Anti-Human von Willebrand factor (vWF) (Caplacizumab) [Clone ALX-0081]

Anti-Human von Willebrand factor (vWF) (Caplacizumab) [Clone ALX-0081]

Product No.: V30

- -
- -
Product No.V30
Clone
ALX-0081
Target
von Willebrand Factor (vWF)
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
F8VWF, VWD, Coagulation factor VIII
Isotype
Human VHH
Applications
B
,
ELISA
,
FA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Baboon
Cynomolgus Monkey
Human
Host Species
Hamster
Expression Host
CHO Cells
Immunogen
Recombinant A1 domain of vWF
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
≤ 1.0 EU/mg as determined by the LAL method
Purity
>95% monomer by analytical SEC
≥95% by SDS Page
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 ?
B,
ELISA,
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 Caplacizumab. ALX-0081 (Caplacizumab) is a humanized single- variable domain, bivalent immunoglobulin that targets the A1-domain of the ultra-large von Willebrand factor (vWF). ALX-0081 (Caplacizumab) cross-reacts with cynomolgus monkey and baboon vWF but not murine vWF.
Background
von Willebrand factor (vWF) is a plasma glycoprotein involved in clot formation via the adhesion of platelets to subendothelial collagen1,2. A severe deficiency of the vWF-cleaving metalloproteinase ADAMTS13 leads to acquired thrombotic thrombocytopenic purpura (aTTP), a rare autoimmune blood clotting disorder caused by anti-ADAMTS13 autoantibodies2. Reduced ADAMTS13 activity leads to the accumulation of ultra-large vWF multimers in the blood. These bind to platelets, leading to the formation of platelet-rich microthrombi in the microvasculature. These microvascular occlusions ultimately lead to thrombocytopenia, hemolytic anemia, tissue ischemia, and organ dysfunction. ADAMTS13 activity can be normalized via a combination of plasma exchange, immunosuppression, and immunotherapy.

ALX-0081 (Caplacizumab) was developed as a treatment for thrombosis in high-risk patients with acute coronary syndrome, including treatment of aTTP2. Caplacizumab was generated as a single-domain antibody fragment and consists of two identical, humanized building blocks linked by a 3-alanine linker2,3. The precursor of Caplacizumab was isolated from a llama immunized with the recombinant A1 domain of vWF, humanized, and used to form a monovalent vWF-binding Nanobody (PMP12A2h1)3. The bivalent form was subsequently produced in Escherichia coli using a secretory system.

Caplacizumab inhibits the interaction between vWF and the platelet glycoprotein Ib-IX-V receptor, preventing platelet adhesion at high shear rates like those observed in stenosed arteries2,3. Administration of caplacizumab in patients leads to rapid suppression of ristocetin cofactor, a biomarker for vWF activity, as well as reductions in VWF antigen and factor VIII.

Antigen Distribution
vWF is found in blood plasma, the subendothelial matrix, storage granules of endothelial cells, platelets, and megakaryocytes. vWF can also be found in macrophages, where it is bound and endocytosed during clearance.
Ligand/Receptor
Platelet glycoprotein Ib-IX-V receptor, coagulation factor VIII
NCBI Gene Bank ID
UniProt.org
Research Area
Autoimmune
.
Biosimilars
.
Cardiovascular Disease
.
Immunology
.
Blood Coagulation

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 Caplacizumab biosimilars are used as calibration standards or reference controls in PK bridging ELISAs by preparing standard curves with known concentrations of the biosimilar, against which unknown serum sample concentrations are quantified. This approach ensures consistency and scientific rigor in measuring Caplacizumab levels in clinical and preclinical samples.

Context and Supporting Details:

  • Calibration Standard Preparation: The biosimilar is serially diluted in pooled human serum to generate a set of calibration standards, typically spanning a range of concentrations (e.g., 0, 31.25, 62.5, 125, 250, 500, 1000, 2000 ng/mL). These standards are critical for generating a calibration curve, which enables quantification of Caplacizumab in unknown samples by interpolation.

  • Bridging ELISA Principle: In a PK bridging ELISA, two capture/detection reagents (often anti-drug antibodies) “bridge” the Caplacizumab molecule in patient serum. The signal intensity generated is proportional to the concentration of Caplacizumab present. The calibration curve (generated using the biosimilar standard) allows this signal to be translated into actual drug concentrations in test samples.

  • Rationale for Using Biosimilars: Research-grade biosimilars are employed instead of clinical-grade reference products due to availability, cost, and control over lot variability. Before biosimilars are used as standards, assay validation studies compare the analytical equivalence of the biosimilar and reference standard within the assay (evaluating accuracy, precision, and linearity). This assures regulators that both molecules behave indistinguishably in the assay system.

  • Validation and Controls: Control samples (Quality Controls, QCs) are prepared with both the biosimilar standard and the reference product at several concentrations to verify that the assay reliably measures drug levels across the expected range. Assay validation measures parameters such as specificity, sensitivity, reproducibility, and lack of interference, using both biosimilar and reference product spiked controls.

  • Advantages of a Single Standard: Using a single, well-qualified biosimilar as the analytical standard in the ELISA reduces inter-assay variability, simplifies validation, and is currently considered best practice in PK bridging studies for biosimilars. The approach streamlines bioanalytical comparability and regulatory submission, provided comprehensive comparability data are generated and accepted by regulatory authorities.

Summary Table: Caplacizumab Biosimilar Role in PK Bridging ELISA

StepDescription
Calibration StandardSerial dilutions of biosimilar prepared in serum
Reference/QC ControlsPrepared with both biosimilar and reference product
Assay SystemBridging ELISA with anti-drug antibodies
QuantificationUnknown serum concentrations are interpolated from the standard curve
Regulatory ExpectationDemonstrate analytical equivalence of biosimilar and reference standard

Research-grade Caplacizumab biosimilars are thus essential for robust, regulatory-compliant PK assays, provided their equivalency with the reference is thoroughly demonstrated through method validation.

The primary in vivo models for studying the effects of anti-von Willebrand Factor (vWF) antibodies on tumor growth inhibition and the characterization of tumor-infiltrating lymphocytes (TILs) are mainly syngeneic mouse models and, more recently, humanized mouse models.

Syngeneic mouse models:

  • These models involve transplanting tumor cells from a mouse of a defined genetic background into immunocompetent mice of the same strain, thereby preserving immune function for the study of TILs.
  • For example, the ret transgenic mouse model of malignant melanoma has been used to investigate the role of VWF. Blocking VWF in this model was associated with tumor weight reduction and survival benefit.
  • Syngeneic models allow for the functional analysis of tumor-infiltrating lymphocytes in the context of an intact murine immune system, which is ideal for characterizing TILs after anti-vWF antibody treatment.
  • Orthotopic tumor models (e.g., implantation into tissue of origin) have also been used for metastasis studies involving VWF.

Humanized mouse models:

  • These models have been genetically engineered to express human genes—in this context, human VWF and human GPIbα—to overcome species differences that complicate the study of VWF-targeted therapeutics.
  • A recent example is the "fully humanized von Willebrand disease type 1 mouse model" (hVWD1 mice). This model expresses human vWF and has been used for in vivo testing of therapeutic antibodies targeting vWF, including a bispecific antibody that bridges VWF to albumin.
  • Humanized models provide more translational relevance for antibodies that are specifically designed to target human vWF, but immune reconstitution (for proper TIL analysis) varies depending on whether the model includes a human immune compartment.

Additional Supporting Details

  • There is limited evidence specifically on the use of research-grade anti-vWF antibodies in published tumor immunology studies. Most work to date has focused on genetic knockout or pharmaceutical inhibition (e.g., via heparins or recombinant proteins) of VWF rather than using anti-VWF antibodies directly.
  • The capacity to characterize TILs depends on the immune competency of the model:
    • Syngeneic models support robust TIL profiling.
    • Humanized models expressing human immune components (e.g., humanized NSG mice engrafted with human immune cells) would be optimal for studies requiring human antibody specificity and TIL analysis.

Summary Table: Model Comparison

Model TypevWF Target OriginImmune SystemAntibody CompatibilityTIL Characterization
Syngeneic (mouse)Mouse vWFMouse (intact)Mouse-reactive AbsExcellent
Humanized (partial)Human vWFMouseHuman-reactive AbsLimited
Humanized (full)Human vWFHuman (reconstituted)Human-reactive AbsExcellent
  • Research targeting tumor growth and TILs with anti-vWF antibodies is thus most actively performed in syngeneic mouse models, with humanized models increasingly used to accommodate human-specific therapeutics and more translational characterization.
  • The choice of model depends on the antibody's species specificity and the complexity of immune responses required for TIL analysis.

Researchers use Caplacizumab biosimilar mainly as a research-grade tool to target von Willebrand Factor (VWF), but its use in immune-oncology models in direct conjunction with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars is not prominently documented in the current literature. Most published research involving Caplacizumab focuses on its role in thrombotic disorders, particularly immune-mediated thrombotic thrombocytopenic purpura (iTTP), often combined with immunosuppressants like corticosteroids and rituximab—not in cancer immunotherapy settings with checkpoint inhibition.

Key context and supporting details:

  • Caplacizumab biosimilar is a monoclonal antibody that specifically detects human VWF, and research-grade versions are available for in-vitro and in-vivo studies but are not approved for clinical oncology use.
  • In immune-oncology, researchers predominantly study the synergistic effects of different checkpoint inhibitors (e.g., PD-1, CTLA-4, and LAG-3) to optimize anti-tumor immune responses. Combination therapies like anti-PD-1/CTLA-4 or anti-PD-1/LAG-3 have demonstrated distinct mechanisms of action and synergistic potential in activating different T-cell populations and overcoming immune resistance in tumor models.

Typical experimental design for combination checkpoint inhibitor studies:

  • Researchers use highly controlled animal models or cell cultures (e.g., mouse models of melanoma) to treat with combinations of checkpoint inhibitors.
  • The synergy is interrogated by analyzing tumor growth inhibition, immune cell activation (CD4, CD8 T cells, Tregs), cytokine profiles, and overall survival. Studies have shown that combining inhibitors leads to different immune cell activation patterns depending on the agents involved.
  • Mechanistic studies often track immune cell subsets via flow cytometry and use gene/protein expression profiling to identify changes unique to each combination regimen.

Role and limitations of Caplacizumab biosimilars:

  • Although research-grade Caplacizumab biosimilars can be used to study VWF function and related platelet-immunity interactions, there is no substantive evidence in current peer-reviewed research supporting their combined use with immune checkpoint inhibitors for synergistic immune-oncology investigation.
  • Caplacizumab’s mechanism centers around inhibiting VWF-mediated platelet adhesion, not directly modulating T-cell or checkpoint pathways.
  • Existing immune-oncology studies focus on checkpoint inhibitors that target T-cell activation and exhaustion pathways, not platelet function or VWF.

Summary table: Use of Caplacizumab biosimilar vs. checkpoint inhibitors

Agent TypeMechanismTypical Oncology UseSynergy Evidence
Caplacizumab biosimilarBlocks VWF, affects plateletsiTTP, thrombosis modelsNot established
Anti-CTLA-4 biosimilarBlocks CTLA-4, restores T-cell activationCancer immunotherapy modelsWell-documented
Anti-LAG-3 biosimilarBlocks LAG-3, enhances T-cell activationCancer immunotherapy, research modelsWell-documented

There is currently no validated approach for using Caplacizumab biosimilars in combination with checkpoint inhibitors to study synergistic effects in complex immune-oncology models, based on available research. Researchers investigating synergy typically focus on combinatorial checkpoint blockade targeting T-cell functions, not platelet or VWF pathways.

A Caplacizumab biosimilar can be used as a capture or detection reagent in a bridging ADA ELISA to monitor anti-drug antibodies (ADAs) that patients may develop against the therapeutic drug during treatment.

Key steps and context:

  • Bridging ADA ELISA Principle:
    This assay detects bivalent ADAs in patient serum by leveraging the fact that these antibodies can simultaneously bind two identical drug molecules (in this case, Caplacizumab or its biosimilar).

  • Role of Biosimilar as Reagent:

    • The Caplacizumab biosimilar is typically labeled (e.g., with biotin for capture or with HRP/dye for detection).
    • One population of biosimilar molecules is immobilized on the ELISA plate (as the “capture” reagent). Patient serum is then added; any anti-Caplacizumab antibodies present bind to the surface-bound molecule.
    • After washing, a second biosimilar molecule—labeled for detection—is added. If ADA is present, it bridges the immobilized biosimilar and the labeled biosimilar, allowing for signal development (usually via HRP and chromogenic substrate).
  • Why use a biosimilar?

    • A biosimilar with matched structure and epitope fidelity to Caplacizumab is usually interchangeable with the originator in ADA assays, provided it is analytically validated for equal or greater sensitivity and specificity.
    • It ensures reagent availability and may help avoid interference or limitation if the originator is in short supply.

Clinical Relevance:

  • The resulting signal directly correlates with ADA presence and titer, allowing for sensitive monitoring of patient immunogenicity against the therapeutic.
  • Such assays are widely validated for biotherapeutics and monoclonal antibodies.
  • Using a biosimilar as the reagent does not affect the fundamental assay principle as long as quality and functional similarity are confirmed.

Technical considerations:

  • Matrix effects (complex serum composition) require careful assay validation for specificity and sensitivity.
  • Choice of label (HRP, biotin) and blocking buffers should be optimized to lower background and increase drug tolerance.
  • Assays typically compare response rates for biosimilar and originator across head-to-head studies for regulatory submissions and clinical safety assurance.

In summary, a Caplacizumab biosimilar can effectively serve as a capture or detection agent in bridging ADA ELISA to detect anti-Caplacizumab antibodies in patient serum, provided its analytical qualities match that of the originator and the assay is properly validated.

References & Citations

1 Lenting PJ, Christophe OD, Denis CV. Blood. 125(13):2019-2028. 2015.
2 Duggan S. Drugs. 78(15):1639-1642. 2018.
3 Ulrichts H, Silence K, Schoolmeester A, et al. Blood. 118(3):757-765. 2011.
4 Peyvandi F, Scully M, Kremer Hovinga JA, et al. N Engl J Med. 374(6):511-522. 2016.
B
Indirect Elisa Protocol
FA

Certificate of Analysis

Formats Available

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