LCR2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to LCR2 Antibody

Based on available information, "LCR2 Antibody" can refer to different targets depending on the context. It is crucial to specify which molecule LCR2 antibody binds to in order to provide accurate information.

CCR2 Antibody

2.1. General Information
CCR2 Antibody, specifically Anti-CCR2 antibody [EPR20844-15] ab273050, is a rabbit monoclonal antibody. It targets the CC chemokine receptor 2 (CCR2) . CCR2, also known as CD192, Cmkbr2, Ccr2, C-C chemokine receptor type 2, C-C CKR-2, CCR-2, JE/FIC receptor, and MCP-1 receptor, is a key functional receptor for CCL2, but it can also bind CCL7 and CCL12 chemokines .

2.2. Applications
This antibody is used in several techniques, including:

  • Western blotting

  • Immunohistochemistry (IHC)

  • Immunofluorescence

  • Flow cytometry

It is suitable for mouse and rat samples .

2.3. Function
CCR2 plays a role in various biological processes :

  • Chemotaxis and Migration: Mediates chemotaxis and migration induction through the activation of the PI3K cascade, the small G protein Rac, and lamellipodium protrusion in monocytes and macrophages .

  • Inflammation: Regulates the expression of T-cell inflammatory cytokines and T-cell differentiation, promoting the differentiation of T-cells into T-helper 17 cells (Th17) during inflammation .

  • Thymocyte Export: Facilitates the export of mature thymocytes by enhancing directional movement of thymocytes to sphingosine-1-phosphate stimulation and up-regulation of S1P1R expression; signals through the JAK-STAT pathway to regulate FOXO1 activity, leading to increased expression of S1P1R .

  • Neuropathic Pain: Plays an important role in mediating peripheral nerve injury-induced neuropathic pain .

  • Synaptic Transmission: Increases NMDA-mediated synaptic transmission in both dopamine D1 and D2 receptor-containing neurons, potentially through MAPK/ERK-dependent phosphorylation of GRIN2B/NMDAR2B .

  • Macrophage Recruitment: Mediates the recruitment of macrophages and monocytes to the injury site following brain injury .

2.4. Validation

  • Knockout Cell Line Validation: Specificity is confirmed using a CCR2 knockout cell line .

  • Multi-Tissue Microarray (TMA) Validation: Specificity and sensitivity are confirmed in IHC with multi-tissue microarray (TMA) validation .

2.5. Data

TechniqueApplicationSpecies
Western BlottingDetection of CCR2 proteinMouse, Rat
Immunohistochemistry (IHC)Staining of CCR2 in tissues, confirmed with CCR2 knockout miceMouse
ImmunofluorescenceVisualizing CCR2 expression in cellsMouse, Rat
Flow CytometryAnalyzing CCR2-expressing cellsMouse, Rat
ImmunoprecipitationEnrichment of CCR2 proteinUnspecified

2.6. Dual Antagonist
Some CCR2 antagonists also have potent CC chemokine receptor 5 (CCR5) activity . These antagonists have demonstrated in vivo efficacy and oral bioavailability, making them suitable clinical candidates .

LCR1-LCR2 Algorithm in Liver Cancer Risk

3.1. General Information
LCR1-LCR2 is a multianalyte blood test used to assess the risk of hepatocellular carcinoma (HCC) . It combines proteins involved in liver cell repair (apolipoprotein A1, haptoglobin), HCC risk factors (gender, age, gamma glutamyl transpeptidase), a marker of fibrosis (alpha2-macroglobulin), and alpha-fetoprotein (AFP), a specific marker of HCC .

3.2. Validation
A study externally validated the LCR1-LCR2 algorithm in patients with chronic hepatitis B (CHB) . The 5-year negative predictive value (NPV) of LCR1-LCR2 was 99.3% (95% confidence interval = 99.0–99.6) . The algorithm's performance was confirmed in patients with mixed causes of liver disease and in those with hepatitis C virus (HCV) .

3.3. Performance
LCR1-LCR2 outperformed the PAGE-B scoring system for the number of patients needed to screen (NNS) to detect one case of HCC . The sensitivity of the FT-LCR1-LCR2 algorithm was higher compared to standard surveillance (78.4% vs 60.8%, p=0.002) .

3.4. Data

OutcomeResult
5-year Negative Predictive Value (NPV)99.3% (95% CI = 99.0–99.6)
Cox Hazard Ratio (Adjusted)6.4 (3.1–13.0; P < .001) after adjustment for exposure to antivirals, age, gender, geographical origin, HBe-Ag status, alcohol consumption, and type-2 diabetes
Number Needed to Screen (LCR1-LCR2)8.5 (3.2–8.1)
Number Needed to Screen (PAGE-B)26.3 (17.5–38.5; P < .0001)
Sensitivity of FT-LCR1-LCR2 vs. Standard78.4% vs 60.8% (p=0.002)

CCRL2/LCCR Antibody

4.1. General Information
CCRL2/LCCR, also known as LCCR, is a 7TM chemokine receptor-like protein . It is expressed on macrophages, glial cells, and mast cells at inflammatory sites .

LRRK2 Antibody

5.1. General Information
LRRK2 antibodies are used for detecting and purifying LRRK2 (Leucine-rich repeat kinase 2) . LRRK2 antibodies have been developed as renewable (i.e., monoclonal) antibodies with more selectivity and sensitivity for LRRK2 detection and purification .

5.2. Applications
These antibodies are utilized in various techniques :

  • Immunoblotting

  • Immunocytochemistry

  • Immunohistochemistry

  • Immunoprecipitation

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M Phosphate-Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
LCR2 antibody; At4g10595 antibody; T4F9Putative defensin-like protein 145 antibody; Putative low-molecular-weight cysteine-rich protein 2 antibody; Protein LCR2 antibody
Target Names
LCR2
Uniprot No.

Target Background

Database Links
Protein Families
DEFL family
Subcellular Location
Secreted.

Q&A

What is the LCR2 test and how does it function within the liver cancer risk assessment paradigm?

LCR2 is a component of the liver cancer risk test algorithm (LCR1-LCR2), a multi-analyte blood test designed to identify subjects at risk of hepatocellular carcinoma (HCC). This algorithm integrates proteins involved in liver cell repair (apolipoprotein A1, haptoglobin), established HCC risk factors (gender, age, GGT), a fibrosis marker (alpha2-macroglobulin), and AFP, which serves as a specific marker for HCC . The test functions by calculating risk scores based on these parameters to stratify patients according to their likelihood of developing HCC over a defined timeframe, typically five years. Unlike traditional surveillance methods that rely heavily on imaging and single biomarkers, the LCR1-LCR2 algorithm provides a more comprehensive risk assessment by analyzing multiple biomarkers simultaneously.

How does the LCR1-LCR2 sequential algorithm differ from standard AASLD surveillance protocols?

The LCR1-LCR2 sequential algorithm represents a significant methodological departure from standard American Association for the Study of Liver Diseases (AASLD) surveillance, which primarily relies on identifying patients with cirrhosis (F4) and measuring AFP levels. The LCR1-LCR2 algorithm employs a two-tier approach: it first identifies high-risk patients through LCR1 assessment in non-cirrhotic patients (F0-F3), then applies LCR2 to further refine risk stratification in both cirrhotic patients and those non-cirrhotic patients with elevated LCR1 . This approach has demonstrated superior sensitivity compared to standard surveillance (78.4% vs. 60.8%, p=0.002) and improved 5-year HCC-free survival prediction (0.90 [0.81-0.99] vs. 0.77 [0.67-0.86], p=0.003) . The algorithm's ability to identify at-risk patients without cirrhosis represents a particular advantage over traditional surveillance methods that might miss this population segment.

What is the negative predictive value (NPV) of LCR2 in chronic hepatitis C patients, and how is this calculated?

In chronic hepatitis C patients, the LCR1-LCR2 test demonstrates a remarkably high negative predictive value (NPV) of 99.4% (95% CI 99.1–99.6) at 5 years . This means that patients classified as low-risk by the algorithm have a 99.4% probability of not developing HCC within the five-year timeframe. This NPV was calculated in a cohort of 4,903 patients (including 1,026 with baseline cirrhosis) who were followed for a median of 5.7 years. Among 3,755 patients classified as low-risk by LCR1-LCR2, only 24 developed HCC at 5 years, compared to 113 HCC cases among 1,148 patients classified as high-risk . The NPV calculation accounts for various confounding factors, as the findings remained consistent after adjustment for exposure to antivirals, age, gender, geographical origin, HCV genotype-3, previous alcohol consumption, and type 2 diabetes.

What experimental designs have been validated for LCR2 implementation in prospective hepatology studies?

Implementation of LCR2 in prospective hepatology research has been validated through several robust experimental designs. The gold standard appears to be large-scale, multi-center prospective cohort studies with long-term follow-up, exemplified by the Hepather cohort study which enrolled patients from 32 hepatology centers in France . This design allows for comprehensive risk assessment across diverse patient populations. Another validated approach involves matched case-control designs within prospective cohorts, where patients who developed HCC are matched with controls who did not develop HCC during a similar follow-up period. Matching criteria typically include gender, age, and fibrosis stage, with researchers remaining blinded to LCR1-LCR2 results during selection . Such designs enable researchers to evaluate the test's performance while controlling for known confounding factors. Additionally, time-dependent Cox proportional hazards models have been employed to quantify the relationship between LCR1-LCR2 results and HCC development while adjusting for treatment response and other covariables .

How should researchers account for temporal changes in LCR2 markers when designing longitudinal studies?

When designing longitudinal studies involving LCR2, researchers must implement methodologies that account for the temporal dynamics of the component biomarkers. Evidence indicates that LCR2 signals can change over time, with different patterns observed in patients who eventually develop HCC versus those who do not. For instance, research has shown that among patients who developed HCC, only 7.8% experienced disappearance of the algorithm signal when retested approximately 1.5 years later, compared to 13.9% of controls . Additionally, patients who later developed HCC showed a mean increase in LCR2 of +0.014 (SE=0.02) per year, while controls showed no increase (0.00, SE=0.01) . To account for these temporal changes, researchers should:

  • Incorporate scheduled serial measurements at consistent intervals (typically annual or biannual)

  • Use mixed-effects models to analyze rate of change rather than absolute values

  • Establish individual baselines and track delta changes

  • Consider implementing time-dependent Cox proportional hazards models

  • Account for potential confounding events (antiviral therapy, alcohol cessation, etc.) that may alter biomarker trajectories

What are the optimal sample collection and processing protocols to ensure reliability of LCR2 components?

To ensure reliability of LCR2 component measurements, researchers should adhere to standardized sample collection and processing protocols. Blood samples should be collected after overnight fasting to minimize the influence of diet on apolipoprotein A1 and other biomarkers. Immediate processing is recommended, with samples being centrifuged within 2 hours of collection at 3000g for 15 minutes at 4°C. Serum should be aliquoted and stored at -80°C to prevent degradation of protein biomarkers. Multiple freeze-thaw cycles must be avoided as they can significantly alter protein stability and concentration measurements. Batch analysis is preferred to minimize inter-assay variability, and laboratories should participate in quality control programs to ensure standardization. For multi-center studies, centralized testing facilities should be utilized whenever possible. If frozen archived samples are being used, researchers must validate the stability of biomarkers under the specific storage conditions and duration relevant to their study. Importantly, the analytical methods for each component (apolipoprotein A1, haptoglobin, alpha2-macroglobulin, and AFP) should be consistent throughout the study and should match those used in the original validation studies to ensure comparability of results.

How does the integration of LCR2 with genetic biomarkers enhance HCC prediction models in non-cirrhotic patients?

The integration of LCR2 with genetic biomarkers represents a frontier in enhancing HCC prediction models, particularly for non-cirrhotic patients who are often missed by conventional surveillance strategies. Current evidence suggests that combining LCR2 with genetic markers creates a multidimensional risk profile that captures both the functional state of the liver and underlying genetic predispositions. For non-cirrhotic patients (F0-F3 stages), where LCR1 first identifies those at elevated risk, the addition of genetic markers such as PNPLA3 rs738409, TM6SF2 rs58542926, and MBOAT7 rs641738 polymorphisms can further refine risk stratification .

Methodologically, this integration requires sophisticated statistical approaches such as machine learning algorithms that can handle non-linear interactions between biomarkers. Researchers should employ nested model comparison techniques to quantify the incremental predictive value of adding genetic markers to the LCR2 algorithm. A key consideration is the differential weighting of markers based on their relative contribution to HCC risk in non-cirrhotic versus cirrhotic contexts. This approach has demonstrated particular value in patients with metabolic risk factors where genetic predisposition may play a more significant role in carcinogenesis even in the absence of advanced fibrosis.

What methodological approaches can resolve discordance between LCR2 results and traditional imaging surveillance findings?

Resolving discordances between LCR2 results and traditional imaging surveillance findings requires systematic methodological approaches that acknowledge the complementary nature of these surveillance modalities. When discordance occurs (e.g., high-risk LCR2 with negative imaging, or low-risk LCR2 with suspicious lesions), researchers should implement the following resolution framework:

  • Temporal resolution analysis: Track the timing of biomarker elevation relative to imaging detectability to establish the lead-time advantage of molecular versus radiological detection.

  • Risk-stratified surveillance intensification: For patients with high-risk LCR2 but negative imaging, implement shortened imaging intervals (3 months rather than 6 months) and consider alternative imaging modalities (MRI with liver-specific contrast rather than ultrasound).

  • Molecular-radiological correlation studies: Conduct paired analyses of sequential LCR2 measurements with corresponding imaging studies to identify patterns of concordance/discordance and their prognostic implications.

  • Bayesian integration models: Develop statistical models that assign conditional probabilities to true disease state based on the combination of biomarker and imaging results, accounting for the known sensitivity and specificity of each modality.

  • Histopathological validation: When feasible, obtain tissue confirmation of suspicious lesions to establish the ground truth for discordant cases, which can subsequently inform refinement of interpretation algorithms.

This methodological framework allows researchers to systematically characterize the nature of discordances and develop evidence-based protocols for clinical decision-making in such scenarios .

How can LCR2 algorithm performance be optimized for patients with sustained virological response (SVR) after HCV treatment?

  • Recalibration of component weightings: The relative contribution of individual biomarkers to HCC risk likely changes post-SVR, with inflammatory markers potentially carrying less weight while fibrosis and metabolic markers retain significance.

  • Post-SVR specific cut-points: Establish dedicated thresholds for post-SVR patients that account for the expected biomarker changes following viral clearance.

  • Temporal dynamics modeling: Incorporate rate of change in biomarkers post-SVR rather than absolute values, as trajectory of improvement may be more informative than static measurements.

  • Competing risk frameworks: Implement statistical models that account for competing risks (particularly liver-related versus non-liver mortality) which change substantially post-SVR.

  • Integration with regression models: Combine LCR2 with validated post-SVR regression models (such as modified PAGE-B or ANTICIPATE) to create hybrid algorithms specifically calibrated for the post-SVR setting.

These methodological refinements should be validated in large, diverse cohorts of post-SVR patients with adequate follow-up to capture late HCC events, which can occur even years after successful viral eradication .

What statistical methods are most appropriate for evaluating the incremental value of LCR2 over existing predictive models?

The evaluation of LCR2's incremental value over existing predictive models requires sophisticated statistical approaches that go beyond traditional measures of discrimination. Researchers should implement a comprehensive analytical framework that includes:

  • Nested model comparison: Likelihood ratio tests comparing models with and without LCR2 components to quantify statistical significance of information gain.

  • Discrimination improvement metrics: Calculate and report both area under the receiver operating characteristic curve (AUROC) improvements and more sensitive measures such as the Integrated Discrimination Improvement (IDI) and Net Reclassification Improvement (NRI), which better capture the ability to correctly reclassify patients.

  • Calibration assessment: Employ Hosmer-Lemeshow tests and calibration plots stratified by risk groups to ensure that predicted probabilities match observed outcomes across the risk spectrum.

  • Decision curve analysis: Quantify the net benefit of clinical decisions based on models including LCR2 versus existing models across a range of decision thresholds.

  • Time-dependent ROC analysis: Implement time-dependent ROC analysis to assess how the discriminative ability of models changes over various prediction horizons (1-year, 3-year, 5-year).

This comprehensive statistical approach has revealed that the LCR1-LCR2 algorithm provides substantial incremental value over standard surveillance methods, with significantly improved sensitivity (78.4% vs. 60.8%) and superior 5-year survival discrimination .

How should researchers address potential biological confounders in LCR2 interpretation, such as comorbid autoimmune conditions?

Addressing biological confounders in LCR2 interpretation is essential for accurate risk assessment, particularly when comorbid conditions might affect component biomarkers. Researchers should implement the following methodological approaches:

  • Stratified analysis: Conduct subgroup analyses within specific comorbidity cohorts to establish whether algorithm performance remains consistent. For instance, autoimmune conditions may influence haptoglobin and alpha2-macroglobulin levels independently of liver pathology.

  • Confounder adjustment models: Employ multivariable models that explicitly adjust for defined comorbidities as covariates when assessing LCR2 performance. The Hepather cohort study demonstrated robustness of results after adjustment for various confounders including age, gender, geographical origin, HCV genotype-3, previous alcohol consumption, and type 2 diabetes .

  • Biomarker interaction studies: Investigate specific interactions between comorbidity-related biomarkers and LCR2 components to identify potential mechanistic overlaps.

  • Sensitivity analyses: Perform analyses excluding patients with specific comorbidities to determine whether results remain consistent in the unaffected population.

  • Comorbidity-specific recalibration: For patient populations with high prevalence of specific comorbidities, consider developing recalibrated algorithms with adjusted weightings for affected biomarkers.

This systematic approach to addressing biological confounders ensures that LCR2 interpretation remains valid across diverse patient populations with varying comorbidity profiles, enhancing the generalizability of research findings.

What are the methodological considerations for normalizing LCR2 results across different laboratory platforms and reagent lots?

Normalizing LCR2 results across different laboratory platforms and reagent lots is essential for multi-center research studies and longitudinal data collection. Researchers should implement a comprehensive normalization framework that includes:

  • Reference standardization: Establish common calibrators and reference materials that can be shared across participating laboratories to anchor measurements to a consistent standard.

  • Bridging studies: Conduct formal method comparison studies when transitioning between platforms or reagent lots, with at least 100 samples spanning the analytical range measured on both systems to derive conversion factors.

  • Commutability assessment: Verify that reference materials behave similarly to patient samples across different measurement systems to ensure applicability of standardization.

  • External quality assessment (EQA): Participate in regular EQA programs where identical samples are analyzed by multiple laboratories to identify and quantify inter-laboratory variability.

  • Statistical harmonization: Apply harmonization algorithms such as quantile normalization or z-score standardization when direct standardization is not feasible.

  • Lot-to-lot verification: For longitudinal studies, implement verification protocols when reagent lots change, including analysis of control samples spanning the measurement range on both the current and new lots.

  • Machine learning normalization: Consider advanced computational approaches that can learn and correct for systematic biases between measurement systems based on paired measurements.

By implementing these methodological considerations, researchers can ensure that observed differences in LCR2 results reflect true biological variation rather than analytical artifacts, thereby enhancing the validity and reproducibility of research findings across different settings.

How might integration of artificial intelligence with LCR2 enhance personalized HCC surveillance protocols?

The integration of artificial intelligence (AI) with LCR2 presents transformative opportunities for personalized HCC surveillance. AI approaches can extend beyond current static risk algorithms to create dynamic, adaptive surveillance protocols tailored to individual patient trajectories. Methodologically, researchers should explore:

  • Deep learning temporal models: Implement recurrent neural networks or transformer-based architectures that can process sequential LCR2 measurements along with other clinical data to identify subtle patterns preceding HCC development.

  • Reinforcement learning frameworks: Develop algorithms that optimize surveillance intervals based on continuously updated risk assessments, potentially reducing unnecessary imaging for consistently low-risk patients while intensifying surveillance for those with concerning biomarker trajectories.

  • Multi-modal integration: Create AI systems that simultaneously process LCR2 biomarker data, imaging findings, and clinical variables to generate integrated risk assessments with higher sensitivity and specificity than any single modality.

  • Explainable AI approaches: Implement attention mechanisms and feature importance analyses to identify which components of the LCR2 algorithm are most influential for individual patients at specific time points, providing clinically actionable insights.

  • Federated learning systems: Develop privacy-preserving AI frameworks that can learn from decentralized data across multiple institutions without requiring direct data sharing, thereby accelerating algorithm refinement through access to larger and more diverse patient populations.

Research in this direction should include comprehensive validation studies comparing AI-enhanced LCR2 surveillance protocols against standard approaches, with particular attention to cost-effectiveness, detection of early-stage HCC amenable to curative treatment, and potential reduction in surveillance-related harms .

What methodological approaches can best evaluate LCR2 performance in non-viral etiologies of chronic liver disease?

Evaluating LCR2 performance in non-viral etiologies of chronic liver disease requires methodological approaches that account for the distinct pathophysiological mechanisms and natural history of these conditions. Researchers should implement:

  • Etiology-specific validation cohorts: Establish dedicated cohorts for specific etiologies (NASH, alcohol-related liver disease, autoimmune hepatitis, etc.) with sufficient sample size to achieve adequate statistical power for rare events like HCC.

  • Comparative effectiveness studies: Directly compare LCR2 performance against etiology-specific risk scores (e.g., GALAD for NAFLD-HCC) using paired analyses within the same patient population.

  • Biomarker interaction analyses: Conduct interaction studies between LCR2 components and etiology-specific biomarkers to identify potential synergistic or antagonistic relationships that might affect algorithm performance.

  • Etiology-specific recalibration: Consider recalibrating component weightings for specific etiologies based on the relative contribution of inflammation, fibrosis, and metabolic factors to carcinogenesis in each condition.

  • Competing risk frameworks: Implement statistical models that account for the distinct competing risks in different etiologies (e.g., higher non-HCC mortality in alcohol-related disease versus NAFLD).

  • Integration with genetic risk factors: Combine LCR2 with known genetic risk variants specific to different etiologies (e.g., PNPLA3 for NAFLD, HSD17B13 for ALD) to enhance predictive accuracy.

These approaches can help establish whether the promising performance of LCR2 observed in viral hepatitis extends to other chronic liver diseases, potentially expanding its clinical utility across the spectrum of patients at risk for HCC .

What are the methodological challenges in applying LCR2 to populations underrepresented in validation studies?

Applying LCR2 to populations underrepresented in validation studies presents several methodological challenges that researchers must systematically address:

  • Baseline distribution calibration: Establish reference distributions of LCR2 components in underrepresented populations, as these may differ from those in validation cohorts due to genetic factors, environmental exposures, and comorbidity patterns.

  • Threshold recalibration: Evaluate whether the established cutoff values for LCR1 and LCR2 maintain optimal sensitivity and specificity in diverse populations; consider population-specific thresholds if necessary.

  • External validation studies: Conduct dedicated external validation studies in underrepresented populations with sufficient sample size and follow-up duration to capture enough HCC events for robust statistical analysis.

  • Interaction with population-specific risk factors: Investigate potential interactions between LCR2 components and population-specific risk factors (e.g., aflatoxin exposure in parts of Africa and Asia).

  • Transfer learning approaches: Employ statistical methods like Bayesian updating or transfer learning to adapt existing models to new populations while preserving information from original validation cohorts.

  • Equity impact analysis: Explicitly evaluate whether LCR2 implementation maintains consistent performance across demographic subgroups, and quantify any potential disparities in algorithm performance.

  • Cultural and access considerations: Develop implementation frameworks that address cultural factors and healthcare access barriers that might affect feasibility of LCR2-based surveillance in diverse populations.

These methodological approaches can help ensure that the benefits of LCR2-based risk stratification extend equitably across diverse populations, rather than exacerbating existing healthcare disparities in HCC outcomes .

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 2025 TheBiotek. All Rights Reserved.