CML5 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to CML5 Antibody

The CML5 Antibody (anti-CD5) is a monoclonal antibody developed for diagnostic and therapeutic applications in CD5+ leukemias and lymphoproliferative disorders. CD5 is a 67 kDa transmembrane glycoprotein expressed on mature T-cells, thymocytes, and subsets of B-cells, including leukemic cells in CML and chronic lymphocytic leukemia (CLL) . Its overexpression in malignant cells makes it a biomarker for disease characterization and a target for immunotherapy.

Antibody Structure

  • Format: Humanized IgG with a PE-Cyanine5.5 conjugate for flow cytometry applications .

  • Epitope: Binds the CD5 extracellular domain, facilitating cell surface labeling and internalization .

  • Isoforms: Recognizes both membrane-bound and soluble CD5 variants .

Mechanism of Action

  • Diagnostic Use: Binds CD5 on leukemic cells for detection via flow cytometry or immunohistochemistry .

  • Therapeutic Potential: Induces antibody-dependent cellular cytotoxicity (ADCC) by recruiting NK cells or macrophages .

Flow Cytometry

The 14-color CD5+ assay quantifies CD5 expression in CML and CLL cells, enabling minimal residual disease (MRD) monitoring. Key performance metrics include :

AntigenPositive PopulationSeparation RatioClinical Relevance
CD5 PECD3+ T-cells131Distinguishes T/B-cell LPDs
CD19 PC7CD20+ B-cells76Identifies B-cell malignancies

Targeting CML Stem Cells

  • CD5 is overexpressed in CML progenitor cells .

  • Preclinical studies show anti-CD5 antibodies enhance TKI efficacy by eliminating residual leukemic stem cells .

Preclinical Efficacy

  • ADCC Activity: Autologous NK cells from CML patients effectively lyse CD5+ leukemic cells when combined with anti-CD5 antibodies .

  • Combination Therapy: Anti-CD5 + TKIs reduce leukemic engraftment in murine models by 85% compared to monotherapy .

Immune Modulation

  • CD5 blockade reduces immunosuppressive Tregs and MDSCs in CML patients, restoring cytotoxic T-cell responses to leukemia-associated antigens (e.g., WT1, PRAME) .

Limitations

  • Tumor Heterogeneity: CD5 expression varies across CML subtypes, limiting universal applicability .

  • Off-Target Effects: CD5 is expressed on normal T-cells, risking immunosuppression .

Innovative Strategies

  • Bispecific Antibodies: Combining anti-CD5 with anti-CD123 (IL-3Rα) enhances specificity for leukemic stem cells .

  • CAR-T Cell Therapy: CD5-targeting CAR-T cells are under investigation for relapsed/refractory CML .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CML5 antibody; MSS3 antibody; At2g43290 antibody; F14B2.33 antibody; T1O24.3Calmodulin-like protein 5 antibody; Protein MULTICOPY SUPPRESSORS OF SNF4 DEFICIENCY IN YEAST 3 antibody
Target Names
CML5
Uniprot No.

Target Background

Function
CML5 is a potential calcium sensor.
Gene References Into Functions
  1. Phylogenetic analysis indicates that AtCML4 and AtCML5 are closely related paralogues, likely arising from a duplication event within the Brassicaceae family. These findings suggest that CML4/5-like proteins constitute a flowering plant-specific subfamily of CMLs, potentially playing a role in vesicle transport within the plant endomembrane system. PMID: 27029353
Database Links

KEGG: ath:AT2G43290

STRING: 3702.AT2G43290.1

UniGene: At.21047

Protein Families
Calmodulin family

Q&A

What is CML5 Antibody and what is its target specificity?

CML5 antibody is a polyclonal antibody raised in rabbits against recombinant Oryza sativa subsp. japonica (Rice) CML5 protein . While the name suggests potential relevance to Chronic Myeloid Leukemia (CML), it's important to distinguish that commercial CML5 antibodies are primarily designed for plant research applications.

For leukemia research, several other antibodies have demonstrated specificity for leukemia-associated antigens. Studies have shown that patients with chronic myeloid leukemia (CML) develop circulating antibodies against leukemic blast cells, which show reactivity against myeloblasts but not against remission lymphocytes or normal lymphocytes . These antibodies recognize multiple leukemia-derived proteins, with research identifying at least eight distinct proteins that elicit immune responses in CML patients .

How are antibodies against leukemia antigens detected in research settings?

Detection of antibodies against leukemia-associated antigens employs multiple complementary techniques:

  • Cytotoxicity Assays: These measure the ability of antibodies to kill target cells, providing functional evidence of antibody activity against leukemia cells .

  • Immunofluorescence Assays: These visualize antibody binding to target cells and can detect circulating antibodies against blast cells .

  • Expression cDNA Library Screening: This technique involves constructing a cDNA library from leukemia cells and screening it with autologous serum to identify proteins that elicit high-titer IgG antibodies .

  • Northern Blot Analysis: This evaluates mRNA expression patterns of potential target antigens in normal tissues versus leukemic cells .

  • Dot Blot Analysis: Used to screen sera from patients and controls for antibodies against specific clones using bacterial lysates .

These methodologies allow researchers to comprehensively characterize antibody responses to leukemia-associated antigens for both basic research and potential therapeutic applications.

What is the relationship between antibody responses and disease progression in leukemia?

The relationship between antibody responses and disease progression appears significant, though complex. Studies of CML patients have observed that patients in the stable phase of disease develop circulating antibodies against leukemic blast cells, while those in the aggressive phase often fail to produce such antibodies despite repeated immunotherapy .

Specifically, eight patients in the stable phase of CML developed circulating antibodies against immunizing blast cells. Seven of these eight patients maintained a steady clinical course ranging from 20 to 40 months, while only one entered the blastic phase and died . In contrast, six patients in the aggressive phase of CML showed progressive leukocytosis, splenomegaly, or increasing myeloblastosis. Five of these patients died within approximately 16 months of diagnosis, and notably, humoral antibodies were not detected in these patients despite repeated immunotherapy .

This suggests that the capacity to mount an antibody response may correlate with disease stability, though the causal relationship remains unclear – it's possible that immune competence facilitates antibody production and also contributes to disease control, rather than the antibodies themselves being protective.

How do researchers distinguish between antibodies that target common versus leukemia-specific antigens?

Distinguishing between antibodies targeting common versus leukemia-specific antigens requires careful experimental design involving multiple control samples and cross-reactivity testing:

These approaches collectively help researchers identify antibodies with genuine specificity for leukemia-associated antigens, which is essential for developing targeted diagnostic and therapeutic applications.

What methodological approaches are used for characterizing novel anti-leukemia antibodies?

Characterization of novel anti-leukemia antibodies involves multiple methodological approaches addressing different aspects of antibody properties:

  • Initial Hybridization and Selection: Novel antibodies are often obtained through hybridization of mouse spleen lymphocytes immunized with blast cells from leukemia patients . This process involves screening large numbers of hybridomas to identify those producing antibodies with desired specificity.

  • Epitope Characterization: Researchers must determine whether an antibody recognizes a new epitope or one already described. This typically involves competitive binding assays with established antibodies and epitope mapping techniques .

  • Internalization Studies: Evaluating whether antibodies efficiently internalize through the cell membrane is critical for applications delivering toxins or therapeutic payloads. Some anti-CD5 monoclonal antibodies have demonstrated efficient internalization, suggesting potential for delivering toxins to immunocompetent T lymphocytes .

  • Multi-well Adapted Immunoprecipitation: For detecting antibodies in patient sera, specialized techniques such as immunoprecipitation assays using radiolabelled recombinant proteins produced by coupled in vitro transcription/translation provide sensitive detection methods .

  • Validation Across Multiple Techniques: Comprehensive characterization involves testing antibody reactivity using multiple techniques, as different methods have varying sensitivities. For example, CRMP5 antibodies were more frequently detected by immunoprecipitation than by immunofluorescence or immune blots .

These methodological approaches provide complementary information about antibody characteristics, enabling researchers to fully evaluate potential research and clinical applications.

How should researchers design experiments to evaluate the therapeutic potential of anti-leukemia antibodies?

Designing experiments to evaluate therapeutic potential requires systematic approaches across multiple levels:

  • In Vitro Efficacy Assessment:

    • Cytotoxicity assays comparing antibody effects on leukemic versus healthy cells

    • Evaluation of multiple mechanisms of action (complement-mediated cytotoxicity, antibody-dependent cellular cytotoxicity, direct induction of apoptosis)

    • Assessment of antibody internalization properties when considering antibody-drug conjugates

  • Patient Sample Testing:

    • Testing against both cell lines and primary patient samples to ensure clinical relevance

    • Evaluation across samples representing different disease stages to assess stage-specific efficacy

    • Testing against the patient's own leukemic cells to confirm autologous activity

  • Combination Approaches:

    • Evaluation alongside standard chemotherapy to assess potential synergies

    • Combined treatment with immune modulators (e.g., BCG vaccine) to enhance antibody responses

  • Dose-Response Relationships:

    • Systematic evaluation of different antibody concentrations

    • Assessment of dosing schedules (e.g., weekly for 4 weeks followed by monthly intervals)

  • Therapeutic Window Determination:

    • Comparison of effects on malignant versus normal cells to establish therapeutic window

    • As noted in development of a novel monoclonal antibody for myelofibrosis: "It's quite rare to get such a wide therapeutic window of opportunity"

These experimental design elements collectively provide robust assessment of therapeutic potential, addressing both efficacy and safety considerations necessary for advancing antibodies toward clinical application.

What control samples are essential when evaluating antibody specificity in leukemia research?

Essential control samples for rigorous evaluation of antibody specificity include:

Control TypePurposeExample From Research
Normal LymphocytesEstablish baseline reactivity with healthy cellsStudies used normal lymphocytes from 20 donors
Remission LymphocytesDetermine whether antibodies target leukemia-specific or general lymphocyte antigensAntibodies showing reactivity against myeloblasts but not corresponding remission lymphocytes from five patients
Panel of MyeloblastsAssess cross-reactivity across different leukemia samplesTesting against myeloblasts from 12 patients
Patient's Own Leukemic BlastsConfirm autologous reactivityTwo cases where antibodies showed reactivity against the patient's own leukemic blasts
Multiple Cell LinesEvaluate specificity across diverse cellular contextsTesting expression in normal bone marrow, leukemia cell lines, fresh leukemia cells, and normal tissues
Normal TissuesAssess potential off-target bindingExpression analysis in normal tissues compared to leukemic cells
Healthy Control SeraEstablish background antibody prevalence in general populationScreening sera from 300 healthy blood donors for CRMP5 antibodies

Incorporating these controls enables researchers to comprehensively characterize antibody specificity, distinguishing between antibodies with broad reactivity versus those with leukemia-specific targeting properties. This information is crucial for both basic research applications and therapeutic development.

How do researchers correlate antibody responses with clinical outcomes in leukemia patients?

Correlating antibody responses with clinical outcomes requires comprehensive patient monitoring alongside immunological assessment:

  • Longitudinal Patient Monitoring: Following patients over extended periods while measuring antibody responses at regular intervals. Research has tracked CML patients for 20-40 months to correlate antibody production with disease stability .

  • Clinical Staging Correlation: Classifying patients according to disease phase and comparing antibody responses between stages. Studies have distinguished between "stable phase" and "aggressive phase" CML, observing different antibody response patterns .

  • Multivariate Analysis: Accounting for confounding factors such as prior treatments, demographic characteristics, and comorbidities that might affect both antibody responses and clinical outcomes.

  • Antibody Titer Quantification: Measuring not just presence/absence but antibody levels, as higher titers may correlate with stronger clinical responses.

  • Functional Antibody Assessment: Evaluating antibody functions (complement activation, ADCC, internalization) to determine which properties correlate with improved outcomes.

The correlation between antibody responses and outcomes appears significant. In one study, seven of eight patients who developed circulating antibodies maintained stable disease for 20-40 months, while patients in aggressive disease phases failed to produce antibodies despite repeated immunotherapy . This suggests antibody responses may be important prognostic indicators, though larger studies are needed to establish definitive correlations.

What challenges exist in translating promising antibody-based approaches from laboratory to clinical applications?

Translating antibody-based approaches to clinical applications faces several key challenges:

  • Antibody Specificity:

    • Ensuring antibodies specifically target leukemia cells while sparing healthy tissues

    • Distinguishing between ubiquitously expressed proteins and those with restricted expression in leukemic cells

    • Addressing potential cross-reactivity with normal tissues that could lead to toxicity

  • Immunogenicity Concerns:

    • Early antibody therapeutics like the murine anti-CD5 antibody T101 encountered limitations partly due to immunogenicity

    • Evolution from murine to humanized antibodies (like CAMPATH-1M to CAMPATH-1H) to reduce immunogenicity while maintaining efficacy

  • Variable Patient Responses:

    • Understanding why antibody responses differ between disease phases

    • Identifying biomarkers that predict which patients will benefit from antibody-based therapies

  • Technical Manufacturing Challenges:

    • Producing consistent antibody preparations with preserved specificity and functionality

    • Developing effective antibody-drug conjugates or CAR T-cell approaches that maintain targeting precision

  • Clinical Trial Design:

    • Establishing appropriate endpoints for evaluating efficacy of antibody therapies

    • Determining optimal timing of antibody therapy within treatment protocols

Addressing these challenges requires multidisciplinary collaboration between laboratory researchers and clinical investigators to develop effective strategies for clinical translation of promising antibody-based approaches.

How might novel antibody engineering approaches enhance targeting of leukemia-specific antigens?

Novel antibody engineering approaches offer several promising avenues for enhancing leukemia-specific targeting:

  • Neoepitope-Directed Antibodies: Developing antibodies that specifically recognize mutated proteins found only in cancer cells. Recent work with CALR mutations in myeloproliferative neoplasms demonstrates this approach: "What's exciting is that our antibody works directly against the mutant protein that's encoded by the CALR mutation by binding to it and pushing it off the surface of the cell so it can no longer signal" . This approach targets "a problematic disease driven by a recurrent somatic mutation normally considered undruggable" .

  • Bispecific Antibody Development: Creating antibodies that simultaneously bind to leukemia cells and immune effectors, bringing cytotoxic T cells into proximity with target cells. This approach enhances natural immune surveillance mechanisms while maintaining specificity.

  • Antibody-Drug Conjugates (ADCs): Coupling antibodies with cytotoxic payloads that are released after internalization. The efficient internalization properties observed with some anti-CD5 monoclonal antibodies make them promising candidates for delivering toxins to target cells .

  • CAR T-Cell Derivation: Using the binding portion of therapeutic antibodies to develop CAR T-cells with curative potential. As noted in recent research: "We can also take the binding portion of the antibody and turn that into a CAR T-cell that could have curative potential" .

  • Combinatorial Targeting: Developing antibody cocktails targeting multiple leukemia-associated antigens simultaneously to minimize escape variants and enhance therapeutic efficacy.

These engineering approaches leverage growing understanding of leukemia-specific antigens and antibody biology to create increasingly precise therapeutic tools for targeting leukemic cells while minimizing effects on healthy tissues.

What emerging methods are advancing our understanding of the immunogenicity of leukemia cells?

Several emerging methodologies are advancing our understanding of leukemia cell immunogenicity:

  • Expression cDNA Library Screening: This technique has enabled identification of multiple immunogenic proteins from leukemia cells. Researchers constructed a cDNA library from CML patient leukemia cells and used autologous serum to screen for proteins eliciting high-titer IgG antibodies, identifying eight distinct clones that suggested multiple immune responses were elicited in the host .

  • Neoepitope Identification: Advanced sequencing and computational approaches enable identification of leukemia-specific mutations that create novel epitopes recognizable by the immune system. These "little neoepitopes that occur and that are not normally found on normal cells" represent promising targets for antibody development .

  • Single-Cell Technologies: Single-cell RNA sequencing and proteomics provide unprecedented resolution of cellular heterogeneity within leukemia populations, enabling identification of subpopulation-specific antigens and immune interactions.

  • Functional Immune Assays: Beyond antibody detection, functional assays evaluating T-cell responses, NK cell activation, and dendritic cell presentation provide broader understanding of anti-leukemia immunity.

  • Longitudinal Immune Monitoring: Tracking immune responses throughout disease progression and treatment provides insights into the dynamic relationship between leukemia and the immune system, potentially identifying critical timepoints for therapeutic intervention.

These methodologies collectively enhance our understanding of leukemia immunogenicity, revealing both the mechanisms by which leukemia cells evade immune detection and potential vulnerabilities that can be exploited for therapeutic benefit through antibody-based approaches.

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.