CML3 Antibody

Shipped with Ice Packs
In Stock

Description

Target Overview: CD123 in CML Pathobiology

CD123, the α subunit of the interleukin-3 (IL-3) receptor, is overexpressed in CML leukemic stem/progenitor cells (LSPCs) compared to normal hematopoietic cells . Key characteristics:

  • Elevated expression:

    • Blast crisis (BC)-CML: 77.5% of CD34+/CD38− cells express CD123 vs. 20.3% in healthy donors .

    • Chronic phase (CP)-CML: CD123 levels increase with disease progression (53.0% in CP vs. 73.2% in BC) .

  • Functional role: Mediates IL-3–induced survival signals, contributing to tyrosine kinase inhibitor (TKI) resistance .

Mechanistic Action of Anti-CD123 Antibodies

CSL362, a humanized anti-CD123 monoclonal antibody, demonstrates dual mechanisms:

MechanismEffect on CML CellsNormal Cell Impact
ADCC (antibody-dependent cellular cytotoxicity)NK cell-mediated lysis of CD123+ LSPCs (e.g., 60–80% reduction in leukemic engraftment in murine models) Minimal toxicity to CD123low HSCs
IL-3 neutralizationBlocks IL-3 rescue of TKI-induced apoptosis (synergistic effect with TKIs) No IL-3 signaling disruption

Preclinical Efficacy of CSL362

  • In vitro:

    • 85% ADCC-mediated lysis of BC-CML CD34+/CD38− cells using autologous NK cells .

    • Combination with TKIs (e.g., imatinib) reduced CML progenitors by 90% vs. 70% with TKI alone .

  • In vivo:

    • 70% reduction in leukemic engraftment in NSG mice transplanted with CML LSPCs .

Clinical Correlations

  • IL-3 plasma levels: Elevated in untreated CP-CML (4.3 pg/mL) vs. healthy donors (1.1 pg/mL) .

  • NK cell activity: CML patients retain functional NK cells capable of CSL362-mediated ADCC .

Clinical Development Challenges

  • Selectivity: CD123 is expressed on dendritic cells and some HSCs, requiring careful therapeutic window assessment .

  • Resistance: Upregulation of inhibitory ligands (e.g., LGALS9-TIM3) in CML cells may dampen NK cell responses .

Comparative Data: CD123 Expression Across Hematologic Malignancies

DiseaseCD123+ CD34+/CD38− CellsMean Fluorescence Intensity (MFI)
Healthy donors20.3%1.6
CP-CML53.0%2.4
BC-CML73.2%8.9
AML72.4%5.6

Data adapted from .

Future Directions

  • Combination therapies: Pairing with FLT3-targeting bispecific antibodies (e.g., CLN-049) to address heterogeneous antigen expression .

  • Biomarker refinement: Correlating CD123 expression levels with TKI resistance patterns .

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
CML3 antibody; At3g07490 antibody; F21O3.20Calmodulin-like protein 3 antibody
Target Names
CML3
Uniprot No.

Target Background

Function
CML3 is a potential calcium sensor.
Gene References Into Functions
  1. AtCML3 facilitates dimerization of peroxisomal processing protease AtDEG15, playing a crucial role in normal peroxisome metabolism. [AtCML3] PMID: 23943091
  2. CML3 and CML30 exhibit distinct subcellular localizations: peroxisomes for CML3 and mitochondria for CML30. CML3's peroxisomal targeting is mediated by an atypical C-terminal PTS1-like tripeptide, while CML30 utilizes an N-terminal, non-cleavable transit peptide. [CML3] PMID: 22116655
Database Links

KEGG: ath:AT3G07490

STRING: 3702.AT3G07490.1

UniGene: At.53214

Protein Families
Calmodulin family

Q&A

What is the role of IL-3 receptor α subunit (CD123) in chronic myeloid leukemia progression?

CD123 (IL-3 receptor α subunit) serves as an established marker for leukemic stem cells and plays a critical role in chronic myeloid leukemia (CML) progression. Research indicates that CD123 expression is significantly higher in CD34+/CD38- cells of both chronic phase and blast crisis CML patients compared to normal donors, with expression levels increasing as the disease progresses. This receptor mediates cytokine-dependent resistance pathways that allow leukemic stem and progenitor cells (LSPCs) to persist despite tyrosine kinase inhibitor (TKI) therapy. The IL-3 signaling pathway specifically contributes to long-term survival of these cells by activating anti-apoptotic mechanisms and promoting proliferation, which explains why targeting CD123 has emerged as a promising therapeutic strategy .

How do antibodies targeting CD123 affect leukemic stem and progenitor cells in CML?

Antibodies targeting CD123, such as the humanized monoclonal antibody CSL362, effectively eliminate CML leukemic stem and progenitor cells through multiple mechanisms. The primary mechanism involves selective antibody-dependent cell-mediated cytotoxicity (ADCC), which facilitates the lysis of CD123+ cells. This process significantly reduces leukemic engraftment in experimental mouse models. Additionally, these antibodies can neutralize IL-3-mediated rescue of TKI-induced cell death, addressing a key resistance mechanism. Research demonstrates that both healthy donor allogeneic natural killer (NK) cells and CML patients' autologous NK cells can mount effective CSL362-mediated ADCC responses against leukemic cells. This dual action—direct cytotoxicity and prevention of cytokine-mediated resistance—makes CD123-targeting antibodies particularly valuable for targeting the persistent leukemic stem cell population that conventional TKI therapy often fails to eliminate .

What distinguishes natural killer cell activity in CML patients compared to healthy donors?

Natural killer (NK) cells in CML patients exhibit distinct characteristics that separate them from those in healthy individuals. Single-cell analysis reveals that CML patients have a higher proportion of CD56dim NK cells with an active phenotype, characterized by elevated expression of cytotoxic molecules (GZMA/B, PRF1), chemokines (CCL3/4), and inflammatory cytokines (IFNG). These cells demonstrate significant cytotoxic potential against leukemic targets. Notably, the interaction between leukemic cells and NK cells occurs through inhibitory LGALS9-TIM3 and PVR-TIGIT pathways, with upregulation of LGALS9 observed in CML target cells. This distinct NK cell profile in CML patients represents an important immunological signature that can be potentially harnessed for therapeutic approaches. The active state of these NK cells makes them particularly effective in antibody-mediated cellular cytotoxicity when combined with targeted antibody therapies .

What structural engineering approaches are used to optimize bispecific antibody design for leukemia therapy?

Bispecific antibody engineering for leukemia therapy employs several sophisticated structural design approaches to ensure both efficacy and safety. The knob-and-hole technology represents one of the most successful methods, enabling the production of bispecific antibodies with high monomer purity (>97-99%) and minimal aggregates (<0.3-0.7%). This technology creates asymmetric modifications in the CH3 domains that favor heterodimer formation. Additionally, strategic mutations (CH3 N297G) are introduced in the Fc domain to reduce interactions with Fcγ receptors, minimizing unwanted antibody-mediated effector functions that could lead to off-target toxicity. Successful bispecific antibody design also requires careful selection of antibody arms with appropriate binding affinities to both targets. For example, in CLL-1/CD3 bispecific antibodies, researchers have investigated various CD3ε affinities ranging from low (50 nM) to very high (0.05 nM) to identify the optimal balance between potency, pharmacokinetics, and safety. This systematic engineering approach ensures that the resulting bispecific antibody maintains structural integrity while effectively redirecting T cell cytotoxicity against leukemic cells .

How can computational modeling accelerate antibody optimization for targeting leukemia antigens?

Computational modeling significantly accelerates antibody optimization through multiple advanced techniques. De novo CDR loop conformation prediction allows researchers to model critical binding regions with high accuracy, enabling refinement without extensive wet-lab iterations. Batch homology modeling facilitates rapid construction of structural models for both parent sequences and variants, dramatically reducing the time required to evaluate modifications. Modern computational platforms can identify promising antibody candidates through sophisticated prediction tools that characterize structural stability, binding potential, and pharmacokinetic properties. For anti-leukemia antibodies specifically, computational approaches enable researchers to detect potential hotspots for aggregation through protein surface analysis, identify favorable antibody-antigen contacts through fast protein-protein docking, and predict the impact of residue substitutions on binding affinity and selectivity. Additionally, technologies like Residue Scan FEP+ with lambda dynamics allow rapid identification of high-quality protein variants with improved targeting of leukemia-specific antigens. These computational tools collectively reduce development timelines while increasing the probability of identifying clinically viable antibody candidates .

What transgenic mouse models are appropriate for preclinical evaluation of antibodies targeting myeloid leukemia?

For preclinical evaluation of antibodies targeting myeloid leukemia, dual-targeting BAC-transgenic mouse models provide a sophisticated system that maintains an intact immune system while expressing human target antigens. A particularly effective model is the 2xBAC-Tg GEMM (genetically engineered mouse model) co-expressing human CD3ε and human CLL-1 antigens. In this model, human CD3ε is expressed on CD4+ and CD8+ T cells, while human CLL-1 is expressed on a subset of myeloid cells including monocytes and eosinophils (though notably not on neutrophils, which differs from human expression patterns). This model allows for dose-dependent evaluation of T-cell activation upon exposure to targeted bispecific antibodies, as confirmed by CD69 upregulation on CD3+CD8+ T cells. Importantly, hematopoietic stem cells (Sca1+/c-kit+ HSCs) in these models do not express the human CLL-1 on their surface, which permits interpretation of efficacy beyond the normal recovery period of blood cells (10-14 days). These characteristics make such models invaluable for screening the in vivo potency and behavior of antibodies targeting myeloid malignancies before advancing to clinical trials .

What quality control processes should be implemented when validating antibodies for research applications?

Implementing rigorous quality control processes for antibody validation requires a multi-step approach to ensure reliability. Initially, all antibody candidates should undergo strict specificity and sensitivity assessment using immunohistochemistry (IHC) prior to any conjugation. Following conjugation (especially for applications like imaging mass cytometry), antibodies must be re-tested to confirm they retain their affinity and specificity, as the conjugation process can significantly alter antibody function. This was explicitly demonstrated with anti-PDGFRa antibody (clone D13C6), which lost signal in IHC after conjugation. For critical antibodies that demonstrate sensitivity to conjugation but remain essential for research panels, alternate approaches should be implemented - such as including appropriate secondary antibodies in the panel. Statistical validation should employ methods like Spearman correlation tests (with coefficient values >±0.5 and p<0.05 considered significant) and Bland-Altman plots to ensure consistent performance. For antibodies intended for complex analyses, additional validation should include assessment of inter- and intra-case heterogeneity using χ2 tests and evaluation of various cell class distributions using appropriate computational tools .

How can researchers evaluate the effectiveness of antibody-mediated cytotoxicity against leukemic stem cells?

Evaluating antibody-mediated cytotoxicity against leukemic stem cells requires a multi-faceted approach. Researchers should first establish baseline expression of target antigens (such as CD123) on CD34+/CD38- cells from both patients and normal donors using flow cytometry to quantify differential expression patterns. To assess antibody-dependent cell-mediated cytotoxicity (ADCC), co-culture assays should be performed using both healthy donor allogeneic natural killer (NK) cells and, crucially, patients' autologous NK cells to determine whether the patient's own immune system can mount an effective antibody-mediated response. Measuring reduction in leukemic engraftment in appropriate mouse models provides a critical in vivo readout of effectiveness. Additionally, researchers should conduct combination studies with standard-of-care treatments (such as tyrosine kinase inhibitors for CML) to determine whether the antibody enhances elimination of leukemic progenitors and stem cells beyond conventional therapy alone. For comprehensive evaluation, it's important to analyze both the direct cytotoxic effects and the ability to neutralize cytokine-mediated rescue mechanisms (such as IL-3 signaling) that contribute to leukemic stem cell persistence .

How should researchers interpret differences in antibody efficacy between in vitro and in vivo models?

Researchers should approach discrepancies between in vitro and in vivo antibody efficacy data with careful consideration of multiple factors. When in vitro studies show dramatic potency differences (e.g., 100-fold greater activity with high-affinity CD3 arms) that become less pronounced in animal models, this often reflects the complex pharmacokinetic landscape in living systems. The prolonged exposure to therapeutic antibodies in vivo can compensate for lower binding affinity, as demonstrated with lower-affinity CD3 TDBs that show comparable efficacy to high-affinity variants in mouse models despite their reduced in vitro activity. Additionally, safety considerations often emerge only in animal models - as seen when only low-affinity CD3/CLL1 TDBs proved well-tolerated in non-human primates while still achieving target cell depletion. When interpreting such data, researchers should specifically evaluate: 1) residence time and biodistribution of different antibody variants, 2) engagement of complex immune components absent in simplified in vitro systems, 3) potential compensatory mechanisms in living organisms, and 4) emergence of adverse effects that may limit dosing of otherwise potent antibody candidates. This multi-factorial analysis prevents premature elimination of promising candidates based solely on in vitro potency metrics .

What strategies can address reduced antibody function following conjugation for imaging or detection applications?

When antibodies show reduced function following conjugation for imaging or detection applications, researchers should implement a systematic troubleshooting strategy. First, perform comparative pre- and post-conjugation testing using identical conditions to precisely quantify the functional changes. For antibodies that demonstrate significant sensitivity to conjugation yet remain essential for research panels (as observed with anti-PDGFRa antibody clone D13C6), consider alternative approaches such as incorporating appropriate secondary antibodies in the panel to detect the primary unconjugated antibody. Adjusting the conjugation chemistry may also restore function - try varying metal tags, fluorophores, or conjugation protocols with smaller batches before scaling up. Additionally, epitope accessibility can be improved through modified sample preparation protocols, including optimized antigen retrieval methods for tissue samples or adjusted fixation procedures that better preserve the target epitope. If direct conjugation consistently compromises antibody function, consider indirect detection systems or alternative antibody clones targeting different epitopes of the same protein. Document all optimization steps thoroughly to establish standardized protocols that ensure reproducibility across experiments and research groups .

How can researchers determine the optimal antibody affinity balance for therapeutic applications?

Determining the optimal antibody affinity balance for therapeutic applications requires systematic evaluation across multiple dimensions. Researchers should first establish an affinity testing matrix that includes variants spanning several orders of magnitude in binding strength (e.g., KD values from 50 nM to 0.05 nM for CD3ε binding). For each variant, conduct parallel assessments of: 1) in vitro potency using relevant cell-based assays measuring target cell killing, T-cell activation markers, and cytokine release; 2) pharmacokinetic profiling to determine plasma half-life and tissue distribution; and 3) comprehensive safety evaluation in appropriate animal models. The critical insight from such studies is that the highest affinity antibody is frequently not the optimal therapeutic candidate - as demonstrated when lower-affinity CD3/CLL1 TDBs showed superior tolerability while maintaining efficacy in non-human primates compared to their high-affinity counterparts. Researchers should specifically look for the inflection point where increasing affinity no longer improves therapeutic index but begins to compromise safety. For T cell-engaging bispecifics particularly, moderate-to-low CD3 affinity often provides the best balance between maintaining sufficient target cell depletion while preventing excessive T cell activation that could lead to cytokine release syndrome and other adverse events .

How do combination approaches with tyrosine kinase inhibitors and antibody therapies enhance elimination of resistant leukemic cells?

Combination approaches with tyrosine kinase inhibitors (TKIs) and antibody therapies create a synergistic effect through complementary mechanisms that target different aspects of leukemic cell survival. TKIs effectively eliminate the majority of differentiated leukemic cells by inhibiting constitutively active signaling pathways, but often fail to eradicate leukemic stem and progenitor cells (LSPCs) that persist through cytokine-mediated resistance mechanisms. Antibody therapies, particularly those targeting CD123 (IL-3 receptor α subunit), directly address this resistance pathway through dual mechanisms: first, by facilitating antibody-dependent cell-mediated cytotoxicity (ADCC) against CD123+ cells, and second, by neutralizing IL-3-mediated rescue of TKI-induced cell death. Research demonstrates that this combination causes significantly greater reduction of CML progenitors than either approach alone. Furthermore, the combination preferentially eliminates leukemic stem cells over normal hematopoietic stem and progenitor cells, improving the therapeutic index. This selective targeting occurs because CD123 expression is higher in leukemic stem cells than in normal stem cells, providing a therapeutic window that allows for the elimination of drug-resistant leukemic cells while preserving normal hematopoiesis .

What role do imaging mass cytometry panels play in understanding the tumor microenvironment in leukemia research?

Imaging mass cytometry (IMC) panels provide unprecedented insights into the spatial relationships between leukemic cells and their microenvironment through highly multiplexed analysis of tissue sections. Advanced IMC panels can simultaneously detect up to 42 different markers, enabling comprehensive characterization of multiple cell types and their functional states within the tumor microenvironment. This approach is particularly valuable for understanding complex cellular interactions that may influence leukemia progression and treatment response. When developing such panels, researchers must implement rigorous antibody validation protocols, including assessment of antibody specificity and sensitivity before and after metal conjugation, as the conjugation process can significantly alter antibody function. Statistical analysis of IMC data typically employs methods such as Spearman correlation tests to determine relationships between markers and computational approaches to examine inter- and intra-case heterogeneity. For leukemia research specifically, IMC panels can identify spatial organization of leukemic cells relative to immune effector cells, stromal components, and vascular structures, providing crucial information about potential escape mechanisms and resistance pathways that might not be apparent from conventional bulk analysis methods .

How can bispecific antibody engineering be optimized to target leukemic stem cells while sparing normal hematopoietic stem cells?

Optimizing bispecific antibody engineering to selectively target leukemic stem cells while sparing normal hematopoietic stem cells requires sophisticated design strategies focused on differential antigen expression and binding kinetics. An effective approach leverages antigens that are overexpressed on leukemic stem cells but have limited expression on normal hematopoietic stem cells - such as CLL-1, which is prevalent in acute myeloid leukemia but not expressed on hematopoietic stem cells, potentially allowing for normal hematopoietic recovery. The engineering process should include: 1) careful selection of target antigens with preferential expression on leukemic stem cells; 2) fine-tuning of binding affinity to both the target antigen and the effector arm (e.g., CD3ε) to maximize the therapeutic window; 3) optimization of antibody format using technologies like knob-and-hole to produce stable, high-quality bispecific molecules; and 4) incorporation of Fc domain mutations to minimize unwanted effector functions. Preclinical validation requires assessment in appropriate animal models to confirm both efficacy against leukemic cells and sparing of normal hematopoiesis. A particularly promising approach involves bispecific T cell-dependent antibodies that redirect T cell cytotoxicity specifically against leukemic stem cells through dual targeting, as demonstrated with CD3/CLL-1 bispecific antibodies that show potential for treating acute myeloid leukemia without compromising normal hematopoietic recovery .

What cell line models are most appropriate for evaluating antibody efficacy against myeloid leukemias?

When evaluating antibody efficacy against myeloid leukemias, researchers should select cell line models that best recapitulate the molecular and phenotypic characteristics relevant to the antibody's mechanism of action. For studies targeting CLL-1 or CD123, human AML cell lines such as Molm-13, ML-2, THP-1, EOL-1, Nomo-1, U937, HL-60, and PL-21 provide a diverse testing panel representing different disease subtypes. These cell lines should be maintained under standardized conditions (RPMI 1640 medium with 10% heat-inactivated fetal bovine serum, 2 mM glutamine, and 1% penicillin-streptomycin at 37°C in 5% CO2) to ensure reproducibility. Critical consideration should be given to the expression levels of target antigens on these cell lines, which should be quantified before experimentation using flow cytometry or other appropriate methods. For bispecific antibody testing, co-culture systems incorporating both the leukemic cell lines and appropriate effector cells (T cells or NK cells) are essential to evaluate antibody-dependent cytotoxicity. Additionally, researchers should consider using cell lines with defined genetic alterations that reflect clinically relevant disease subtypes, as response to antibody therapy may vary based on underlying molecular pathology .

How should antibody structure prediction be approached for novel therapeutic candidates?

Antibody structure prediction for novel therapeutic candidates requires a multifaceted computational approach that integrates homology modeling with advanced simulation techniques. Begin with a fully guided homology modeling workflow that incorporates de novo CDR loop conformation prediction, as these hypervariable regions are critical for antigen binding but often challenge traditional modeling methods. When pursuing rational antibody humanization, generate humanized antibodies through CDR grafting combined with targeted residue mutations, followed by careful evaluation of the percentage of humanness in the resulting constructs to minimize immunogenicity. For antibody-antigen interaction analysis, employ ensemble protein-protein docking to predict complex structures and identify favorable contact points. The resolution of experimental epitope mapping data can be enhanced from peptide to residue-level detail, providing crucial insights for optimization. To identify potential development risks early, computational protein surface analysis should be used to detect potential hotspots for aggregation and highlight potential surface sites for post-translational modification and chemical reactivity. Finally, for in silico engineering, accurately predict the impact of residue substitutions on binding affinity, selectivity, and thermostability using advanced techniques like Residue Scan FEP+ with lambda dynamics, which allows rapid identification of high-quality protein variants .

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.