LCR62 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Antibodies

Antibodies are proteins produced by the immune system in response to foreign substances. They are crucial for identifying and neutralizing pathogens and are widely used in medical research and diagnostics. Given the lack of specific information on "LCR62 Antibody," we will discuss general principles of antibody research and how they might apply to any newly discovered or less-studied antibodies.

Understanding Antibody Structure and Function

Antibodies, or immunoglobulins, consist of two heavy chains and two light chains. They bind to specific antigens through their variable regions, which are highly diverse and allow for the recognition of a vast array of pathogens. The specificity and affinity of an antibody for its antigen are critical for its effectiveness in therapeutic or diagnostic applications.

Research Findings on Similar Antibodies

While there is no specific data on "LCR62 Antibody," research on other antibodies provides valuable insights into their development and application:

  • L-Selectin/CD62L Antibody: This antibody targets L-Selectin, a cell surface glycoprotein involved in leukocyte adhesion and homing. It is used in research to study immune cell interactions and has applications in diagnostics for conditions involving immune dysregulation .

  • Broadly Neutralizing Antibodies: These are antibodies that can neutralize multiple strains of a virus, such as influenza or HIV. They are of great interest for developing universal vaccines or treatments .

Development and Application of Monoclonal Antibodies

Monoclonal antibodies are produced by a single clone of cells and are highly specific to their target. Techniques like RAMIHM allow for the rapid development of fully human monoclonal antibodies, which can be used against a variety of targets, including viral antigens and cancer markers .

Data Table Example: Characteristics of Monoclonal Antibodies

AntibodyTargetApplicationSource
L-Selectin/CD62LL-SelectinImmune cell studies, diagnosticsMouse IgG 2A
CR6261Influenza HA stemBroadly neutralizing antibodyHumanized mice
RAMIHM-generatedSARS-CoV-2 RBDNeutralizing antibodiesHumanized mice

This table illustrates how different antibodies are developed and applied, which could serve as a model for studying any newly identified antibody like "LCR62 Antibody."

References:

  1. R&D Systems. (2024). Human L-Selectin/CD62L Antibody MAB728-100.

  2. PubMed. (2005). The application of a monoclonal antibody to CD62L on paraffin.

  3. Nature. (2012). Structural and genetic basis for development of broadly neutralizing influenza antibodies.

  4. PMC. (2023). RAMIHM generates fully human monoclonal antibodies by rapid mRNA immunization of humanized mice and BCR-seq.

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
LCR62 antibody; At2g29045 antibody; T9I4 antibody; Putative defensin-like protein 170 antibody; Putative low-molecular-weight cysteine-rich protein 62 antibody; Protein LCR62 antibody
Target Names
LCR62
Uniprot No.

Target Background

Database Links

KEGG: ath:AT2G29045

STRING: 3702.AT2G29045.1

UniGene: At.64378

Protein Families
DEFL family
Subcellular Location
Secreted.

Q&A

What is ICR62 and what is its primary target in research applications?

ICR62 is a rat monoclonal antibody specifically targeting the epidermal growth factor receptor (EGFR). It functions by effectively blocking the binding of multiple EGFR ligands including EGF, transforming growth factor (TGF)-alpha, and HB-EGF to the receptor. This blocking mechanism inhibits the growth of tumor cell lines that overexpress EGFR in vitro and has demonstrated capability to eradicate such tumors when grown as xenografts in athymic mice .

The antibody has been evaluated in clinical settings, particularly for patients with squamous cell carcinomas where EGFR overexpression is common. Phase I clinical trials have demonstrated that ICR62 can be safely administered to patients and effectively localizes to metastatic tumor sites even at relatively low doses .

How are monoclonal antibodies like ICR62 typically purified for research applications?

Research-grade monoclonal antibodies undergo multi-step affinity chromatography purification methods to ensure high specificity and purity. The specific technique varies based on the antibody's species of origin and isotype. For rat monoclonal antibodies like ICR62, Protein A or Protein G chromatography is commonly employed depending on the specific isotype characteristics .

The purification process typically includes:

  • Initial capture using affinity chromatography (Protein A/G)

  • Additional purification steps to remove non-specific proteins

  • Buffer exchange to stabilize the antibody (commonly phosphate-buffered saline)

  • Quality control testing for purity, specificity, and activity

  • Formulation with stabilizers like BSA (bovine serum albumin) and preservatives

For example, purified antibody preparations are often formulated in solutions containing 0.01 M phosphate buffered saline (150 mM NaCl, pH 7.4), 1% BSA, and 0.09% sodium azide as a preservative to maintain stability and activity .

What are the recommended storage and handling conditions for maintaining ICR62 antibody activity?

To maintain optimal activity and stability, monoclonal antibodies like ICR62 should be stored under controlled conditions:

  • Temperature: Store at 2-8°C (refrigerated)

  • Avoid freezing, as freeze-thaw cycles can denature antibodies

  • Protect from prolonged exposure to light

  • Maintain sterility of the preparation

  • Follow manufacturer's recommendations for shelf life

When using the antibody, researchers should aliquot the stock solution to avoid repeated freeze-thaw cycles and minimize contamination risk. The working concentration should be determined for each specific application through titration experiments. For flow cytometry applications of similar antibodies, concentrations of ≤0.25 μg per 10^6 cells in a volume of 100 μl are typically recommended .

How should researchers determine optimal antibody concentrations for different experimental applications?

Determining the optimal antibody concentration is critical for experimental success and should be approached methodically:

  • Begin with a pilot titration experiment using a broad concentration range (e.g., 0.1-10 μg/ml)

  • Evaluate both positive and negative controls to establish signal-to-noise ratios

  • Consider tissue/sample type, as different applications may require different concentrations

  • Perform dose-response experiments for quantitative applications

  • Validate specificity using appropriate controls

For clinical applications of ICR62, researchers have used doses ranging from 2.5 mg to 100 mg, with detectable antibody levels in serum at 4h and 24h post-administration at doses of 40 mg or greater . For research applications with similar antibodies like anti-CD62L, concentrations of ≤0.25 μg per 10^6 cells have been effective for flow cytometry .

What techniques are most effective for evaluating antibody-antigen binding kinetics and affinities?

Several complementary techniques can be employed to accurately determine antibody-antigen interactions:

For example, in studies with anti-V monoclonal antibodies, researchers used SPR to determine binding affinities by capturing antibodies (approximately 300 RU/flow cell) on anti-mouse Fc-immobilized CM5 chips, then flowing the target antigen at concentrations ranging from 1 nM to 1.5 μM to determine association constants .

How can researchers validate antibody specificity and cross-reactivity for their target of interest?

Rigorous validation of antibody specificity is essential for reliable research outcomes:

  • Positive and negative control samples: Use samples known to express or lack the target

  • Competitive binding assays: Pre-incubate with unlabeled antibody or purified antigen

  • Western blotting: Confirm binding to proteins of expected molecular weight

  • Immunoprecipitation followed by mass spectrometry: Identify all proteins captured by the antibody

  • Knockout/knockdown validation: Compare staining in cells with and without target expression

For competitive binding assays, researchers can use biotinylated antibody preparations and measure displacement by unlabeled antibodies. This approach has been used effectively to map epitopes and evaluate binding competition between different monoclonal antibodies targeting the same antigen .

What factors influence successful antibody localization to tumor tissues in vivo?

Several critical factors determine the efficiency of antibody localization to tumor tissues:

  • Antibody dose: Higher doses (40 mg or greater) have shown more prominent tumor localization in clinical studies with ICR62

  • Tumor vascularity and permeability: More vascularized tumors typically show better antibody penetration

  • Antigen density: Higher target expression improves localization

  • Antibody affinity: Higher affinity can improve retention but may reduce tissue penetration

  • Antibody size and format: Whole IgG versus fragments (Fab, scFv)

  • Host immune response: Development of human anti-rat antibodies (HARA) can reduce efficacy

In clinical studies with ICR62, researchers observed effective localization to metastatic squamous cell carcinoma lesions when biopsies were obtained 24 hours after antibody administration. Membrane localization was more prominent at higher doses (100 mg compared to 40 mg) .

How should researchers address inconsistent staining patterns or unexpected cross-reactivity?

When troubleshooting staining issues with research antibodies:

  • Optimize fixation protocols: Different fixatives can mask or reveal epitopes

  • Perform antigen retrieval optimization: Test multiple methods (heat-induced, enzymatic)

  • Block non-specific binding: Use appropriate blocking reagents (serum, BSA)

  • Titrate primary and secondary antibodies: Sub-optimal concentrations can cause background or weak signals

  • Consider alternative detection systems: Non-avidin/biotin techniques may reduce background

For example, researchers successfully applied CD62L antibodies to formalin-fixed paraffin-embedded tissues using non-avidin/biotin techniques, which had previously been limited to frozen tissue applications, demonstrating how methodological adaptations can overcome technical limitations .

What approaches are recommended for evaluating therapeutic efficacy of antibodies in preclinical models?

A comprehensive evaluation of therapeutic antibody efficacy should include:

  • Dose-response studies: Test multiple concentrations to establish minimum effective dose

  • Timing studies: Evaluate prophylactic versus therapeutic administration

  • Combination studies: Test with standard treatments or other therapeutic agents

  • Multiple disease models: Evaluate in different preclinical models of the target disease

  • Pharmacokinetic/pharmacodynamic (PK/PD) analysis: Measure antibody clearance and target engagement

In studies with protective monoclonal antibodies against plague, researchers evaluated efficacy through passive protection studies where antibodies were administered intraperitoneally 24 hours before challenge. The challenge dose was delivered either subcutaneously for a bubonic plague model or by whole-body aerosol for a pneumonic plague model, with observations conducted twice daily for the study period .

What immune responses should researchers monitor when using non-human antibodies in clinical studies?

When administering non-human antibodies like ICR62 (rat origin) to patients:

  • Anti-antibody responses: Monitor for human anti-rat antibodies (HARA) or relevant species responses

  • Distinguish between anti-idiotypic (directed against the variable region) and anti-isotypic (directed against the constant region) responses

  • Evaluate timing of response development (typically 1-2 weeks post-administration)

  • Assess impact on antibody clearance and efficacy

  • Consider premedication strategies to reduce hypersensitivity reactions

In clinical trials with ICR62, only 4 out of 20 patients (20%) developed HARA responses across various dose levels (one at 20 mg, one at 40 mg, and two at 100 mg doses). Importantly, only two of these responses were anti-idiotypic, suggesting limited neutralizing potential against the therapeutic effect .

How can researchers quantitatively assess antibody-mediated changes in cell signaling pathways?

For mechanistic studies of antibodies like ICR62 that target signaling receptors:

  • Phospho-specific Western blotting: Measure changes in downstream signaling proteins

  • Phospho-flow cytometry: Quantify signaling changes at single-cell resolution

  • Proximity ligation assays: Detect protein-protein interactions affected by antibody binding

  • RNA-seq or proteomics: Evaluate broader pathway effects

  • Functional assays: Measure biological outcomes (proliferation, migration, apoptosis)

When studying receptor-targeting antibodies like ICR62, researchers should focus on measuring not only direct target engagement (EGFR binding) but also downstream signaling consequences, which provides mechanistic insight into the antibody's therapeutic effects beyond simple receptor blockade .

What considerations are important when comparing the efficacy of different antibody clones targeting the same epitope?

When comparing antibody clones with similar targets:

  • Epitope mapping: Determine precise binding regions through peptide arrays or mutagenesis

  • Avidity and affinity measurements: Quantify binding strength through ELISA (avidity) and SPR (affinity)

  • Functional consequences of binding: Assess whether binding leads to blocking, internalization, or signaling changes

  • Isotype effects: Consider how different isotypes might affect effector functions

  • In vivo potency: Evaluate doses required for equivalent biological effects

In studies comparing anti-V monoclonal antibodies, researchers found that the most protective antibody (mAb 7.3) bound to a unique conformational site on the V-antigen, while less protective antibodies bound to different sites. Interestingly, the protective efficacy did not directly correlate with avidity or affinity measurements, suggesting that epitope specificity was more critical for protection than binding strength alone .

What are the optimal conditions for using antibodies in immunohistochemistry on formalin-fixed, paraffin-embedded tissues?

For successful application of antibodies to FFPE tissues:

  • Deparaffinization and rehydration: Complete removal of paraffin is essential

  • Antigen retrieval: Heat-induced epitope retrieval (HIER) using citrate or EDTA buffers at pH 6.0-9.0

  • Blocking endogenous peroxidase: Use H₂O₂ treatment before antibody application

  • Non-specific binding block: Apply protein block (serum, BSA) for 30-60 minutes

  • Primary antibody incubation: Optimize concentration and incubation time/temperature

  • Detection system: Consider non-avidin/biotin systems to avoid background

Researchers have successfully applied antibodies like anti-CD62L to formalin-fixed tissues using non-avidin/biotin techniques, achieving reliable staining that helped distinguish between reactive and neoplastic T-cell infiltrates. This approach showed CD62L can exhibit enhanced specificity compared to other markers like CD7 in evaluating certain cell populations .

How should researchers quantify diminished antigen expression in comparative studies?

When measuring changes in antigen expression levels:

  • Use consistent acquisition parameters across all samples

  • Include internal positive controls in each experiment

  • Measure percent reduction compared to appropriate reference populations

  • Consider both intensity changes and proportion of positive cells

  • Use digital image analysis for unbiased quantification when possible

In studies of T-cell markers, researchers quantified expression by calculating percentage reduction in staining. For example, in reactive groups, CD62L showed a 15% and 22% reduction in epidermal and dermal staining respectively, while in non-lymphomatous endogenous T-cell dyscrasia and lymphoma categories, an 80% diminution in expression was observed .

What approaches can optimize antibody concentration determination for specialized applications like surface plasmon resonance?

For SPR applications, antibody concentration optimization requires specific considerations:

  • Antibody capture approach: Immobilize anti-species antibodies (e.g., rabbit anti-mouse Fc γ) at high density (~10,000 RU) using amine coupling

  • Capture test antibody: Inject at moderate concentration (e.g., 200 nM) and flow rate (20 μL/min) to achieve consistent capture levels (~300 RU/flow cell)

  • Surface stabilization: Rinse with buffer (10 min at 20 μL/min) to remove poorly captured antibodies

  • Antigen concentration range: Test broad concentration range (e.g., 1 nM to 1.5 μM) to determine accurate binding constants

  • Regeneration conditions: Establish conditions (e.g., 10 mM EDTA, pH 8.0, and 2M NaCl) that remove bound antigen without damaging the captured antibody

This approach, as used in studies of anti-V monoclonal antibodies, allows for accurate determination of association constants (ka) between antibodies and their targets while maintaining a controlled experimental environment .

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.