Anti-Human HER2 (Pertuzumab)

Anti-Human HER2 (Pertuzumab)

Product No.: H290

- -
- -
Product No.H290
Clone
2C4
Target
HER-2/neu
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
ERBB2, CD340, NGL, TKR1
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
N

- -
- -
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
Humanized antibody derived from mouse clone 2C4.
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
FA
N
IP
IF
FC
Antagonist
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 Pertuzumab. This product is for research use only. Pertuzumab binds specifically to human and cynomolgus HER2 near the center of extracellular domain II of the dimerization arm, adjacent to the binding pocket used for receptor dimerization.
Background
HER2 (ERBB2) is a member of the epidermal growth factor (EGF) family of receptor tyrosine kinases that regulate cell growth, survival and differentiation 1,2. HER2 activates downstream signaling pathways by forming a heterodimer with other ligand-bound EGF receptor family members (EGF receptor, HER3, HER4). Dysregulation of HER2 contributes to tumorigenesis in breast, ovarian, gastric, and other cancers 1. Additionally, HER2-HER3 heterodimers are potent signaling dimers required for HER2-mediated cancer cell proliferation 3.

Pertuzumab is a humanized monoclonal antibody used in the treatment of breast cancers that have either HER2 protein overexpression or ERBB2 gene amplification 2. Pertuzumab blocks HER2 function as a coreceptor by sterically inhibiting its heterodimerization with other HER family members, including EGF receptor, HER3, and HER4 3,4,5,6. As a result, HER2’s ability to activate pathways associated with cancer cell proliferation and survival is limited 2. Additionally, when pertuzumab binds to a cancer cell, antibody-dependent cellular cytotoxicity is triggered.

Pertuzumab is a full-length, chimeric IgG1 antibody generated by cloning VLκI and VHIII of murine 2C4 into a vector containing human kappa and CH1 domains 7. Pertuzumab was initially expressed and purified as a Fab from E. coli for residue optimization and subsequently was stably produced in Chinese hamster ovary cells.

Contact between pertuzumab and HER2 occurs at the HER2 heterodimerization interface 4 and is primarily made with the heavy chain of the antibody fragment, with a small contribution from the light chain 8. Additionally, Leu295 and His296 are important for binding.
Antigen Distribution
HER2 is ubiquitously expressed in epithelial, mesenchymal, and neuronal cells and their cellular progenitors. It is mostly localized to the plasma membrane and is generally excluded from clathrin-coated pits.
Ligand/Receptor
EGF receptors, SHC1, c-Src, Integrin B4, Grb2, SOS1, JAK2
NCBI Gene Bank ID
UniProt.org
Research Area
Biomarker
.
Biosimilars
.
Cancer
.
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 Pertuzumab biosimilars play a crucial role in pharmacokinetic (PK) bridging ELISA assays by serving as calibration standards and reference controls to ensure accurate quantification of drug concentrations in serum samples. This approach is essential for establishing bioequivalence between biosimilar candidates and the originator drug.

Calibration Standard Implementation

Research-grade Pertuzumab biosimilars are primarily used to create calibration curves that enable precise quantification of drug concentrations in biological samples. These biosimilars are calibrated against innovator drugs to ensure a high degree of accuracy in measurements. The calibration process involves creating a series of known concentrations that span the expected range of drug levels in clinical samples.

Several commercial ELISA kits utilize biosimilars that have been calibrated against International Standards from the National Institute of Biologicals and Control (NIBSC), providing standardization across different testing platforms. This international standardization ensures that measurements are consistent and comparable across different laboratories and studies.

Reference Control Functions

Biosimilar reference controls serve multiple quality assurance functions in PK bridging studies. They are used to validate assay performance through precision testing, where both intra-assay and inter-assay reproducibility are assessed using pools with low, medium, and high concentrations. The acceptance criteria typically require coefficient of variation (CV) values below 10% for low concentrations and below 5% for medium and high concentrations.

Bioanalytical Method Validation

The use of biosimilars in PK bridging ELISA follows stringent validation protocols based on ICH M10 and FDA bioanalytical method validation guidance. These methods undergo comprehensive seven-point validation procedures to ensure quality and robustness across different production lots. The validation process includes assessment of sensitivity, specificity, linearity, and matrix effects.

Modern approaches employ anti-idiotypic monoclonal antibodies in sandwich assay formats, which provide exceptional specificity and sensitivity in drug detection, even at low concentrations. This technology ensures that biosimilar standards can accurately represent the behavior of the originator drug in biological matrices.

Clinical Application Example

A practical implementation of this approach was demonstrated in a phase I biosimilarity study where KM118 (a Pertuzumab biosimilar) was compared against the reference drug Perjeta®. The study used ELISA methodology to determine Pertuzumab concentrations over an 84-day period, with biosimilarity established when the 90% confidence intervals for geometric mean ratios fell within the predefined range of 80.00-125.00%. The geometric mean AUC₀₋ₜ values showed that the test formulation (74,465.82 ng·h/mL) and reference formulation (69,097.83 ng·h/mL) met bioequivalence criteria with confidence intervals ranging from 99.92 to 114.72%.

Technical Specifications

Contemporary PK bridging assays typically operate within a dynamic concentration range of 100-25,000 ng/mL, with the lower limit of quantification set at 100 ng/mL for both analytes. Advanced methodologies may employ hybrid immunoaffinity capture liquid chromatography tandem mass spectrometry (LC-MS/MS) approaches that use biotinylated recombinant HER2 extracellular domain to simultaneously enrich both Pertuzumab and other therapeutic antibodies from human serum.

The acceptance criteria for analytical runs require that at least 75% of calibration standards, or at least 6 calibration standards including the lower and upper limits of quantification, must not deviate by more than ±20.0% (±25.0% at the lower limit) from their nominal values. This ensures consistent and reliable quantification throughout the analytical range.

Primary In Vivo Models for Anti-HER2/neu Antibody Studies

Research on anti-HER2/neu antibodies, such as trastuzumab, commonly employs two main types of animal models for in vivo tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization: syngeneic mouse models (with either murine or human HER2/neu) and humanized mouse models.

Syngeneic Mouse Models

Syngeneic models are immunocompetent mice that carry tumors derived from mouse cell lines, either overexpressing the mouse (rodent) neu/Erbb2 or a modified human HER2 (huHER2) construct. These models are the standard for studying the tumor-inhibitory effects of anti-HER2/neu antibodies and for characterizing TILs and adaptive immune responses, because they allow for the evaluation of therapy within a fully functional immune system.

  • Murine HER2/neu-expressing models (e.g., MMTV-neu, TUBO): Traditional models use mouse-derived neu/Erbb2, which mimics HER2+ tumor biology but is incompatible with human HER2-targeted antibodies.
  • Engineered huHER2 syngeneic models: To directly study human HER2-targeting antibodies (like trastuzumab), researchers have developed syngeneic mouse tumors expressing a truncated, non-immunogenic form of human HER2 (HER2T) in immunocompetent mice (e.g., BALB/c 4T1.2-HER2T). This allows for assessment of native anti-tumor immune responses, including TIL dynamics, following treatment with anti-HER2/neu antibodies.
  • Key features: These models support evaluation of both the direct anti-tumor effects (e.g., interference with HER2 signaling, antibody-dependent cellular cytotoxicity—ADCC) and the immune-mediated effects (e.g., T-cell activation, NK cell infiltration, and generation of immunological memory).

Humanized Mouse Models

Humanized models, which involve engrafting human tumors (patient-derived xenografts, PDX) into immunodeficient mice with a humanized immune system, are less commonly used for this type of research. They are primarily reserved for translational studies aiming to replicate the human immune microenvironment and evaluate human-specific effects of anti-HER2/neu antibodies.

  • Use case: Humanized models are particularly relevant when the research question involves interactions between human immune cells, human HER2, and therapeutic antibodies.
  • Limitation: These models do not fully recapitulate a functional murine immune system and are less suitable for studying the native, antigen-specific TIL response observed in syngeneic settings.

Model Comparison Table

Model TypeImmune SystemHER2/neu TargetTIL CharacterizationTypical Use Case
Syngeneic (murine HER2)Intact, endogenous mouseMouse neu/Erbb2YesFundamental tumor biology, ADCC, TILs
Syngeneic (huHER2T)Intact, endogenous mouseEngineered human HER2YesHuman HER2-targeted antibody TIL studies
HumanizedHuman immune componentsHuman HER2Partially (human TIL)Translational, human-specific questions

Mechanisms and Findings

  • Anti-HER2/neu antibodies inhibit tumor growth not only by blocking HER2 signaling but also by engaging Fc receptors (FcR) on immune cells, leading to ADCC and activation of both innate and adaptive immunity.
  • T-cell dependence: The therapeutic effect of anti-HER2/neu antibodies is T-cell dependent, and the generation of a robust anti-tumor immune response is critical for sustained tumor regression.
  • TIL dynamics: Both CD4+ and CD8+ T cells, NK cells, B cells, and dendritic cells infiltrate tumors in response to treatment, and their presence correlates with improved outcomes and immunological memory.
  • Model selection impact: Syngeneic models are essential for elucidating these immune mechanisms, whereas humanized models are best suited for validating human-specific aspects of therapy.

Conclusion

The primary in vivo models for administering research-grade anti-HER2/neu antibodies and characterizing TILs are syngeneic mouse models—either with murine HER2/neu or engineered human HER2 constructs in immunocompetent hosts. These models are preferred for mechanistic studies of tumor growth inhibition and immune infiltration. Humanized models are used secondarily, mainly for translational research where human-specific immune interactions are of interest.

Based on the provided search results, there appears to be a misunderstanding in your query. The research on pertuzumab biosimilars and immune checkpoint inhibitors represents two distinct areas of cancer treatment that are not typically combined in the way your question suggests.

Pertuzumab: A HER2-Targeted Therapy, Not a Checkpoint Inhibitor

Pertuzumab is not an immune checkpoint inhibitor - it is a HER2-targeted monoclonal antibody used specifically for treating HER2-positive breast cancer. The biosimilar studies for pertuzumab (including KM118 and QL1209) focus on demonstrating bioequivalence and safety compared to the reference drug Perjeta® in treating HER2-positive breast cancer patients.

The pertuzumab biosimilar research centers on:

  • Pharmacokinetic similarity to the reference drug
  • Efficacy in neoadjuvant treatment when combined with trastuzumab and chemotherapy
  • Safety profiles in breast cancer patients
  • Cost-effectiveness as an alternative to the expensive reference drug

Immune Checkpoint Inhibitor Combinations

The research on combining multiple checkpoint inhibitors involves entirely different therapeutic targets. Current combination strategies focus on agents like:

  • Anti-CTLA-4 (such as ipilimumab) combined with anti-PD-1/PD-L1 inhibitors (such as nivolumab)
  • These combinations target different mechanisms: anti-CTLA-4 primarily acts in lymph nodes to restore T cell activation, while anti-PD-1 acts at tumor sites to prevent T cell neutralization

The rationale for checkpoint inhibitor combinations is that targeting multiple immune pathways can overcome individual therapy limitations and enhance antitumor efficacy.

Why These Approaches Are Separate

Pertuzumab biosimilars and checkpoint inhibitor combinations represent fundamentally different treatment paradigms:

  • Pertuzumab targets HER2 protein overexpression in specific breast cancers
  • Checkpoint inhibitors modulate immune system responses across various cancer types

There is no evidence in the current literature suggesting researchers are combining pertuzumab biosimilars with checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 agents to study synergistic effects in immune-oncology models. These represent distinct therapeutic approaches for different cancer types and mechanisms.

A Pertuzumab biosimilar can be used as the capture and/or detection reagent in a bridging ADA (anti-drug antibody) ELISA to monitor a patient’s immune response against the therapeutic drug by exploiting its structural similarity to the reference antibody, ensuring that any ADAs generated against the therapeutic will also bind to the biosimilar.

In a typical bridging ELISA:

  • Biosimilar Pertuzumab is immobilized on the ELISA plate as the capture reagent. Patient serum is then added; if ADAs specific to Pertuzumab are present, they will bind to the immobilized biosimilar.
  • After washing, biosimilar Pertuzumab labeled with a detection marker (e.g., biotin, HRP) is added as the detection reagent. This detection reagent binds the other "arm" of the ADA, forming a “bridge” between the capture biosimilar and the detection biosimilar via the ADA.
  • The signal, typically chromogenic or luminescent, is proportional to the ADA concentration and is measured for quantification.

Key considerations:

  • Because the biosimilar is highly similar in sequence and structure to the originator, it provides equivalent epitope recognition, allowing detection of immune responses against either the biosimilar or the original drug.
  • Either the biosimilar or the originator can serve as capture/detection reagents depending on reagent availability and analytical validation; the same approach applies to both, provided analytical comparability has been established.

Additional details:

  • The bridging format is crucial for detecting bivalent antibodies (IgGs) but cannot distinguish between isotypes unless further modified.
  • Controls must demonstrate that the biosimilar’s binding profile mimics the originator to avoid assay artifacts or missed ADAs.
  • In practice, analogous bridging ELISAs have been used for other monoclonal antibodies and biosimilars, validating the approach for immunogenicity assessment.

In summary, a Pertuzumab biosimilar is used as both capture and detection reagent in a bridging ELISA due to its structural and antigenic comparability to the therapeutic, reliably enabling the monitoring of anti-drug antibody development in patients treated with either the reference or biosimilar formulation.

References & Citations

1. https://www.ncbi.nlm.nih.gov/gene/2064
2. Dean L, Kane M. Pertuzumab Therapy and ERBB2 Genotype. 2015 Sep 10 [Updated 2021 Jan 21]. In: Pratt VM, Scott SA, Pirmohamed M, et al., editors. Medical Genetics Summaries [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2012-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK315949/
3. Metzger-Filho O, Winer EP, Krop I. Clin Cancer Res. 19(20):5552-5556. 2013.
4. Franklin MC, Carey KD, Vajdos FF, et al. Cancer Cell. 5(4):317-328. 2004.
5. Hughes JB, Berger C, Rødland MS, et al. Mol Cancer Ther. 8(7):1885-1892. 2009.
6. Keating GM. Drugs. 72(3):353-360. 2012.
7. Adams CW, Allison DE, Flagella K, et al. Cancer Immunol Immunother. 55(6):717-727. 2006.
8. Roskoski R Jr. Pharmacol Res. 79:34-74. 2014.
9. Tanner M, Kapanen AI, Junttila T, et al. Mol Cancer Ther. 3(12):1585-1592. 2004.
10. Friess T, Scheuer W, Hasmann M. Clin Cancer Res. 11(14):5300-5309. 2005.
11. Nahta R, Yuan LX, Zhang B, et al. Cancer Res. 65(23):11118-11128. 2005.
12. Erjala K, Sundvall M, Junttila TT, et al. Clin Cancer Res. 12(13):4103-4111. 2006.
13. Arpino G, Gutierrez C, Weiss H, et al. J Natl Cancer Inst. 99(9):694-705. 2007.
14. Osipo C, Meeke K, Cheng D, et al. Int J Oncol. 30(2):509-520. 2007.
15. Sakai K, Yokote H, Murakami-Murofushi K, et al. Cancer Sci. 98(9):1498-1503. 2007.
16. Nagumo Y, Faratian D, Mullen P, et al. Mol Cancer Res. 7(9):1563-1571. 2009.
17. Scheuer W, Friess T, Burtscher H, et al. Cancer Res. 69(24):9330-9336. 2009.
18. Sak MM, Szymanska M, Bertelsen V, et al. Carcinogenesis. 34(9):2031-2038. 2013.
19. Yamashita-Kashima Y, Shu S, Harada N, et al. Oncol Rep. 30(3):1087-1093. 2013.
20. Zahnd C, Pecorari F, Straumann N, et al. J Biol Chem. 281(46):35167-35175. 2006.
21. Fábián Á, Horváth G, Vámosi G, et al. Cytometry A. 83(4):375-385. 2013.
Indirect Elisa Protocol
FA
N

Certificate of Analysis

- -
- -

Formats Available

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