACaptures any anti-Olaratumab ADAs
DetectionN/ABiosimilar (or reference)Binds ADAs, enables signal detection
Signal GenerationN/AEnzyme/substrateQuantifies ADA presence

In summary, a Olaratumab biosimilar can be used as either the capture or detection reagent in a bridging ADA ELISA to monitor patient immune responses against the therapeutic drug, ensuring detection of antibodies that may impact both the biosimilar and reference product. This approach supports comparative immunogenicity assessment essential for biosimilar development and patient safety.

References & Citations

Indirect Elisa Protocol
FA
Flow Cytometry
Immunoprecipitation Protocol
General Western Blot Protocol

Certificate of Analysis

Formats Available

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