Anti-Human IGF1R (CD221) (Teprotumumab) [Clone RG1507] — Fc Muted™

Anti-Human IGF1R (CD221) (Teprotumumab) [Clone RG1507] — Fc Muted™

Product No.: I-2215

- -
- -
Product No.I-2215
Clone
RG1507
Target
IGF1R (CD221)
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Insulin-like growth factor I receptor (IGF-I receptor), CD221, IGF-1R
Isotype
Human IgG1κ
Applications
ELISA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Muted
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 Teprotumumab. Teprotumumab, also known as RG1507, is a fully human monoclonal antibody that targets the insulin-like growth factor-1 receptor (IGF- 1R).
Background
The insulin-like growth factor-1 receptor (IGF1R) is a key receptor tyrosine kinase that regulates cell growth, proliferation, and survival, playing a critical role in cancer development and progression. Overexpression of IGF1R is commonly observed in various cancers, where it promotes tumor growth, transformation, motility, and metastasis. Targeting IGF1R has become a promising therapeutic approach, with strategies including the reduction of receptor expression, inhibition of kinase activity through small-molecule inhibitors, disruption of receptor function using monoclonal antibodies, and neutralization of its ligands. A deeper understanding of the downstream signaling pathways and IGF1R's role in different cancer types is essential for developing more effective treatments1,2.

Teprotumumab has shown significant potential, particularly in the treatment of thyroid eye disease (TED). Its effectiveness stems from its ability to inhibit IGF1R, which is linked to the thyrotropin receptor, a central factor in TED's pathology. However, teprotumumab therapy is associated with side effects such as hyperglycemia, hearing changes, fatigue, muscle spasms, hair loss, weight loss, gastrointestinal issues, menstrual irregularities, and infusion reactions. Recognizing and managing these adverse effects is crucial for optimizing teprotumumab’s use in clinical practice, ensuring a favorable balance between its risks and benefits3,4.
Antigen Distribution
IGF1R (Insulin-like Growth Factor 1 Receptor) is primarily found on the surface of human cells and is a transmembrane receptor activated by insulin-like growth factor 1 (IGF-1) and IGF-21. It is widely expressed in various tissues, including the brain, liver, muscle, and bone.
Ligand/Receptor
IGF1, IGF2, insulin
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cell Biology
.
Immunology
.
Opthamology
.
Thyroid Eye Disease

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 Teprotumumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs by serving as the analytical standard curve material, allowing drug concentration in serum samples to be quantified consistently across both biosimilar and originator contexts.

Key points explaining their application:

  • Single Analytical Standard Approach: Regulatory and industry best practice is to use a single PK assay with a single, well-characterized analytical standard—often the biosimilar itself—to quantify both the reference (originator) and biosimilar products in clinical samples. This improves precision and minimizes method variability compared to separate assays for each product.

  • Assay Calibration: The biosimilar (or sometimes originator drug) is prepared in serial dilutions to generate a standard curve (calibration curve) across the relevant concentration range. The mean absorbance values for these standards are plotted (typically on a semi-log or log-log graph), enabling interpolation of unknown sample concentrations. The same standard is used to quantify Teprotumumab in all assay samples.

  • Reference Controls: Both biosimilar and reference product are tested during assay validation for bioanalytical comparability—this involves measuring parallelism, accuracy, and precision to ensure the biosimilar behaves equivalently to the reference drug within the assay. Well-characterized samples (quality controls, or QCs) of both are analyzed to validate the standard curve and verify the method’s robustness.

  • Biosimilar as Reference Material: Research-use biosimilar antibodies are expressly manufactured and quality-controlled for such applications, serving either as calibrators (standards) or as reference controls within the ELISA, according to availability and validation data. Careful equivalence testing is required to confirm that either product can serve as the standard.

  • Quantitation in Serum: The typical ELISA protocol involves spiking known concentrations of Teprotumumab (biosimilar standard) into drug-naïve serum to build the standard curve, followed by measurement of unknown serum samples from preclinical or clinical studies. Unknown concentrations are interpolated using the best-fit curve (often with cubic spline or four-parameter logistic fitting).

  • Assay Validation Parameters: During PK assay validation, multiple runs are performed with independent sets of standards and QCs from both the biosimilar and reference drug to confirm linearity, accuracy, precision, and comparability under regulatory guidelines.

Summary Table—Use of Biosimilars in PK Bridging ELISA:

StepMaterial UsedPurpose/Role
Standard curveBiosimilar (lyophilized)Calibration standard for quantification
Reference/controlBiosimilar & originatorMethod validation, ensure assay equivalence
Sample analysisSerum samplesDrug concentration quantitation

This approach ensures accurate, reproducible measurement of Teprotumumab concentrations in serum for both PK bridging studies and routine bioanalysis, provided rigorous comparability of the biosimilar and reference is demonstrated in the assay system.

The primary in vivo models for studying anti-IGF1R (CD221) antibody-mediated tumor inhibition and characterization of tumor-infiltrating lymphocytes (TILs) are murine syngeneic tumor models and, less commonly, human xenograft models in immunodeficient mice.

Key model types:

  • Syngeneic murine models: These involve transplanting mouse tumors into immunocompetent mice of the same genetic background (e.g., CT-26, MC38, 4T1 in BALB/c or C57BL/6 mice), allowing for direct assessment of both tumor growth inhibition and immune response (including TILs).

  • Human xenograft models: Human tumors are implanted in immunodeficient mice (e.g., nude or SCID mice), permitting the study of anti-tumor activity of antibodies like anti-IGF1R with limited scope for immune characterization (since these mice lack functional lymphocytes).

  • Humanized mouse models: These are immunodeficient mice engrafted with human immune cells, enabling study of TILs alongside human tumor xenografts. However, while increasingly relevant for immunotherapy, use of anti-IGF1R antibodies in such models is less commonly reported in the literature compared to syngeneic setups.

Supporting details:

  • Syngeneic models (CT-26, MC38, 4T1) are frequently used for immunotherapy studies because they allow for robust immune cell profiling, including flow cytometric analysis of TILs following antibody administration (benchmarking commonly performed for checkpoint blockade, but the same experimental set-up applies to other antibodies including anti-IGF1R).

  • Example: Studies have characterized immune populations—including CD8+ T cells, regulatory T cells, and myeloid cells—after antibody treatment in these models, enabling pharmacodynamic analysis of immune modulation in response to therapy.

  • Human xenograft models using nude mice (e.g., RH-30 rhabdomyosarcoma in nude mice with anti-IGF1R antibody hR1) primarily characterize tumor growth inhibition but lack a functional immune system for TIL characterization.

  • For comprehensive TIL characterization, syngeneic mouse models remain the gold standard, as they possess a fully functional mouse immune system that allows detailed evaluation of immune cell infiltration and phenotyping within the tumor microenvironment post-antibody treatment.

  • Humanized models are used in immuno-oncology for dual characterization of anti-tumor activity and human TILs, but published use with research-grade anti-IGF1R antibodies remains rare.

Summary Table:

Model TypeTumor SourceMouse SystemTIL Characterization PossibleRelevant Citation
SyngeneicMouse tumorImmunocompetentYes (full profiling)
Human xenograftHuman tumorImmunodeficientNo (lacks mouse TILs)
Humanized xenograftHuman tumorHumanized immune sys.Yes (human TILs, less common)

In conclusion, syngeneic mouse models (e.g., CT-26, MC38, 4T1) are the primary system for in vivo studies of anti-IGF1R antibodies where both tumor growth inhibition and TIL characterization are required. Human xenograft and humanized models may be used for tumor inhibition studies but are less prevalent for detailed immune characterization with IGF1R-targeted agents.

Current published research does not specifically document the use of Teprotumumab biosimilars in combination with checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3) in immune-oncology models. Teprotumumab primarily targets the insulin-like growth factor 1 receptor (IGF-1R) and has been approved for the treatment of thyroid eye disease (TED), with its mechanism and benefits well characterized in autoimmune and fibrotic contexts, rather than in cancer immunotherapy.

Context from Current Literature

  • Teprotumumab Mechanism and Use:

    • Teprotumumab is a monoclonal antibody against IGF-1R, used in TED to inhibit pathogenic signaling in orbital fibroblasts and immune cells.
    • Its clinical data and research applications are almost exclusively within autoimmune and inflammatory diseases, particularly thyroid eye disease, not in oncology or combination immunotherapy studies.
  • Checkpoint Inhibitor Combination Strategies:

    • Checkpoint inhibitors such as anti-CTLA-4, anti-PD-1, and anti-LAG-3 are frequently combined in preclinical and clinical models to harness synergistic immune activation against tumors, with well-documented results in models of melanoma and other cancers.
    • The logic for combining different checkpoint inhibitors is based on their distinct mechanisms: CTLA-4 blockade primarily boosts T cell priming and expansion, while PD-1/PD-L1 and LAG-3 inhibitors work within the tumor microenvironment to sustain effector activity and prevent exhaustion.

Hypothetical Synergy and Preclinical Model Considerations

  • No direct studies combining Teprotumumab with checkpoint inhibitors in immune-oncology models were identified in the provided literature nor in current major reviews or clinical trial reports.
  • The theoretical basis for combination might stem from the role of IGF-1R in the tumor microenvironment, where IGF-1R signaling has been associated with immunosuppressive and proliferative effects. Inhibiting IGF-1R has been hypothesized to augment anti-tumor immunity, potentially synergizing with checkpoint blockade, but this remains speculative and untested in published immune-oncology preclinical models.
  • Translational hurdles include the distinct indications for these agents (autoimmunity vs. oncology), safety concerns, and species differences in IGF-1R pathway relevance.

Summary Table: Reported Use of Teprotumumab vs. Checkpoint Inhibitors in Immuno-Oncology Models

AgentIndication/TargetImmune-Oncology Evidence of Combination
TeprotumumabIGF-1R (Autoimmune: TED)None found
Anti-CTLA-4, Anti-LAG-3Immune checkpoints (Cancer: Multiple)Extensive, including synergistic models

Additional Notes

  • Researchers routinely combine checkpoint inhibitors (such as anti-CTLA-4 with anti-PD-1 or anti-LAG-3) to overcome resistance and increase response rates in complex immune-oncology models, both in preclinical and clinical settings.
  • The literature supports the potential synergy of combining targeted therapies with checkpoint inhibitors in cancer, but teprotumumab and its biosimilars have not been among the targeted agents studied for this purpose.
  • Any report or experiment involving teprotumumab biosimilars in cancer immunotherapy would be ground-breaking and novel, likely appearing in recent conference proceedings or trial registries not captured in the current peer-reviewed literature.

In summary: there are currently no published studies on the use of Teprotumumab biosimilars combined with immune checkpoint inhibitors to study synergies in cancer immunotherapy models. The intersection of these fields is speculative and would require new preclinical research investment.

A Teprotumumab biosimilar can be used as both the capture and detection reagent in a bridging ADA (anti-drug antibody) ELISA to monitor patient immune responses by exploiting its structural identity to the therapeutic drug. In this assay format, the same (or biosimilar) drug is utilized on both ends: one immobilized on the plate to capture ADA from the sample and one labeled for detection.

Essential context and technical details:

  • In a bridging ELISA for ADA detection, the assay typically involves:

    • Coating a microtiter plate with the biosimilar form of the therapeutic antibody (here, Teprotumumab or its biosimilar).
    • Adding the patient's serum; if anti-Teprotumumab antibodies (ADAs) are present, they will bind the immobilized drug.
    • Adding a detection reagent, which is Teprotumumab (or the biosimilar) labeled with an enzyme (such as HRP) or biotin.
    • If patient's serum contains ADA, these will bridge between the immobilized drug and the labeled drug, forming a "sandwich."
    • The detection is completed via an enzymatic or colorimetric reaction, yielding a measurable signal proportional to ADA concentration.
  • Why use a biosimilar as capture and detection reagent?

    • For immunogenicity assays, using a biosimilar ensures the detection reagents closely mimic the structure and epitopes of the therapeutic, providing specificity for ADAs that react to clinically relevant parts of the molecule.
    • Biosimilars are particularly advantageous when the original drug is costly or restricted in supply, or when detection reagent production benefits from minor modifications.
  • Key points for bridging ELISA with Teprotumumab biosimilar:

    • The ADA must be bivalent (able to bind two identical targets) for the "bridge" to form.
    • The assay detects antibodies regardless of subclass (IgG, IgM, etc.), though special configurations can target specific ADA classes.
    • Any ADA specific for epitopes present on both the therapeutic and biosimilar is detectable, whether neutralizing or non-neutralizing.

ADA monitoring in Teprotumumab therapy:

  • While published literature extensively describes bridging ELISA for other monoclonal antibodies, the principle is directly applicable to Teprotumumab and its biosimilars.
  • No source describes the Teprotumumab-specific ADA ELISA in detail; however, use of biosimilars as bridging reagents is standard practice for mAb immunogenicity, supporting the validity of this approach.

In summary, a Teprotumumab biosimilar is used in a bridging ADA ELISA by serving as the capture agent on the plate and as the labeled detection reagent, allowing sensitive and specific monitoring of ADAs against Teprotumumab in patient samples.

References & Citations

1. Park E, Park SY, Kim H, et al. J Pathol Transl Med. 2015;49(5):382-388.
2. Haisa M. J Int Med Res. 2013;41(2):253-264.
3. Yvon C, Khong JJ, Malhotra R, Patel BC. Teprotumumab. In: StatPearls. StatPearls Publishing; 2024. Accessed October 5, 2024. http://www.ncbi.nlm.nih.gov/books/NBK585036/
4. Mn S, Cc K. The Journal of clinical endocrinology and metabolism. 2023;108(9).
5. Teprotumumab Recombinant Monoclonal Antibody (MA5-41915). Accessed October 5, 2024. https://www.thermofisher.com/antibody/product/Teprotumumab-Antibody-Recombinant-Monoclonal/MA5-41915
Indirect Elisa Protocol

Certificate of Analysis

Formats Available

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