NPM1 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

NPM1 monoclonal antibodies are laboratory-produced immunoglobulins designed to bind selectively to epitopes on the NPM1 protein. Key features include:

PropertyDetails
Target ProteinNPM1 (Nucleophosmin 1, B23)
Molecular Weight~33–37 kDa (varies by isoform and post-translational modifications)
Subcellular LocalizationNucleus (wild-type), cytoplasm (mutant NPM1 in AML)
Mutant-Specific RecognitionAntibodies like T26 and clone 2E2 detect cytoplasmic mutant NPM1 in AML

Immunohistochemistry (IHC)

  • Detects NPM1 expression in formalin-fixed, paraffin-embedded tissues (e.g., skin, bone marrow) at concentrations of 1–3 µg/ml .

  • Mutant-specific clones (e.g., clone 3F9) differentiate cytoplasmic NPM1 in AML cells from nuclear wild-type NPM1 .

Flow Cytometry and Immunofluorescence

  • Identifies NPM1 mutations in AML patient samples with high specificity (100% concordance with molecular testing) .

  • TCR-like antibody 2E2 binds NPM1 mutant/HLA-A2 complexes on leukemia cells, enabling immunotherapeutic targeting .

Western Blotting

  • Validates NPM1 expression in cell lines (e.g., OCI-AML3, HeLa) at ~37 kDa .

Diagnostic Utility

  • Mutant Detection: Antibodies such as T26 recognize the C-terminal mutant epitope in ~95% of NPM1-mutated AML cases, aiding rapid diagnosis .

  • Minimal Residual Disease (MRD) Monitoring: Clone 3F9 detects residual leukemic cells in post-treatment bone marrow samples .

Therapeutic Potential

  • Checkpoint Inhibition Enhancement: Anti-PD-1 antibodies augment T-cell cytotoxicity against NPM1-mutated AML cells in vitro .

  • Targeted Immunotherapy: TCR-like antibody 2E2 demonstrates nanomolar affinity (KD = 158 nM) for NPM1 mutant/HLA-A2 complexes, showing promise in preclinical models .

Cross-Reactivity and Specificity

NPM1 monoclonal antibodies undergo rigorous validation:

AssayFindings
Protein Microarray (19,000 human proteins)Z-score >43 for NPM1; S-score >29 confirms specificity
Mutant vs. Wild-TypeClone T26 shows no cross-reactivity with wild-type NPM1
Tumor-SpecificityClone 3F9 unreactive with 93 non-NPM1-mutated AMLs and 150 non-AML tumors

Research Findings

  • Mechanistic Insights: Cytoplasmic NPM1 mutants disrupt ARF/p53 signaling, promoting leukemogenesis .

  • Therapeutic Resistance: NPM1-mutated cells upregulate PD-L1, suggesting combinatorial therapy with αPD-1 antibodies .

Limitations and Challenges

  • Cross-Reactivity: Some antibodies (e.g., 2G3, 3F9) weakly bind wild-type NPM1, necessitating confirmatory molecular testing .

  • Epitope Variability: Not all NPM1 mutations (e.g., rare subtypes beyond A, B, D) are detected by existing clones .

Product Specs

Form
Purified mouse monoclonal antibody in a buffer containing 0.1M Tris-Glycine (pH 7.4, 150 mM NaCl) with 0.2% sodium azide and 50% glycerol.
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery times.
Synonyms
nucleophosmin numatrin nucleolar protein NO38 nucleophosmin/nucleoplasmin family, member

Q&A

What is the significance of NPM1 mutations in acute myeloid leukemia?

NPM1 mutations represent the most frequent genetic aberrations in acute myeloid leukemia (AML) and define a clinically distinct subset of this disease. These mutations are essential for initiating malignant transformation during the early stages of leukemogenesis. The mutated NPM1 protein is retained in AML cells to maintain its malignant phenotype, with a relatively conserved alternative reading frame, positioning it as an ideal target for AML immunotherapy and diagnostics . NPM1-mutated AML is recognized as a distinct entity in WHO and ICC classifications, characterized by unique clinical and molecular features, including the aberrant cytoplasmic dislocation of the NPM1 mutant protein that is critical for leukemogenesis .

How do NPM1 monoclonal antibodies differ from conventional AML diagnostic tools?

NPM1 monoclonal antibodies provide a rapid, simple, and cost-effective molecular diagnostic approach compared to conventional sequencing methods. They enable direct visualization of the mutant protein rather than detecting the genetic alteration. Unlike PCR-based techniques, antibody-based detection can be performed using routine laboratory techniques such as immunofluorescence, immunohistochemistry, and flow cytometry without specialized molecular biology equipment . Additionally, antibody-based methods allow for simultaneous assessment of protein localization and cellular morphology, providing insights into the relationship between NPM1 mutations and cellular differentiation that are not possible with genetic testing alone .

What are the main types of NPM1 monoclonal antibodies currently available for research?

Several significant NPM1 monoclonal antibodies have been developed for research purposes:

  • T26 antibody: Raised against a 19-amino acid polypeptide containing the unique C-terminus of the type A NPM1 mutant protein. It recognizes 10 of the 21 known NPM1 mutants, including types A, B, and D, which cover approximately 95% of all cases .

  • TCR-like fully human IgG1 antibody: Developed to target the NPM1 mut A 283-291/HLA0201 complex as a neoantigen, mimicking T-cell receptor recognition mechanisms .

  • Recent tumor-specific mAb: Specifically recognizes mutant but not wild-type NPM1 protein, suitable for immunostaining of bone marrow paraffin sections with high specificity (positive in 128/128 NPM1-mutated AMLs but negative in 93 AMLs with different genotypes) .

How should NPM1 monoclonal antibodies be validated for experimental use?

Proper validation of NPM1 monoclonal antibodies requires a multi-step approach:

  • Specificity verification: Test against known NPM1-mutated and wild-type samples. A properly validated antibody should demonstrate selective binding to mutant forms without cross-reactivity with wild-type NPM1 or unrelated cellular proteins . For example, the T26 antibody recognized 10 of 21 known NPM1 mutants without cross-reacting with wild-type NPM1 .

  • Cross-platform validation: Verify performance across different detection methodologies (immunofluorescence, immunohistochemistry, flow cytometry) to ensure consistent results . The T26 antibody demonstrated efficiency across all these platforms.

  • Clinical correlation: Compare antibody-based detection with molecular confirmation of NPM1 mutation status. In validation studies, all antibody-positive cases should be confirmed to carry NPM1 mutations by molecular analysis .

  • Sensitivity assessment: Determine the lower detection limit using dilution experiments with known positive samples, particularly important for minimal residual disease (MRD) applications .

What are the optimal methodological approaches for using NPM1 monoclonal antibodies in bone marrow samples?

When using NPM1 monoclonal antibodies on bone marrow samples, researchers should consider these methodological approaches:

  • For paraffin-embedded sections:

    • Use optimized antigen retrieval protocols to ensure accessibility of the epitope

    • Apply appropriate blocking to minimize background staining

    • Include positive and negative controls in each batch

    • Ensure adequate fixation (neutral-buffered formalin for 12-24 hours) for optimal antigen preservation

  • For flow cytometry applications:

    • Perform permeabilization to allow antibody access to intracellular NPM1

    • Use multiparameter approaches to correlate NPM1 mutant expression with other markers (CD34, CD7)

    • Include adequate controls for permeabilization efficiency

  • For immunofluorescence:

    • Use confocal microscopy for precise subcellular localization studies

    • Employ co-staining with markers for nuclear and cytoplasmic compartments to accurately assess NPM1 localization

How can researchers quantitatively assess NPM1 mutant protein expression using monoclonal antibodies?

Quantitative assessment of NPM1 mutant protein requires standardized approaches:

  • Flow cytometry-based quantification:

    • Use calibration beads with known antibody-binding capacity

    • Calculate molecules of equivalent soluble fluorochrome (MESF) values

    • Establish a standardized mean fluorescence intensity (MFI) ratio between positive samples and negative controls

  • Image analysis for immunohistochemistry:

    • Apply digital image analysis software to quantify staining intensity

    • Establish an H-score (combining intensity and percentage of positive cells)

    • Correlate with variant allele frequency (VAF) data from molecular testing

  • Western blot quantification:

    • Use densitometric analysis with appropriate loading controls

    • Create standard curves with recombinant proteins of known concentration

    • Normalize to total protein or housekeeping proteins

How can NPM1 monoclonal antibodies elucidate the relationship between subcellular localization and leukemogenic activity?

Recent research using NPM1 monoclonal antibodies has revealed important insights regarding subcellular localization and leukemogenic mechanisms:

  • Nuclear versus cytoplasmic distribution: While NPM1 mutant positivity was primarily found in the cytoplasm of leukemic cells, approximately 30% of cases also showed nuclear positivity. This finding was especially evident in leukemic proerythroblasts . This dual localization pattern correlates with recent observations that the NPM1 mutant can interact with XPO1 bound to chromatin to exert its leukemogenic activity .

  • Experimental approaches to study localization:

    • Use confocal microscopy with Z-stack analysis to precisely determine subcellular distribution

    • Employ subcellular fractionation followed by western blotting with NPM1 monoclonal antibodies

    • Perform co-immunoprecipitation with chromatin-associated proteins to confirm nuclear interactions

  • Correlation with differentiation status: In NPM1-mutated AML with monocytic differentiation, the NPM1 mutant protein shows variable expression patterns, with more mature leukemic cells being negative or only weakly positive. This suggests that although the mutation remains present (as evidenced by VAF of 40-50%), the NPM1 mutant protein is lost during monocytic differentiation .

What methodological considerations are important when using NPM1 monoclonal antibodies for clonality assessment?

Clonality assessment with NPM1 monoclonal antibodies requires careful methodological considerations:

  • Limitations in monocytic lineage: The downregulation of NPM1 mutant protein during monocytic differentiation makes it unsuitable as a standalone marker for tracking clonality in mature monocytic cells. Researchers have observed that cells with strong monocytic maturation (including starry sky macrophages) may not express detectable levels of NPM1 mutant protein despite belonging to the leukemic clone .

  • Alternative approaches for clonality tracking:

    • Combine NPM1 antibody staining with other mutation-specific antibodies (such as anti-IDH1 R132H) for co-mutated cases

    • Employ sequential staining protocols to detect multiple antigens in the same cell

    • Use laser capture microdissection of specific cell populations followed by molecular analysis

  • Experimental validation: In studies examining NPM1-mutated AMLs co-mutated for IDH1 R132H, staining with specific anti-IDH1 R132H mAb revealed strong positivity in NPM1-mutant negative mature macrophages, demonstrating that these terminally differentiated cells belong to the leukemic clone despite lacking detectable NPM1 mutant protein .

How do different NPM1 mutation variants affect monoclonal antibody binding and detection sensitivity?

The diversity of NPM1 mutations impacts antibody binding characteristics:

What methodological considerations are important when using NPM1 monoclonal antibodies for minimal residual disease (MRD) monitoring?

NPM1 monoclonal antibodies offer potential for MRD monitoring with specific methodological considerations:

  • Sensitivity thresholds:

    • Immunohistochemistry typically detects 1-5% positive cells

    • Flow cytometry can detect 0.1-0.01% positive cells

    • Compare with molecular methods that can detect 0.001-0.0001% burden

  • Protocol optimization for MRD:

    • Increase antibody incubation time for low-level detection

    • Apply signal amplification methods

    • Use multiparameter approach to reduce false positives

    • Consider acquisition of higher cell numbers for flow cytometry (>500,000 events)

  • Validation against molecular MRD:

    • Establish correlation between antibody-based and PCR-based MRD quantification

    • Determine concordance rates at different disease burden levels

    • Recent studies demonstrated that anti-NPM1 mutant mAb could detect rare NPM1 mutant-expressing cells in patients in molecular relapse

How does the prognostic value of NPM1 mutation detection by monoclonal antibodies compare with molecular methods?

The prognostic assessment using antibody-based methods has shown important correlations:

  • Concordance with genetic testing: Studies show excellent correlation between cytoplasmic NPM1 immunoreactivity and presence of NPM1 mutations. All NPMc+ cases (54% of total) had NPM1 mutations, while none of the nucleus-restricted (NPMc–) cases (46% of total) had NPM1 mutations (P < .0001) .

  • Association with other prognostic markers:

    • NPM1 mutations detected by immunohistochemistry correlate with FLT3-ITD (P = .0062)

    • Association with absence of CD34 (P = .0001) and CD7 (P = .041)

    • Favorable survival outcome in AML-NK cases that were NPM1 mutated and FLT3-ITD non-mutated

  • Methodological approach for prognostic assessment:

    • Combine NPM1 antibody staining with FLT3-ITD testing

    • Correlate with CD34 and CD7 expression by immunophenotyping

    • Integrate findings with other molecular and cytogenetic prognostic factors

What are the technical challenges in developing antibody-based therapies targeting intracellular NPM1 mutant proteins?

Development of antibody-based therapeutics against NPM1 mutant protein faces unique challenges:

  • Intracellular target accessibility:

    • Conventional therapeutic antibodies recognize surface antigens but cannot penetrate intracellular domains

    • Innovative approaches mimic T-cell receptor (TCR) recognition of the complex of major histocompatibility class I and peptide on the cell surface derived from processed intracellular antigen

  • Technical approaches to overcome intracellular barriers:

    • Development of TCR-like fully human IgG1 antibody against NPM1 mut A 283-291/HLA0201 complex

    • Utilization of CHO expression system to produce recombinant complexes

    • Application of mouse hybridoma technology to generate monoclonal antibody cells specific to these complexes

  • Experimental validation methods:

    • Surface plasmon resonance analyses to determine binding kinetics and affinity constants

    • Flow cytometry-based screening for protein-specific monoclonal antibodies

    • Assessment of specificity using control peptides

How should researchers interpret discrepancies between NPM1 protein detection by antibodies and genetic mutation analysis?

Discrepancies between protein-level and genetic-level detection require careful analysis:

  • Sources of potential discordance:

    • Post-transcriptional or post-translational regulation affecting protein expression

    • Technical limitations in antibody sensitivity or specificity

    • Heterogeneous expression within the leukemic population

    • Sampling variations between specimens used for different techniques

  • Methodological approach to resolve discrepancies:

    • Perform sequential testing of the same sample with both methods

    • Use microdissection to isolate specific cell populations for comparative analysis

    • Apply quantitative methods to assess correlation between variant allele frequency and protein expression levels

  • Biological interpretation: Studies have demonstrated that despite consistent VAF of 40-50% in NPM1 mutations, protein expression levels can vary significantly, particularly during monocytic differentiation, suggesting differential protein stability or degradation mechanisms rather than technical limitations .

What statistical approaches are recommended for analyzing NPM1 monoclonal antibody staining patterns across patient cohorts?

Appropriate statistical analysis of NPM1 antibody staining requires:

  • Quantitative assessment methods:

    • Staining intensity scoring (0, 1+, 2+, 3+)

    • Percentage of positive cells quantification

    • Combined H-score calculation (intensity × percentage)

  • Statistical tests for meaningful comparisons:

    • Chi-square or Fisher's exact test for categorical comparisons (positive vs. negative)

    • Mann-Whitney U test for non-parametric continuous data (staining intensity scores)

    • Kaplan-Meier survival analysis with log-rank test for prognostic correlations

    • Multivariate analysis to assess independent prognostic value

  • Data presentation recommendations:

    • Include scatter plots showing distribution of staining intensity/percentage

    • Create contingency tables comparing antibody results with mutation status

    • Present survival curves stratified by NPM1 status and additional prognostic factors

How does the performance of different NPM1 monoclonal antibodies compare in terms of sensitivity and specificity?

Comparative analysis of different NPM1 monoclonal antibodies reveals varied performance characteristics:

AntibodyRecognition SpectrumSensitivitySpecificityDetection MethodsKey Advantages
T2610/21 NPM1 mutants (types A, B, D) covering ~95% of cases40-38% of de novo AML patientsNo cross-reactivity with wild-type NPM1IF, IHC, Flow cytometryFirst antibody specifically recognizing leukemia-associated mutant protein
Recent tumor-specific mAbAll tested NPM1-mutated AMLs (128/128)Detected rare cells in molecular relapseNo reactivity with 93 non-NPM1 mutated AMLs and 150 other tumorsIHC on paraffin sectionsSuperior performance in tissue sections; nuclear + cytoplasmic detection capability
TCR-like human IgG1NPM1 mut A 283-291/HLA0201 complexRequires further clinical validationSignificantly different binding to NPM1 vs. control peptideFlow cytometryPotential therapeutic application; targets intracellular mutant protein

IF: Immunofluorescence, IHC: Immunohistochemistry

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.