CALR Antibody Pair

Shipped with Ice Packs
In Stock

Product Specs

Buffer
**Capture Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
**Detection Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery timelines.
Notes
We recommend a capture antibody concentration of 0.3 µg/mL and a detection antibody concentration of 0.3 µg/mL. Optimal dilutions should be determined experimentally by the researcher.
Synonyms
CRT,Calreticulin ,CALR, CRTC
Target Names

Q&A

What is CALR and why are antibody pairs important in research?

Calreticulin (CALR) is a multifunctional calcium-binding chaperone that promotes protein folding and quality control in the endoplasmic reticulum. It interacts with monoglucosylated glycoproteins via the calreticulin/calnexin cycle . CALR antibody pairs are crucial for quantitative measurement of CALR protein levels in biological samples through techniques like ELISA.

The significance of CALR antibody pairs has increased substantially since the 2013 discovery that CALR mutations are present in approximately 25% of essential thrombocythemia (ET) and 35% of myelofibrosis (MF) cases . These mutations create a novel C-terminus that aberrantly activates the thrombopoietin receptor (TPOR/MPL), driving malignant hematopoietic stem cell proliferation. Antibody pairs enable precise detection of both wild-type and mutant CALR forms, facilitating diagnosis, research, and therapeutic development.

How do CALR antibody pairs function in immunoassay applications?

CALR antibody pairs typically operate through a sandwich-based detection system that requires two antibodies recognizing different epitopes:

  • Capture antibody: Usually an unlabeled antibody (monoclonal or polyclonal) that is immobilized on a solid phase surface, such as a 96-well microtiter plate. This antibody binds and immobilizes CALR from the sample.

  • Detection antibody: Typically biotinylated or directly conjugated to a reporter molecule (e.g., horseradish peroxidase or fluorophore). This antibody binds to a different epitope on the captured CALR protein.

In ELISA applications, antibody pairs like those described in product documentation demonstrate the following specifications:

ComponentCharacteristicsTypical AmountApplication
Capture antibodyMouse monoclonal anti-CALR, IgG1 Kappa100 μgELISA, sufficient for 3-5 plates
Detection antibodyRabbit polyclonal anti-CALR (biotinylated)50 μgELISA, sufficient for 3-5 plates
StandardRecombinant human CALR protein10 μgGeneration of standard curve

The typical working range for CALR ELISA using these antibody pairs is 6.24-400 ng/mL . The sensitivity and specificity of the assay depend on the quality of the antibody pair and optimization for the specific sample type.

How should researchers optimize CALR antibody pair conditions for experimental applications?

Optimizing CALR antibody pair conditions requires systematic evaluation of multiple parameters:

  • Antibody concentrations: Perform checkerboard titrations to determine optimal concentrations for both capture and detection antibodies. Starting with manufacturer recommendations (often 1-5 μg/mL for capture antibodies), test serial dilutions to identify the combination that provides the highest signal-to-noise ratio.

  • Blocking conditions: Evaluate different blocking agents (BSA, milk protein, commercial blockers) to minimize background while maintaining specific signal. A 2-5% concentration of blocking agent in PBS or TBS is typically effective.

  • Sample dilution: Test various sample dilutions to ensure measurements fall within the linear range of the assay. Cell lysates often require 1:5 to 1:20 dilution, while serum/plasma samples may need 1:50 to 1:200 dilution.

  • Incubation parameters: Optimize incubation times and temperatures for antigen capture (typically 1-2 hours at room temperature or overnight at 4°C) and detection antibody binding (usually 1-2 hours at room temperature).

  • Washing protocol: Develop a consistent washing protocol (typically 3-5 washes) to remove unbound reagents without disturbing specific interactions.

Researchers should document optimization parameters and include appropriate controls, as antibody performance can vary between lots and applications.

What are the key controls necessary when working with CALR antibody pairs?

Implementing proper controls is essential for generating reliable data with CALR antibody pairs:

Control TypePurposeImplementation
Positive controlsVerify assay functionalityRecombinant CALR protein; cell lines with known CALR expression
Negative controlsAssess background/non-specific bindingBuffer only; CALR-knockout samples; isotype control antibodies
Standard curveQuantification referenceSerial dilutions of recombinant CALR (6.24-400 ng/mL)
Specificity controlsConfirm antibody specificityPre-adsorption with CALR peptide; irrelevant protein controls
Sample matrix controlsAssess matrix interferenceSpike recovery in relevant matrices; dilution linearity
Technical replicatesEvaluate technical variationDuplicate/triplicate samples; inter-plate calibrators
Mutation-specific controlsValidate mutation detectionWild-type CALR; Type 1 mutant (52bp deletion); Type 2 mutant (5bp insertion)

For mutation-specific applications, it is particularly important to include both wild-type CALR and known mutant CALR samples to verify the specificity of detection. When evaluating therapeutic antibodies, functional controls demonstrating inhibition of CALR-TPOR interaction should be included .

How should samples be prepared for optimal CALR detection?

Proper sample preparation is critical for accurate CALR detection:

  • Cell lysate preparation:

    • Wash cells with cold PBS to remove media components

    • Lyse cells using a buffer containing: 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1% NP-40/Triton X-100, with protease inhibitors

    • Incubate on ice for 30 minutes with occasional vortexing

    • Centrifuge at 14,000 × g for 15 minutes at 4°C

    • Collect supernatant and determine protein concentration

  • Considerations for mutant CALR detection:

    • Mutant CALR is often expressed on the cell surface due to loss of the KDEL ER-retention motif

    • For flow cytometry applications, surface staining can be performed without permeabilization

    • For detection of both surface and intracellular CALR, a two-step staining protocol may be optimal

  • Storage considerations:

    • Store samples at -20°C or -80°C in aliquots to avoid freeze-thaw cycles

    • For long-term storage of antibody reagents, temperatures below -20°C are recommended

    • Glycerol (50%) is often used in antibody storage buffers to prevent freeze-thaw damage

The optimal sample preparation method depends on the specific application and should be validated for each experimental system.

How can antibody pairs distinguish between wild-type and mutant CALR?

Detection of mutant CALR requires specialized approaches due to the frameshift mutations that create a novel C-terminus:

  • Neoepitope-specific antibodies: Antibodies like 4D7 and INCA033989 specifically target the mutant CALR neoepitope, allowing direct identification of mutated proteins . These antibodies show no binding to wild-type CALR, enabling highly specific detection.

  • Molecular weight differentiation: Wild-type CALR has a molecular weight of approximately 48 kDa, while Type 1 mutant CALR (52bp deletion) is approximately 36 kDa and Type 2 mutant CALR (5bp insertion) is approximately 47 kDa. This size difference can be exploited in Western blot analysis .

  • Subcellular localization: Wild-type CALR primarily localizes to the endoplasmic reticulum due to its KDEL retention motif, while mutant CALR shows increased surface expression. Flow cytometry using non-permeabilized cells can exploit this difference .

  • Differential binding partners: Mutant CALR aberrantly binds to the thrombopoietin receptor (TPOR/MPL), which wild-type CALR does not. Co-immunoprecipitation followed by detection can distinguish based on binding partners .

For comprehensive detection, researchers often combine antibody-based methods with genomic analysis, as each provides complementary information about CALR mutation status.

What therapeutic approaches use antibodies targeting mutant CALR?

Several innovative therapeutic strategies targeting mutant CALR are under development:

  • Blocking antibodies: Monoclonal antibodies like INCA033989 antagonize mutated CALR-driven signaling by preventing binding to the thrombopoietin receptor. In preclinical studies, these antibodies effectively prevented thrombocytosis and megakaryocyte accumulation in bone marrow .

  • Mechanism of action: Anti-mutant CALR antibodies function by:

    • Disrupting the constitutive association between mutant CALR and TPOR

    • Blocking TpoR tyrosine phosphorylation and downstream STAT/ERK signaling

    • Promoting dynamin-dependent endocytosis of the antibody/mutCALR/TPOR complex

    • Inhibiting TPO-independent megakaryocyte differentiation

  • Clinical development: The 4D7 monoclonal antibody has shown promising results in preclinical studies, including:

    • Inhibition of cytokine-independent growth in cell lines expressing mutant CALR

    • Blockade of constitutive factor-independent phospho-STAT5 and phospho-ERK signaling

    • Effectiveness against both Type 1 and Type 2 CALR mutations

  • Advanced approaches: Emerging strategies include:

    • Bispecific antibodies targeting both MPL and mutant CALR

    • Antibody-toxin conjugates for increased efficacy

    • CAR-T cells directed against the mutant CALR neoepitope

    • Therapeutic vaccines derived from mutant CALR peptides

These approaches represent a significant advancement over current treatments like JAK inhibitors, which do not effectively target the underlying mutational drivers of the disease.

What challenges exist in developing antibodies against the mutant CALR neoepitope?

Developing antibodies against the mutant CALR neoepitope presents several technical challenges:

  • Epitope complexity: The neoepitope contains an unusual positively charged sequence with high arginine content, creating challenges for immunogen design and antibody generation .

  • Specificity requirements: Antibodies must precisely distinguish between wild-type and mutant CALR despite substantial sequence similarity outside the neoepitope region.

  • Secreted versus cell-bound forms: Mutant CALR is both secreted and cell-bound, potentially creating a "decoy" effect where secreted protein captures therapeutic antibodies before they can reach cell-bound targets .

  • Mutation heterogeneity: While Type 1 (52bp deletion) and Type 2 (5bp insertion) mutations create similar neoepitopes, subtle differences may affect antibody binding affinity and functional inhibition.

  • Validation challenges: Limited availability of patient samples with different CALR mutation types complicates comprehensive validation of antibody specificity and efficacy.

Despite these challenges, successfully developed antibodies like 4D7 have demonstrated the feasibility of targeting the mutant CALR neoepitope, with dissociation constants in the nanomolar range (Kd ~1.53 nM) .

How do post-translational modifications affect CALR antibody detection?

Post-translational modifications (PTMs) can significantly impact CALR antibody binding:

  • Glycosylation effects: CALR contains potential N-glycosylation sites that can mask epitopes or alter protein conformation. Antibodies raised against E. coli-expressed recombinant CALR (non-glycosylated) may show different binding patterns compared to native glycosylated CALR.

  • Phosphorylation considerations: CALR contains multiple phosphorylation sites that can change during cellular stress and disease states. Phosphorylation near antibody epitopes may enhance or inhibit binding.

  • Oxidation sensitivity: CALR contains multiple cysteine residues susceptible to oxidation. Oxidative stress can alter CALR conformation through disulfide bond formation, potentially affecting epitope accessibility.

  • PTM differences between wild-type and mutant CALR: Mutant CALR may have altered PTM patterns due to mislocalization. Surface-exposed mutant CALR may undergo different modifications than ER-resident wild-type CALR.

Researchers should consider these factors when designing experiments and interpreting results, particularly when comparing data across different sample types or experimental conditions.

How can CALR antibody pairs be used in biomarker development?

CALR antibody pairs offer valuable tools for biomarker development in several contexts:

  • Prognostic assessment: Studies using CALR antibody pairs have demonstrated that CALR expression levels correlate with relapse-free survival in various cancers . Quantitative ELISAs using optimized antibody pairs can measure CALR levels in patient samples.

  • Diagnostic applications: Mutant-specific CALR antibodies can distinguish between JAK2-negative ET/MF cases with CALR mutations (~70% of cases) versus those with other genetic drivers . This distinction has important therapeutic implications.

  • Therapeutic monitoring: Measuring circulating mutant CALR using specific antibody pairs can potentially monitor treatment response to targeted therapies. Decreasing levels may indicate effective therapeutic activity.

  • Minimal residual disease (MRD) detection: High-sensitivity antibody-based assays targeting mutant CALR could potentially detect small populations of residual malignant cells following treatment, though further validation is needed.

  • Patient stratification: Different CALR mutation types (Type 1 vs. Type 2) are associated with different clinical outcomes. Antibody-based methods that can distinguish mutation types could help stratify patients for appropriate treatment approaches.

As antibody-based therapeutics targeting mutant CALR enter clinical development, companion diagnostics using these antibody pairs will become increasingly important for patient selection and monitoring.

What future directions are emerging for CALR antibody pair applications?

The field of CALR antibody research is rapidly evolving, with several promising future directions:

  • Single-cell analysis: Integration of CALR antibody-based detection with single-cell technologies will enable more precise characterization of heterogeneity within CALR-mutated cell populations.

  • Multiplexed detection systems: Development of multiplexed assays combining CALR mutation detection with other MPN-associated markers (JAK2, MPL) will provide more comprehensive patient profiling.

  • Enhanced therapeutic antibodies: Engineering of next-generation anti-CALR antibodies with improved properties:

    • Increased affinity for neoepitopes

    • Enhanced tissue penetration

    • Reduced immunogenicity

    • Optimized effector functions

  • Bispecific approaches: Creation of bispecific antibodies targeting both mutant CALR and immune effector cells (T cells, NK cells) to enhance therapeutic efficacy through immune recruitment .

  • Biomarker integration: Development of integrated biomarker profiles combining genomic, proteomic, and functional assays based on CALR antibody pairs to better predict patient outcomes and treatment responses.

  • Point-of-care diagnostics: Adaptation of CALR antibody pair technologies into rapid, point-of-care testing formats for more accessible clinical application, particularly in settings where advanced molecular testing is unavailable.

These emerging applications highlight the continuing importance of high-quality CALR antibody pairs in both research and clinical settings.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.