PRAMEF17 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Structure and Conjugation

The PRAMEF17 antibody is a polyclonal immunoglobulin raised in rabbits, recognizing epitopes in the human PRAMEF17 protein. Biotin conjugation involves covalently attaching biotin molecules to the antibody’s lysine residues, enabling high-affinity binding to streptavidin or avidin in detection systems.

Key Features:

  • Conjugate: Biotin with a 6-atom spacer (Biotin-SP) enhances accessibility to streptavidin, improving assay sensitivity .

  • Host Species: Rabbit (polyclonal), ensuring broad epitope recognition .

  • Purification: Affinity chromatography removes cross-reactivities, minimizing non-specific binding .

Product CodeSupplierReactivityApplications
CSB-PA018612LD01HUCusabioHumanELISA, WB
ABIN7164117Antibodies-onlineHumanELISA, WB

Applications in Research

The biotin-conjugated PRAMEF17 antibody is optimized for:

ELISA

  • Used in sandwich ELISA to quantify PRAMEF17 in lysates or serum .

  • Requires streptavidin-HRP for signal amplification, achieving detection limits as low as 1 ng/mL .

Western Blotting (WB)

  • Recommended dilution: 1:1000–1:5000 .

  • Compatible with PVDF membranes, with enhanced signal using streptavidin-alkaline phosphatase .

Immunohistochemistry (IHC)

  • Detects PRAMEF17 in tumor tissues, aiding cancer biomarker studies .

  • Requires antigen retrieval and blocking of endogenous biotin .

Validation and Specificity

  • Antibodypedia: Validated for WB and ELISA via independent user reviews .

  • Cusabio: Tested against recombinant PRAMEF17 protein, ensuring no cross-reactivity with human, mouse, or rat serum proteins .

  • Antibodies-online: Confirmed specificity via immunoprecipitation and mass spectrometry .

Challenges and Considerations

  • Endogenous Biotin Interference: High biotin levels in samples (e.g., egg yolk) can cause false positives. Blocking solutions or alternative detection systems (e.g., fluorophores) are recommended .

  • Optimal Dilution: Variability in antigen density necessitates empirical titration .

Research Implications

PRAMEF17 is a tumor-associated antigen overexpressed in cancers like melanoma and ovarian carcinoma . The biotin-conjugated antibody enables:

  • Biomarker Discovery: Identifying PRAMEF17 as a therapeutic target for antibody-drug conjugates (ADCs) .

  • Immunotherapy Monitoring: Tracking immune responses to PRAMEF17 in clinical trials .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. For specific delivery times, please contact your local distributor.
Synonyms
PRAMEF17 antibody; PRAME family member 17 antibody
Target Names
PRAMEF17
Uniprot No.

Q&A

What is PRAMEF17 and why is it studied?

PRAMEF17 (PRAME family member 17) is part of the PRAME family of proteins, which have gained significant research interest due to their association with various malignancies. The PRAME gene family includes tumor-associated antigens that were first identified in melanoma patients and have since been found to be expressed in multiple cancer types . PRAMEF17 specifically is a member of this family, with the UniProt ID Q5VTA0 .

Researchers study PRAMEF17 primarily to understand its potential role in cancer biology, particularly its expression patterns in normal versus neoplastic tissues. Though less extensively studied than some other PRAME family members, PRAMEF17 may share functional characteristics with PRAME, which has been demonstrated to have value as a biomarker in melanoma and other malignancies. Investigation of PRAMEF17 could provide insights into cancer pathogenesis and potentially identify new biomarkers or therapeutic targets.

What are the technical specifications of PRAMEF17 Antibody, Biotin conjugated?

PRAMEF17 Antibody (Biotin) is a rabbit polyclonal antibody specifically designed for the detection of human PRAMEF17. The antibody has been conjugated to biotin, which facilitates its use in various detection systems. Key specifications include:

ParameterSpecification
Host SpeciesRabbit
ReactivityHuman
ClonalityPolyclonal
ConjugationBiotin
IsotypeIgG
Purity> 95%
Purification MethodProtein G chromatography
Physical FormLiquid
Buffer Composition0.01 M PBS, pH 7.4, 0.03% Proclin-300 and 50% glycerol
Validated ApplicationsELISA
UniProt IDQ5VTA0

The antibody has been validated for ELISA applications, though optimal dilutions should be determined by the researcher for their specific experimental conditions .

How should PRAMEF17 Antibody, Biotin conjugated be stored and handled?

Proper storage and handling of PRAMEF17 Antibody (Biotin) is crucial for maintaining its integrity and performance in experimental applications. The manufacturer recommends aliquoting the antibody and storing it at -20°C . This approach minimizes the number of freeze-thaw cycles the antibody experiences, which can degrade protein structure and reduce antibody performance.

When handling the antibody, researchers should:

  • Thaw aliquots completely before use, but keep them cold (on ice) during experimental setup

  • Avoid repeated freeze-thaw cycles as this can lead to protein denaturation and loss of activity

  • Work in clean conditions to prevent contamination

  • Follow good laboratory practices such as using calibrated pipettes and clean tubes

  • Return unused portions to -20°C promptly after use

The antibody is provided in a buffer containing 50% glycerol, which helps stabilize the protein during freeze-thaw cycles. The inclusion of 0.03% Proclin-300 acts as a preservative to prevent microbial growth .

What are the common applications for PRAMEF17 Antibody, Biotin conjugated?

  • ELISA: For quantitative detection of PRAMEF17 in various sample types, including cell lysates, tissue homogenates, and potentially serum samples.

  • Immunohistochemistry (IHC): Although not explicitly validated, biotin-conjugated antibodies are often suitable for IHC applications when used with appropriate streptavidin-based detection systems.

  • Western Blotting: Some researchers report using PRAMEF17 antibodies for Western Blot applications , though specific validation for the biotin-conjugated version should be performed.

  • Flow Cytometry: Potentially useful for detecting PRAMEF17 in cell populations when paired with streptavidin-fluorophore conjugates.

  • Immunoprecipitation: The biotin tag can facilitate pull-down assays when combined with streptavidin beads.

Each application requires optimization and validation in the researcher's specific experimental system. Preliminary experiments to determine optimal dilutions and conditions are strongly recommended .

How should I validate PRAMEF17 Antibody, Biotin conjugated for my specific application?

Thorough validation of PRAMEF17 Antibody (Biotin) for your specific application is essential to ensure reliable and reproducible results. A comprehensive validation process should include:

  • Positive and Negative Controls: Include samples known to express (positive control) and not express (negative control) PRAMEF17. Positive controls might include cell lines reported to express PRAMEF17, while negative controls could be tissues or cells where expression is absent.

  • Antibody Dilution Optimization: Perform a titration series to determine the optimal antibody concentration that provides maximum specific signal with minimal background. Start with the manufacturer's recommended dilution range and adjust as needed .

  • Specificity Tests:

    • Peptide Competition Assay: Pre-incubate the antibody with purified PRAMEF17 protein or immunizing peptide prior to application. A specific antibody will show reduced or eliminated signal.

    • Knockdown/Knockout Validation: Compare staining between wild-type samples and those where PRAMEF17 has been knocked down (siRNA) or knocked out (CRISPR-Cas9).

  • Cross-Reactivity Assessment: Test the antibody on samples from other species or on related PRAME family proteins to assess potential cross-reactivity.

  • Protocol Optimization: Systematically vary experimental conditions (incubation times, temperatures, blocking reagents, washing steps) to identify optimal parameters.

  • Comparison with Alternative Antibodies: When possible, compare results with other PRAMEF17 antibodies, particularly those using different epitopes or conjugation methods.

  • Reproducibility Testing: Repeat experiments multiple times to ensure consistent results across experimental replicates.

Documentation of these validation steps is crucial for publication and ensures confidence in experimental findings.

What are the best protocols for using PRAMEF17 Antibody, Biotin conjugated in ELISA assays?

For optimal ELISA results using PRAMEF17 Antibody (Biotin conjugated), consider the following protocol guidelines:

Direct ELISA Protocol:

  • Coating:

    • Coat ELISA plate wells with target protein or sample at 1-10 μg/ml in carbonate-bicarbonate buffer (pH 9.6)

    • Incubate overnight at 4°C

  • Blocking:

    • Wash wells 3 times with PBST (PBS + 0.05% Tween-20)

    • Block with 2-5% BSA or milk in PBST for 1-2 hours at room temperature

  • Primary Antibody:

    • Dilute PRAMEF17 Antibody (Biotin) in blocking buffer (initial test range: 1:500 - 1:5000)

    • Add to wells and incubate for 1-2 hours at room temperature

    • Wash 4-5 times with PBST

  • Detection:

    • Add streptavidin-HRP (typically 1:1000 - 1:5000) in blocking buffer

    • Incubate for 30-60 minutes at room temperature

    • Wash 4-5 times with PBST

  • Development:

    • Add appropriate substrate (TMB, ABTS, etc.)

    • Monitor color development

    • Stop reaction (if using TMB, add H₂SO₄)

    • Read absorbance at appropriate wavelength

Optimization Considerations:

  • Determine optimal antibody dilution specifically for your sample type

  • Include a standard curve using recombinant PRAMEF17 protein if quantitation is needed

  • Run positive and negative controls in each assay

  • If high background is observed, consider:

    • More stringent washing

    • Higher dilution of antibody

    • Different blocking reagents

    • Pre-adsorption of antibody

The biotin conjugation offers enhanced sensitivity through signal amplification with streptavidin systems, but requires careful optimization to minimize background and maximize specific signal detection .

Can PRAMEF17 Antibody, Biotin conjugated be used for immunohistochemistry (IHC)?

While the product specification for PRAMEF17 Antibody (Biotin) primarily lists ELISA as its validated application , biotin-conjugated antibodies can often be adapted for immunohistochemistry with appropriate optimization. Drawing from approaches used with other PRAME family antibodies in IHC applications , researchers might consider the following protocol framework:

IHC Protocol Adaptation:

  • Tissue Preparation:

    • Use formalin-fixed paraffin-embedded (FFPE) tissue sections (4-6 μm thickness)

    • Deparaffinize and rehydrate through graded alcohols to water

  • Antigen Retrieval:

    • Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)

    • Boil for 15-20 minutes in pressure cooker or similar device

    • Cool to room temperature

  • Endogenous Biotin Blocking (critical step):

    • Block endogenous biotin using a commercial avidin/biotin blocking kit

    • This step is essential for biotin-conjugated antibodies to reduce background

  • Peroxidase Blocking:

    • Treat with 3% hydrogen peroxide for 10 minutes

    • Wash in PBS

  • Protein Blocking:

    • Block with 5-10% normal serum (from species used for secondary detection) for 30-60 minutes

  • Primary Antibody:

    • Apply diluted PRAMEF17 Antibody (Biotin) (starting dilution 1:100-1:500)

    • Incubate overnight at 4°C or 1-2 hours at room temperature

    • Wash thoroughly with PBS

  • Detection:

    • Apply streptavidin-HRP conjugate (1:200-1:1000)

    • Incubate for 30-60 minutes at room temperature

    • Wash thoroughly with PBS

  • Visualization:

    • Develop with DAB substrate

    • Monitor under microscope for optimal development time

    • Counterstain with hematoxylin

    • Dehydrate, clear, and mount

Validation and Controls:

  • Include positive control tissues (potentially melanoma samples, based on PRAME expression patterns)

  • Include negative controls (omitting primary antibody)

  • Consider peptide competition controls to verify specificity

  • Optimization of antibody dilution and incubation conditions is essential

Researchers should note that staining patterns for PRAME family members in IHC can show nuclear localization in melanoma cells, with different patterns in non-neoplastic tissues , which may inform interpretation of PRAMEF17 staining.

How can I troubleshoot common issues when using PRAMEF17 Antibody, Biotin conjugated?

When working with PRAMEF17 Antibody (Biotin conjugated), researchers may encounter several common issues. Here are systematic troubleshooting approaches for each:

1. High Background Signal:

  • Potential Causes and Solutions:

    • Insufficient blocking: Increase blocking time/concentration or try alternative blocking reagents

    • Antibody concentration too high: Perform titration to determine optimal dilution

    • Insufficient washing: Increase number/duration of washes

    • Endogenous biotin interference: Implement avidin/biotin blocking step before antibody application

    • Sample autofluorescence: Include autofluorescence controls and consider quenching protocols

    • Cross-reactivity: Pre-absorb antibody with related proteins

2. Weak or No Signal:

  • Potential Causes and Solutions:

    • Insufficient antigen: Increase sample concentration

    • Antibody concentration too low: Decrease antibody dilution

    • Inadequate antigen retrieval: Optimize antigen retrieval method (pH, time, temperature)

    • Antibody degradation: Use fresh aliquot, avoid freeze-thaw cycles

    • Inactive detection system: Test streptavidin-conjugate with a different biotinylated antibody

    • Target protein denaturation: Modify fixation or sample preparation methods

    • Low target expression: Confirm PRAMEF17 expression in your sample type

3. Non-specific Binding:

  • Potential Causes and Solutions:

    • Cross-reactivity with related proteins: Validate antibody specificity

    • Fc receptor binding: Add Fc receptor blockers to protocol

    • Hydrophobic interactions: Increase detergent (Tween-20) concentration in wash buffer

    • Inappropriate blocking agent: Test different blocking proteins (BSA, milk, normal serum)

4. Inconsistent Results:

  • Potential Causes and Solutions:

    • Variable sample preparation: Standardize sample collection and processing

    • Inconsistent antibody aliquots: Prepare multiple small aliquots from original stock

    • Protocol timing variations: Strictly control incubation times and temperatures

    • Reagent quality changes: Use consistent lots of detection reagents

    • Equipment variations: Calibrate equipment regularly

5. Biotinylation-Specific Issues:

  • Potential Causes and Solutions:

    • Over-biotinylation affecting epitope recognition: Use different clone or unbiotinylated version

    • Steric hindrance: Try different streptavidin conjugates of varying sizes

    • Competition with endogenous biotin: Implement thorough biotin blocking steps

When troubleshooting, change only one variable at a time and include appropriate controls to systematically identify the source of the problem .

How does PRAMEF17 Antibody, Biotin conjugated compare with other detection methods for PRAME family proteins?

The detection of PRAME family proteins, including PRAMEF17, can be accomplished through various methodologies, each with distinct advantages and limitations compared to using PRAMEF17 Antibody (Biotin conjugated):

Antibody-Based Methods Comparison:

Detection MethodAdvantagesLimitationsComparison to PRAMEF17 Antibody (Biotin)
Unconjugated Primary AntibodiesGreater flexibility in detection systems; No biotin interferenceRequires secondary antibody; Potentially lower sensitivityPRAMEF17 Antibody (Biotin) offers direct detection with streptavidin systems without secondary antibody
HRP-Conjugated AntibodiesDirect detection without additional reagentsLess signal amplification potential; Fixed detection systemPRAMEF17 Antibody (Biotin) provides greater amplification potential through streptavidin systems
Fluorophore-Conjugated AntibodiesDirect visualization; Multiplexing capabilityPhotobleaching; Autofluorescence interferencePRAMEF17 Antibody (Biotin) can be paired with various streptavidin-fluorophores for flexible detection

Non-Antibody Detection Methods:

  • mRNA Detection (RT-PCR, RNA-Seq):

    • Provides quantitative expression data at the transcript level

    • Often used for PRAME expression analysis in melanoma research

    • Unlike PRAMEF17 Antibody detection, cannot confirm protein localization or post-translational modifications

    • Higher sensitivity but may not correlate with protein levels

  • Mass Spectrometry:

    • Allows unbiased protein identification and quantification

    • Can detect post-translational modifications

    • Requires specialized equipment and expertise

    • Less sensitive than antibody-based methods for low-abundance proteins

  • CRISPR Knock-in Tags:

    • Endogenous tagging for detection without antibodies

    • Eliminates antibody specificity concerns

    • Requires genetic modification of cells/organisms

    • Not feasible for clinical samples unlike PRAMEF17 Antibody methods

The choice between these methods depends on research objectives. For investigating protein localization in fixed tissues or detecting native PRAMEF17 in various sample types, PRAMEF17 Antibody (Biotin) offers advantages in sensitivity and versatility compared to non-antibody methods. For absolute quantification or transcript-level analysis, mRNA-based methods might be preferable. Recent antibody design advancements using computational approaches like RFdiffusion may eventually improve antibody specificity and performance .

What are the considerations for multiplex immunoassays including PRAMEF17 Antibody, Biotin conjugated?

Incorporating PRAMEF17 Antibody (Biotin conjugated) into multiplex immunoassays requires careful planning to ensure compatibility with other detection systems while maintaining specificity and sensitivity. Here are key considerations for researchers:

Technical Considerations for Multiplexing:

  • Detection System Compatibility:

    • Streptavidin Conjugate Selection: Choose streptavidin conjugates (fluorophores, enzymes) that are spectrally distinct from other detection systems in your multiplex assay

    • Signal Separation: Ensure adequate separation between detection channels to prevent bleed-through (particularly important in fluorescence-based systems)

    • Sequential Detection: Consider sequential rather than simultaneous detection if cross-reactivity is observed

  • Antibody Compatibility:

    • Species Cross-Reactivity: Avoid secondary antibodies that might recognize the rabbit IgG of the PRAMEF17 Antibody

    • Blocking Optimization: Develop blocking strategy that works for all antibodies in the panel

    • Dilution Balancing: Optimize individual antibody dilutions to achieve comparable signal intensities

  • Sample Preparation Harmonization:

    • Antigen Retrieval: Select a retrieval method compatible with all target epitopes

    • Fixation Protocol: Use fixation that preserves all antigens of interest

    • Endogenous Biotin Blocking: Implement thorough biotin blocking to prevent interference with the biotin-conjugated PRAMEF17 Antibody

Experimental Design Strategies:

  • Panel Design Approach:

    • Start with validated single-marker assays

    • Add antibodies one at a time to identify interference

    • Include appropriate single-stain controls

    • Consider antibody order of application (typically from weakest to strongest signal)

  • Validation Requirements:

    • Verify each marker individually before multiplexing

    • Compare multiplex results with single-plex for each marker

    • Include appropriate compensation controls for fluorescence-based detection

  • Potential Applications with PRAMEF17:

    • Tumor Microenvironment Analysis: Combine PRAMEF17 with immune cell markers

    • Cancer Phenotyping: Multiplex with other diagnostic markers

    • Signaling Pathway Investigation: Combine with phosphorylation-specific antibodies

  • Special Considerations for Biotin-Conjugated Antibodies:

    • Apply the PRAMEF17 Antibody (Biotin) early in sequential protocols to minimize cross-reactivity

    • Consider tyramide signal amplification (TSA) for enhanced sensitivity in multiplex IHC

    • Implement stringent washing between detection steps to remove unbound reagents

By addressing these considerations systematically, researchers can successfully incorporate PRAMEF17 Antibody (Biotin) into multiplex assays while maintaining specificity and generating meaningful co-localization data .

How can PRAMEF17 Antibody, Biotin conjugated be used in cancer research?

PRAMEF17 Antibody (Biotin conjugated) offers valuable applications in cancer research, particularly given the established role of PRAME family proteins as cancer/testis antigens expressed in various malignancies . Here are advanced research applications in oncology:

Tumor Expression Profiling:

PRAMEF17 Antibody can be utilized to characterize expression patterns across different tumor types, stages, and grades. Based on PRAME studies, researchers should focus on:

  • Comparative Tissue Analysis:

    • Examine PRAMEF17 expression in tumor versus matched normal tissues

    • Analyze expression across tumor progression stages

    • Compare expression in primary tumors versus metastatic lesions

  • Correlation with Clinical Outcomes:

    • Assess relationship between PRAMEF17 expression and patient survival

    • Evaluate association with treatment response

    • Investigate correlation with recurrence or metastasis

  • Co-expression Analysis:

    • Examine relationship with other PRAME family members

    • Investigate association with established cancer biomarkers

Mechanistic Studies:

  • Subcellular Localization:

    • Determine whether PRAMEF17 exhibits nuclear, cytoplasmic, or other localization patterns

    • Compare with known PRAME family localization (nuclear in melanoma cells)

    • Investigate potential translocation under different cellular conditions

  • Functional Investigations:

    • Combine PRAMEF17 detection with markers of cell proliferation, apoptosis, or differentiation

    • Correlate expression with activation of specific signaling pathways

    • Examine changes in expression following treatment with targeted therapies

  • Tumor Microenvironment Interactions:

    • Analyze PRAMEF17 expression in relation to immune infiltrates

    • Investigate potential role in immune evasion mechanisms

    • Explore relationship with tumor microenvironment markers

Translational Applications:

  • Biomarker Development:

    • Evaluate PRAMEF17 as a diagnostic or prognostic biomarker

    • Assess utility for monitoring treatment response

    • Investigate potential for early detection applications

  • Therapeutic Target Assessment:

    • Determine if PRAMEF17 expression correlates with response to immunotherapy

    • Evaluate potential as a target for antibody-drug conjugates

    • Investigate as a cancer vaccine candidate

  • Liquid Biopsy Development:

    • Explore detection of PRAMEF17 in circulating tumor cells

    • Assess potential as a serum biomarker

    • Investigate utility in minimal residual disease monitoring

Given the emerging understanding of PRAME family proteins in cancer, researchers should design careful validation studies when extending findings from other family members to PRAMEF17. The biotin conjugation facilitates sensitive detection across multiple platforms, enabling comprehensive characterization of this potential biomarker in cancer research applications .

What approaches can be used to compare PRAMEF17 expression between different tissue samples?

Comprehensive comparison of PRAMEF17 expression across different tissue samples requires methodological rigor to ensure reliable and reproducible results. Here are advanced approaches using PRAMEF17 Antibody (Biotin conjugated) alongside complementary techniques:

Quantitative Analysis Approaches:

  • Tissue Microarray (TMA) Analysis:

    • Create TMAs containing multiple tissue cores from different samples

    • Apply standardized IHC protocols using PRAMEF17 Antibody (Biotin)

    • Implement digital image analysis for objective quantification

    • Scoring parameters:

      • Percentage of positive cells (0-100%)

      • Staining intensity (0: negative, 1+: weak, 2+: moderate, 3+: strong)

      • H-score calculation: Σ(intensity × percentage), range 0-300

  • Multiplexed IHC/Immunofluorescence:

    • Combine PRAMEF17 Antibody with other markers of interest

    • Use spectral unmixing to separate overlapping signals

    • Perform cell-by-cell quantification of expression levels

    • Apply spatial analysis to evaluate distribution patterns

  • Semi-Quantitative ELISA:

    • Prepare standardized tissue lysates

    • Develop quantitative ELISA using PRAMEF17 Antibody (Biotin)

    • Generate standard curves with recombinant PRAMEF17

    • Express results as ng/mg total protein

Complementary Analytical Methods:

  • Multi-omics Integration:

    • Correlate protein expression (PRAMEF17 Antibody detection) with:

      • mRNA expression (qRT-PCR, RNA-seq)

      • Methylation status of the PRAMEF17 promoter

      • Proteomic profiles via mass spectrometry

    • Integrate data using computational approaches for comprehensive expression analysis

  • Single-Cell Analysis:

    • Perform single-cell immunofluorescence using PRAMEF17 Antibody

    • Combine with single-cell RNA-seq for correlation analysis

    • Evaluate heterogeneity of expression within tissues

  • 3D Tissue Analysis:

    • Apply PRAMEF17 Antibody in cleared tissue samples

    • Perform confocal or light-sheet microscopy

    • Reconstruct 3D expression patterns for spatial comparison

Statistical Analysis Framework:

Analysis TypeApplicationStatistical Approach
Between-Group ComparisonTumor vs. NormalMann-Whitney U test or t-test
Multi-Group ComparisonDifferent tumor types/gradesANOVA with post-hoc tests
Correlation AnalysisRelation to clinical parametersSpearman's or Pearson's correlation
Survival AnalysisAssociation with outcomesKaplan-Meier with log-rank test
Multivariate AnalysisIndependent prognostic valueCox proportional hazards model

Normalization and Quality Control:

  • Technical Standardization:

    • Include identical positive and negative controls across batches

    • Utilize automated staining platforms when possible

    • Implement tissue-specific normalization strategies

  • Biological Normalization:

    • Account for cellular composition differences between samples

    • Consider normalization to housekeeping proteins

    • Adjust for tissue-specific baseline expression

  • Batch Effect Correction:

    • Implement statistical methods to correct for batch effects

    • Consider reference sample inclusion across batches

    • Apply computational correction algorithms when combining datasets

When publishing comparative analyses, researchers should report detailed methodological parameters, quantification approaches, and statistical methods to ensure reproducibility .

How might advanced antibody design methods impact future PRAMEF17 detection tools?

Recent advances in computational antibody design, particularly approaches like RFdiffusion highlighted in the literature, are poised to significantly transform PRAMEF17 detection tools in the near future . These emerging technologies offer potential improvements across multiple dimensions:

Enhanced Specificity and Reduced Cross-Reactivity:

The atomically accurate de novo design of antibodies using computational methods like RFdiffusion enables precise engineering of binding interfaces . For PRAMEF17 detection, this could lead to:

  • Epitope-Specific Recognition: Design of antibodies that precisely target unique epitopes on PRAMEF17, distinguishing it from other closely related PRAME family members

  • Reduced Off-Target Binding: Minimization of cross-reactivity with other proteins, addressing a common limitation of current polyclonal antibodies

  • Custom Binding Properties: Optimization of binding affinity and kinetics for specific research applications

Improved Structural and Functional Characteristics:

Advanced computational design methods demonstrated in recent research could yield PRAMEF17 antibodies with:

  • Optimized CDR Loops: Specifically designed complementarity-determining regions (CDRs) that maximize target interaction while minimizing non-specific binding

  • Enhanced Stability: Improved thermal and chemical stability to withstand harsh experimental conditions

  • Controlled Binding Orientation: Precise control over antibody-antigen binding geometry to improve detection sensitivity and consistency

Novel Formats and Applications:

Computational design approaches open possibilities for innovative PRAMEF17 detection tools:

  • Multispecific Antibodies: Development of antibodies that simultaneously bind PRAMEF17 and other biomarkers of interest for multiplexed detection

  • Intracellular Antibodies (intrabodies): Generation of antibodies optimized for intracellular expression and detection of PRAMEF17 in living cells

  • Antibody Fragments: Creation of smaller binding domains (scFvs, nanobodies) with enhanced tissue penetration for improved histological detection

Integration with Emerging Technologies:

The convergence of computational antibody design with other technological advances could lead to:

  • AI-Enhanced Imaging Analysis: Pairing of highly specific PRAMEF17 antibodies with machine learning algorithms for automated quantification and pattern recognition

  • Proximity-Based Detection Systems: Integration with proximity ligation assays or FRET-based systems to study PRAMEF17 protein interactions

  • Nanotechnology Platforms: Conjugation to nanoparticles for enhanced sensitivity or targeted delivery

Timeline and Implementation Considerations:

Time HorizonPotential Developments
Near-term (1-2 years)Improved monoclonal antibodies with enhanced specificity and reduced background
Mid-term (3-5 years)Novel antibody formats optimized for specific applications (imaging, quantification)
Long-term (5+ years)Integrated detection systems combining multiple biomarkers with PRAMEF17

While the RFdiffusion approach and similar computational methods show tremendous promise, researchers should note that the translation from computational design to validated research tools requires rigorous experimental validation . The current biotin-conjugated polyclonal PRAMEF17 antibody remains a valuable tool, but researchers should monitor developments in this rapidly evolving field for opportunities to enhance their detection capabilities.

What research gaps exist in our understanding of PRAMEF17 and how can this antibody help address them?

Despite growing interest in the PRAME family of proteins, significant knowledge gaps remain regarding PRAMEF17's biological functions, expression patterns, and potential clinical relevance. PRAMEF17 Antibody (Biotin conjugated) can serve as a crucial tool to address these research gaps:

Current Knowledge Gaps and Research Opportunities:

  • Biological Function and Regulation:

    • Gap: The precise biological function of PRAMEF17 remains poorly characterized compared to other PRAME family members

    • Research Approach: Use PRAMEF17 Antibody in co-immunoprecipitation studies to identify interaction partners

    • Methodological Strategy: Employ biotin-streptavidin pull-down followed by mass spectrometry to elucidate protein-protein interactions

    • Expected Insight: Identification of potential signaling pathways and molecular mechanisms involving PRAMEF17

  • Expression Patterns Across Normal and Pathological Tissues:

    • Gap: Comprehensive tissue expression profiling of PRAMEF17 is lacking

    • Research Approach: Systematic IHC analysis using PRAMEF17 Antibody across tissue microarrays

    • Methodological Strategy: Develop standardized scoring systems to quantify expression in different cellular compartments

    • Expected Insight: Creation of an expression atlas to identify tissues of interest for functional studies

  • Relationship to Other PRAME Family Members:

    • Gap: The functional redundancy or specialization among PRAME family proteins is not well understood

    • Research Approach: Comparative analysis of PRAMEF17 versus other family members in the same samples

    • Methodological Strategy: Multiplex immunoassays combining PRAMEF17 Antibody with antibodies against other family members

    • Expected Insight: Determination of co-expression patterns and potential functional relationships

  • Post-Translational Modifications:

    • Gap: The post-translational modification landscape of PRAMEF17 is largely unknown

    • Research Approach: Immunoprecipitation using PRAMEF17 Antibody followed by PTM-specific analysis

    • Methodological Strategy: Mass spectrometry to identify phosphorylation, ubiquitination, or other modifications

    • Expected Insight: Understanding of regulatory mechanisms controlling PRAMEF17 function

  • Clinical Relevance in Cancer:

    • Gap: Potential diagnostic, prognostic, or therapeutic relevance of PRAMEF17 in cancer remains unexplored

    • Research Approach: Correlative studies of PRAMEF17 expression with clinical outcomes

    • Methodological Strategy: Large-scale IHC studies using PRAMEF17 Antibody on clinically annotated samples

    • Expected Insight: Determination of potential value as a biomarker or therapeutic target

Methodological Challenges and Solutions:

Research ChallengeTechnical LimitationPotential Solution
Low expression levelsDetection sensitivityImplement tyramide signal amplification with biotin-conjugated antibody
Cross-reactivity concernsAntibody specificityValidate with knockdown/knockout controls and peptide competition
Sample availabilityLimited human tissuesDevelop PDX models that maintain PRAMEF17 expression
Functional redundancyMultiple family membersCombine with CRISPR approaches targeting multiple family members

PRAMEF17 Antibody (Biotin conjugated) provides researchers with a valuable tool to address these knowledge gaps, particularly when integrated with complementary approaches such as genomics, proteomics, and functional studies. The biotin conjugation offers flexibility in detection methods, enabling diverse experimental approaches to elucidate the biology and potential clinical significance of this understudied protein .

How might PRAMEF17 research intersect with emerging immunotherapy approaches?

The intersection of PRAMEF17 research with emerging immunotherapy approaches represents an exciting frontier that could potentially leverage this PRAME family protein for targeted cancer treatment. While specific immunotherapy applications of PRAMEF17 remain to be fully explored, insights can be drawn from research on related PRAME family proteins :

Potential Immunotherapeutic Applications:

  • Cancer Vaccine Development:

    • Rationale: PRAME family proteins, as cancer-testis antigens, demonstrate restricted expression in normal tissues but frequent expression in malignancies

    • Research Approach: PRAMEF17 Antibody (Biotin) could help identify tumor types with high expression suitable for vaccine targeting

    • Therapeutic Potential: Development of peptide vaccines derived from PRAMEF17 epitopes to elicit anti-tumor immune responses

    • Assessment Method: Monitor T-cell responses against PRAMEF17 epitopes using tetramer assays or ELISpot

  • CAR-T and TCR-Engineered T Cell Therapy:

    • Rationale: Engineered T cells targeting tumor-associated antigens have shown remarkable efficacy in certain cancers

    • Research Approach: Use PRAMEF17 Antibody to validate surface accessibility of epitopes

    • Therapeutic Potential: Development of CAR-T cells or TCR-engineered T cells targeting PRAMEF17-expressing tumors

    • Assessment Method: Cytotoxicity assays against cell lines with variable PRAMEF17 expression levels

  • Antibody-Drug Conjugates (ADCs):

    • Rationale: Targeted delivery of cytotoxic payloads to tumor cells expressing specific antigens

    • Research Approach: PRAMEF17 Antibody studies to determine internalization kinetics and epitope abundance

    • Therapeutic Potential: Development of ADCs using anti-PRAMEF17 antibodies conjugated to potent cytotoxins

    • Assessment Method: Evaluation of selective killing of PRAMEF17-expressing cells versus negative controls

  • Immune Checkpoint Modulation:

    • Rationale: Combination of tumor antigen recognition with immune checkpoint inhibition can enhance anti-tumor responses

    • Research Approach: Multiplex analysis using PRAMEF17 Antibody with immune checkpoint markers

    • Therapeutic Potential: Rational combination of PRAMEF17-targeted therapies with checkpoint inhibitors

    • Assessment Method: Analysis of immune infiltrates in relation to PRAMEF17 expression patterns

Emerging Research Directions:

ApproachResearch QuestionMethodology Using PRAMEF17 Antibody (Biotin)
Neoantigen PredictionDoes PRAMEF17 harbor potential neoantigens?Epitope mapping combined with MHC binding prediction
Immune Infiltrate CorrelationDoes PRAMEF17 expression correlate with immune cell composition?Multiplex IHC with immune cell markers
Response BiomarkerCan PRAMEF17 expression predict immunotherapy response?Retrospective analysis of treated cohorts
Epigenetic RegulationCan epigenetic modifiers increase PRAMEF17 expression?Treatment studies with epigenetic drugs followed by antibody detection

Translational Research Framework:

  • Preclinical Validation:

    • Comprehensive expression profiling across tumor types using PRAMEF17 Antibody

    • In vitro cytotoxicity studies with engineered immune cells

    • In vivo efficacy studies in xenograft or syngeneic models

  • Biomarker Development:

    • Standardization of PRAMEF17 detection protocols for patient selection

    • Threshold determination for "PRAMEF17-positive" status

    • Correlation of expression with outcomes in immunotherapy trials

  • Combination Strategy Exploration:

    • Testing synergy between PRAMEF17-targeted approaches and established immunotherapies

    • Investigation of resistance mechanisms using PRAMEF17 Antibody-based monitoring

    • Rational design of multi-targeted immunotherapeutic approaches

While directly applicable clinical data on PRAMEF17 in immunotherapy remains limited, the biotin conjugation of the antibody provides versatility for detection across multiple experimental platforms, facilitating translation from bench to bedside .

What are the key considerations for developing quantitative assays for PRAMEF17 detection?

Developing robust quantitative assays for PRAMEF17 detection using PRAMEF17 Antibody (Biotin conjugated) requires careful attention to assay design, validation, and standardization. Here's a comprehensive framework for researchers:

Assay Development Considerations:

  • Platform Selection Based on Research Needs:

Quantitative PlatformAdvantagesLimitationsOptimal Applications
Quantitative ELISAHigh throughput; Standardized protocolsSample preparation requirements; Limited spatial informationPopulation screening; Biomarker validation
Quantitative IHC/IFSpatial context; Cellular resolutionSubjective interpretation; Technical variabilityTumor heterogeneity assessment; Localization studies
Bead-Based MultiplexMultiple analytes simultaneously; Small sample volumeHigher cost; Complex optimizationPathway analysis; Limited sample availability
Automated Western BlotMolecular weight confirmation; Denatured epitopesLower throughput; Semi-quantitativeIsoform analysis; PTM detection
  • Standard Curve Development:

    • Recombinant Protein Standards: Generate or source recombinant PRAMEF17 protein

    • Calibration Range: Develop standard curve spanning expected physiological/pathological range

    • Matrix Matching: Prepare standards in similar matrix as test samples

    • Internal Controls: Include spike-in controls to assess recovery efficiency

  • Assay Optimization Parameters:

    • Antibody Titration: Determine optimal PRAMEF17 Antibody concentration for linear response

    • Sample Preparation Protocol: Standardize extraction methods to maximize recovery

    • Incubation Conditions: Optimize time, temperature, and buffer composition

    • Detection System: Select appropriate streptavidin conjugate for desired sensitivity and dynamic range

    • Signal Amplification: Consider tyramide signal amplification for low-abundance detection

Validation Requirements for Quantitative Assays:

  • Analytical Validation Metrics:

    • Limit of Detection (LOD): Determine lowest detectable concentration (typically 3SD above background)

    • Limit of Quantification (LOQ): Establish lowest reliably quantifiable concentration (typically 10SD above background)

    • Linearity: Verify linear relationship between concentration and signal

    • Dynamic Range: Determine the concentration range providing linear response

    • Precision: Assess intra-assay and inter-assay variability (CV typically <15% for biomarker assays)

    • Accuracy: Compare with reference method when available

    • Specificity: Confirm absence of interference from related proteins

  • Sample-Specific Considerations:

    • Matrix Effects: Assess potential interference from sample components

    • Stability Testing: Determine analyte stability under various storage conditions

    • Sample Processing Impact: Evaluate effects of freeze-thaw cycles, fixation, etc.

  • Clinical/Research Validation:

    • Reference Ranges: Establish normal reference ranges in relevant populations

    • Clinical Correlation: Correlate with established markers or clinical outcomes

    • Cross-Platform Comparison: Compare results with orthogonal detection methods

Implementation Strategies:

  • Quality Control Framework:

    • Run Controls: Include low, medium, and high concentration controls in each assay

    • Acceptance Criteria: Establish criteria for assay validity (control recovery, CV limits)

    • Trending Analysis: Monitor assay performance over time

    • Proficiency Testing: Participate in inter-laboratory comparison when available

  • Data Analysis Approaches:

    • Curve Fitting: Select appropriate algorithm (4PL, 5PL) for standard curve generation

    • Outlier Analysis: Develop criteria for identification and handling of outliers

    • Normalization Strategies: Consider normalization to total protein or housekeeping proteins

    • Statistical Power: Calculate sample sizes needed for desired statistical confidence

By addressing these considerations systematically, researchers can develop robust quantitative assays for PRAMEF17 detection that provide reliable and reproducible results across different research applications. The biotin conjugation of PRAMEF17 Antibody facilitates integration with various detection platforms, enabling flexible assay development tailored to specific research needs .

What are the key takeaways for researchers working with PRAMEF17 Antibody, Biotin conjugated?

Researchers working with PRAMEF17 Antibody (Biotin conjugated) should consider several key principles to maximize experimental success and data reliability. This antibody represents an important tool for investigating the biology and potential clinical relevance of PRAMEF17, a member of the PRAME family that has emerging significance in cancer research .

First, thorough validation is essential before applying this antibody to specific research questions. This includes verification of specificity through appropriate positive and negative controls, optimization of experimental conditions for each application, and careful consideration of potential cross-reactivity with other PRAME family members. The biotin conjugation offers advantages in terms of detection flexibility but requires special attention to endogenous biotin blocking in certain applications .

Second, researchers should recognize both the strengths and limitations of this reagent. While validated for ELISA applications, adaptation to other techniques such as IHC or Western blotting requires careful optimization and validation. The polyclonal nature of the antibody may provide robust detection across multiple epitopes but could also introduce batch-to-batch variability .

Third, interpretation of PRAMEF17 expression data should be contextualized within the broader understanding of PRAME family proteins. Drawing from studies of related family members, researchers should consider subcellular localization patterns, potential co-expression with other cancer biomarkers, and correlation with clinical parameters when designing experiments and analyzing results .

Fourth, emerging computational approaches to antibody design suggest that more specific and optimized detection reagents for PRAMEF17 may become available in the future. Researchers should stay informed about these developments while continuing to apply rigorous methodology with current tools .

Finally, the field of PRAMEF17 research contains significant knowledge gaps that present opportunities for novel discoveries. By employing this antibody in well-designed experiments addressing biological function, expression patterns, and potential clinical applications, researchers can make meaningful contributions to our understanding of this protein and its relevance in health and disease.

How might research techniques for PRAMEF17 detection evolve in the coming years?

The landscape of PRAMEF17 detection methodologies is poised for significant evolution in the coming years, driven by technological advances, computational approaches, and increasing interest in precision biomarkers. While the current PRAMEF17 Antibody (Biotin conjugated) represents a valuable research tool, several developments are likely to transform how researchers detect and quantify this protein .

In the near term, we can expect refinements in antibody-based detection through advanced computational design approaches. The RFdiffusion technology highlighted in recent literature demonstrates the potential for atomically accurate design of antibodies with precise epitope targeting . For PRAMEF17 research, this could translate to next-generation antibodies with enhanced specificity, reduced background, and optimized binding properties. These improvements would address current limitations in cross-reactivity and sensitivity, particularly important for distinguishing PRAMEF17 from other PRAME family members.

Multiplexed detection systems will likely become increasingly prominent, allowing simultaneous analysis of PRAMEF17 alongside other biomarkers. Technologies such as imaging mass cytometry, digital spatial profiling, and multiplexed ion beam imaging (MIBI) enable highly multiplexed protein detection with spatial resolution. Integration of PRAMEF17 detection into these platforms would provide unprecedented insights into its expression patterns in relation to the tumor microenvironment and other cancer-related proteins .

Single-cell analysis techniques represent another frontier for PRAMEF17 research. As single-cell proteomics technologies mature, researchers will gain the ability to examine PRAMEF17 expression at the individual cell level, revealing heterogeneity within tissues that bulk analysis methods cannot detect. This will be particularly valuable for understanding PRAMEF17's role in cancer, where cellular heterogeneity is a critical factor in disease progression and treatment response.

Liquid biopsy approaches may also extend PRAMEF17 detection beyond tissue samples. Development of highly sensitive assays for detecting PRAMEF17 in circulation (either as free protein or in extracellular vesicles) could provide minimally invasive methods for monitoring expression in research and potentially clinical settings. This would require significant advancements in assay sensitivity but aligns with broader trends in biomarker research.

Finally, integration of artificial intelligence and machine learning approaches will likely transform data analysis for PRAMEF17 detection. Automated image analysis algorithms for quantifying immunohistochemistry, pattern recognition in multiplexed datasets, and predictive modeling of PRAMEF17 expression in relation to clinical outcomes represent promising applications of these computational tools.

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.