Anti-Human PDGFR (Olaratumab)

Anti-Human PDGFR (Olaratumab)

Product No.: LT2600

- -
- -
Product No.LT2600
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
Active
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,
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 Olaratumab. 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 are used as calibration standards or reference controls in PK bridging ELISA assays to generate a standard curve, enabling accurate quantification of Olaratumab concentrations in serum samples. The biosimilar is typically selected to serve as the analytical standard when bioanalytical equivalence with the reference product is established, ensuring that both test and reference drug concentrations can be directly compared within a single validated method.

Key steps and rationale:

  • Standard Preparation and Calibration: Known concentrations of the Olaratumab biosimilar are prepared (typically through serial dilutions) and run in parallel with unknown serum samples in the ELISA assay. This set of standards generates a standard curve, which correlates absorbance values to Olaratumab concentrations, allowing quantification of the drug in test samples.

  • Matrix Considerations: Since serum is a complex biological matrix, standards should ideally be prepared in a similar matrix (human serum) to account for potential matrix effects (interference from serum components). This approach ensures any matrix-related signal shifts are accounted for in both calibration standards and unknowns.

  • Single-Method Calibration for PK Bridging: When developing pharmacokinetic bioanalytical assays for biosimilar development, the preferred practice is to use a single assay and a single well-characterized analytical standard—often the biosimilar itself—for both biosimilar and reference product quantification. This reduces analytical variability and supports regulatory requirements for analytical robustness and equivalence testing.

  • Validation and Equivalence Demonstration: Multiple validation samples of biosimilar and reference Olaratumab are spiked into serum at relevant concentrations. Their recoveries are quantified against the biosimilar standard curve, and statistical analysis confirms whether the assay quantifies both products equivalently. Only if bioanalytical equivalence is confirmed can the biosimilar standard serve as the reference for both arms.

  • Controls: In addition to calibration standards, quality control (QC) samples at various concentrations are included on each plate to monitor assay performance and reliability throughout the study.

  • Result Interpretation: Drug concentrations in serum samples are interpolated from the standard curve generated with the biosimilar calibrators, yielding precise, comparable PK data for both biosimilar and reference Olaratumab.

In summary: Research-grade Olaratumab biosimilars are meticulously qualified and used to create the calibration curve in PK bridging ELISAs. This enables direct measurement and cross-comparison of drug concentrations in serum samples, supporting pharmacokinetic equivalence and regulatory submissions.

The primary in vivo models used for studying the effects of a research-grade anti-PDGF Rα antibody on tumor growth and tumor-infiltrating lymphocyte (TIL) characterization are syngeneic murine models and, to a lesser extent, humanized mouse models.

Key Details:

  • Syngeneic Models (Most Common):

    • These use murine tumor cell lines implanted into immunocompetent mice of the same genetic background, preserving native immune responses.
    • Syngeneic models are considered the standard for evaluating immunotherapies like checkpoint inhibitors or antibodies affecting the tumor microenvironment, such as anti-PDGFRα, because they enable rigorous analysis of tumor-immune interactions and TILs in a fully functional immune system.
    • These models have been fully characterized for gene expression profiles, baseline TIL populations, and responses to immunotherapies, which is essential for TIL analysis.
    • Examples include commonly used syngeneic models such as MC38 (colon carcinoma), TC-1 (lung carcinoma), and B16 (melanoma), where intervention with immune-modulating antibodies can be tracked for both tumor growth inhibition and changes in immune infiltrates.
  • Humanized Mouse Models (Supplementary, More Complex):

    • Humanized models, where human immune cells and/or tumor tissue are engrafted into immunodeficient mice, are sometimes used if the research demands analysis with fully human immune components.
    • These models are less frequently used for anti-PDGF Rα studies due to complexity, limited immune system reconstitution, and availability of compatible reagents.
  • PDGF Rα-Driven Tumor Models:

    • Experimental systems often use tumors with overexpression or genetic alteration of PDGF Rα, such as engineered glioblastoma or brain tumor models in mice.
    • In these models, a research-grade anti-mouse PDGF Rα antibody is administered to directly block receptor function, with downstream effects on tumor growth and immune infiltration measured using proliferation assays and immunohistochemistry (IHC) for TIL analysis.

Tumor-Infiltrating Lymphocytes (TILs):

  • Syngeneic models are specifically optimized for TIL characterization, as these models reflect the effects of immunotherapies on endogenous immune populations.
  • Studies typically assess TILs pre- and post-antibody treatment by flow cytometry, IHC, or gene expression analysis in tumor tissues harvested from treated and control groups.

In summary:
The syngeneic mouse model represents the primary preclinical platform for in vivo administration of research-grade anti-PDGF Rα antibodies to study both tumor growth inhibition and TIL dynamics. Humanized models can be employed if human-specific interactions are required but are less commonly used for this research focus.

Key Concepts

Olaratumab is a monoclonal antibody targeting platelet-derived growth factor receptor α (PDGFR-α), inhibiting ligand binding and downstream signaling important for cancer cell proliferation and survival. It is distinct from checkpoint inhibitors such as anti-CTLA-4 and anti-LAG-3 antibodies, which primarily modulate T-cell activity by disrupting immune checkpoint signaling, thereby enhancing antitumor immune responses.

Current research has focused on combination therapies in immune-oncology (IO), especially using multiple checkpoint inhibitors with complementary mechanisms (e.g., anti-PD-1, anti-CTLA-4, anti-LAG-3). However, there is no published evidence in the provided search results of researchers using olaratumab biosimilars in combination with anti-CTLA-4 or anti-LAG-3 biosimilars to study synergistic effects in preclinical or clinical models.

Research Approaches for Combination Studies

Typical IO Combination Studies

  • Checkpoint inhibitor combinations: Researchers often combine agents like anti-PD-1/PD-L1 with anti-CTLA-4 or anti-LAG-3 to exploit different immune activation mechanisms and overcome resistance.
  • Mechanistic focus: These studies often dissect how each inhibitor modulates immune cell subsets (e.g., CD4+ T cells, CD8+ T cells, Tregs) and identify immune correlates of response or resistance.
  • Biosimilar use: Biosimilars of checkpoint inhibitors (e.g., Peresolimab biosimilar as a PD-1/PD-L1 inhibitor analogue) are used in research to replicate the effects of originator drugs, enabling cost-effective and scalable preclinical studies.

Olaratumab’s Place in IO Research

  • Mechanism of action: Olaratumab targets the PDGFR-α pathway, primarily impacting tumor stromal and cancer cell survival, rather than directly modulating T-cell immunity.
  • Rationale for combination: While olaratumab has been studied in combination with chemotherapy (e.g., doxorubicin), there is no clear rationale—based on the provided data—for combining it with checkpoint inhibitors, as their mechanisms are largely non-overlapping and target different biological processes.
  • Potential applications: If used in IO models, olaratumab could, in theory, be tested alongside checkpoint inhibitors to determine if stromal modulation (via PDGFR-α inhibition) augments the efficacy of immune activation. However, such studies are not reported in the current literature cited here.

Current Limitations and Gaps

  • No reported studies: The search results do not identify any research where olaratumab biosimilars are combined with anti-CTLA-4 or anti-LAG-3 biosimilars in immune-oncology models.
  • Mechanistic mismatch: The biological rationale for such a combination is unclear, since olaratumab primarily affects tumor stroma and survival pathways, whereas checkpoint inhibitors directly engage the immune system.
  • Biosimilar availability: Biosimilars of checkpoint inhibitors are used in research, but olaratumab biosimilars are not mentioned in the context of IO combination studies.

Summary Table: Research Context

Agent TypePrimary TargetMain Mechanism in IOEvidence of Combination with Olaratumab
Olaratumab (biosimilar)PDGFR-αStromal/cancer cell inhibitionNot reported
Anti-CTLA-4 (biosimilar)CTLA-4 (T cells)T-cell activationNot reported
Anti-LAG-3 (biosimilar)LAG-3 (T cells)T-cell activationNot reported

Conclusion

Based on the available data, researchers are not currently using olaratumab biosimilars in combination with anti-CTLA-4 or anti-LAG-3 biosimilars to study synergistic effects in complex immune-oncology models. The field has focused on combining checkpoint inhibitors with complementary immune targets, and while biosimilars are increasingly used in IO research, olaratumab’s distinct mechanism and lack of direct immune modulation make it an unconventional partner for checkpoint inhibitor combinations at this time. Future research could theoretically explore such combinations if a biological rationale emerges linking stromal modulation to enhanced immune response, but no such studies are documented in the current literature.

A Olaratumab biosimilar in a bridging ADA ELISA can be used as either the capture or detection reagent to measure anti-drug antibodies (ADAs) in patient samples and monitor immune responses against Olaratumab therapy.

In a bridging ADA ELISA:

  • The assay relies on the bivalent nature of ADAs, which can simultaneously bind two identical molecules of therapeutic antibody (drug or biosimilar).
  • Typically, one form of the drug (either Olaratumab or its biosimilar) is immobilized on the ELISA plate as the capture reagent.
  • Patient serum is added; any ADAs present will bind to the immobilized Olaratumab biosimilar.
  • A second form of Olaratumab biosimilar, labeled with a detection tag (such as HRP or biotin), is added as the detection reagent. If ADAs are present, they will act as molecular bridges between the immobilized and the labeled biosimilar, resulting in a detectable signal.

Key details:

  • Choice of biosimilar as reagent: The biosimilar can be used instead of the original biologic to ensure detection of ADAs that recognize epitopes shared between both products. This is crucial for immunogenicity comparison and can be particularly relevant in biosimilar development and monitoring studies.
  • Detection protocol: For example, one approach would use streptavidin-coated plates with a biotinylated Olaratumab biosimilar as the capture reagent and an HRP-labeled Olaratumab biosimilar for detection. The presence of ADAs is identified by signal produced when bridging occurs.
  • Comparative immunogenicity: To assess immune responses, matched bridging ELISAs may employ parallel reagent sets: one with the reference Olaratumab and one with the biosimilar. This helps characterize potential differences in immunogenicity between the original and biosimilar.

Practical context:

  • Using a biosimilar in the ADA assay ensures the assay is sensitive to antibodies that may develop in response to either the originator or the biosimilar. It also supports regulatory and clinical study requirements by facilitating comparative immunogenicity analysis.
  • The bridging format is highly sensitive due to the dual binding feature of bivalent ADAs, but it can be affected by matrix components or high drug concentrations in serum.

Summary of protocol steps (basic bridging ELISA):

  • Coat plate with capture Olaratumab biosimilar (biotinylated if using streptavidin plates).
  • Add patient serum.
  • Add detection Olaratumab biosimilar (labeled, e.g., HRP).
  • Detect signal (e.g., colorimetric readout correlating to ADA presence).

This approach is widely used to monitor immunogenicity throughout clinical studies involving monoclonal antibodies and their biosimilars, and is recommended for both regulatory submissions and routine pharmacovigilance.

References & Citations

1. Shah GD, Loizos N, Youssoufian H, et al. Cancer. 116(4 Suppl):1018-1026. 2010.
2. Kazlauskas A. Gene. 614:1-7. 2017.
3. Loizos N, Xu Y, Huber J, et al. Mol Cancer Ther. 4:369-379. 2005.
4. Stock P, Monga D, Tan X, et al. Mol Cancer Ther. 6(7):1932-1941. 2007.
5. Matei D, Emerson RE, Lai YC, et al. Oncogene. 25(14):2060-2069. 2006.
6. Youssoufian H, Amato RJ, Sweeney CJ, et al. J Clin Oncol. 26(15):suppl. 2008.
7. Wagner AJ, Kindler H, Gelderblom H, et al. Ann Oncol. 2017 28(3):541-546. 2017.
8. Pender A, Jones RL. Future Oncol. 13(24):2151-2157. 2017.
9. Tap WD, Wagner AJ, Schöffski P, et al. JAMA. 323(13):1266-1276. 2020.
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.