CD45 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Significance

CD45 (leukocyte common antigen) is a transmembrane glycoprotein expressed on all nucleated hematopoietic cells. Recombinant monoclonal antibodies against CD45 are engineered to recognize specific epitopes, enabling precise identification of leukocytes in research and clinical diagnostics . Their recombinant format ensures batch-to-batch consistency, overcoming limitations of traditional hybridoma-derived mAbs .

Key Steps in Antibody Generation

  • Antigen Preparation: CD45 extracellular domains are expressed in eukaryotic systems (e.g., HEK293T cells) to preserve post-translational glycosylation, critical for antigen-antibody interaction .

  • Immunization: Mice are immunized with glycosylated recombinant CD45-his protein, yielding antisera with titers >1:32,000 .

  • Hybridoma Screening: Hybridomas are selected via ELISA and flow cytometry, followed by sub-cloning to isolate high-affinity clones .

Applications in Research and Diagnostics

ApplicationProtocol DetailsReference
Flow Cytometry1×10⁶ cells stained with 1:100–1:400 antibody dilution; detects leukocytes
Immunohistochemistry1:250 dilution on FFPE tissues; validated in human tonsil and colon carcinoma
Western Blot1:1000 dilution; detects endogenous CD45 (180–240 kDa) in Jurkat, MOLT-4 cells
Immunofluorescence1:100–1:400 dilution; localizes CD45 on cell surfaces in PBMCs

Specificity Testing

  • Flow Cytometry: Anti-CD45 mAbs showed 98% binding to CD45⁺ HL60 cells vs. <2% to CD45⁻ SKBR-3 cells .

  • Cross-Reactivity: Rabbit mAbs (e.g., ab40763) exhibit no off-target binding in multi-tissue microarray assays .

Comparative Affinity

Antibody CloneHostAffinity (ELISA Titer)Specificity Confirmed
EP322Y (ab40763)Rabbit1:10,000Yes (55+ publications)
BL-178-12C7 (NBP3-14668)Rabbit1:1000 (WB)Yes (IHC, IF)
4D3 (PMC Study)Mouse1:32,000Yes (CTC isolation)

Challenges and Solutions

  • Glycosylation Complexity: Native CD45 has extensive N-/O-linked glycans, necessitating eukaryotic-expressed antigens for immunization .

  • Commercial Limitations: Traditional mAbs often show batch variability; recombinant formats (e.g., Cell Signaling #13917) address this via standardized production .

Recent Advances and Future Directions

  • CTC Detection: Anti-CD45 mAbs reliably differentiate leukocytes from circulating tumor cells in breast cancer patients, enhancing diagnostic accuracy .

  • Therapeutic Potential: Targeting CD45’s intracellular domain (e.g., #13917) may modulate T-cell activation pathways .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

CD45 antibody CSB-RA019049A0HU is a recombinant monoclonal antibody produced through the expression of plasmids constructed from the CD45 monoclonal antibody DNA sequence (generated from animals immunized with a human CD45 synthesized peptide). The antibody was purified using an affinity chromatography method. This is a rabbit IgG antibody that reacts with the CD45 protein from human samples and is suitable for ELISA analysis.

CD45, a receptor-like tyrosine phosphatase, is exclusively found in the hematopoietic system. It plays a crucial role in the growth and activation of T lymphocytes. Due to its leukocyte-specific tissue distribution, CD45 is a valuable marker for the differential diagnosis of undifferentiated lymphoma.

Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. For specific delivery time information, please consult your local distributors.
Synonyms
B220 antibody; CD 45 antibody; CD45 antibody; CD45 antigen antibody; CD45R antibody; GP180 antibody; L-CA antibody; LCA antibody; Leukocyte common antigen antibody; loc antibody; Ly-5 antibody; LY5 antibody; Ly5, homolog of antibody; Lyt-4 antibody; OTTHUMP00000033813 antibody; OTTHUMP00000033816 antibody; OTTHUMP00000033817 antibody; OTTHUMP00000038574 antibody; Protein tyrosine phosphatase receptor type c polypeptide antibody; Protein tyrosine phosphatase, receptor type C antibody; protein tyrosine phosphatase, receptor type, C antibody; Protein tyrosine phosphatase, receptor type, c polypeptide antibody; Ptprc antibody; PTPRC_HUMAN antibody; Receptor-type tyrosine-protein phosphatase C antibody; T200 antibody; T200 glycoprotein antibody; T200 leukocyte common antigen antibody
Target Names
Uniprot No.

Target Background

Function
Protein tyrosine-protein phosphatase required for T-cell activation through the antigen receptor. Functions as a positive regulator of T-cell coactivation upon binding to DPP4. The first PTPase domain exhibits enzymatic activity, while the second one influences the substrate specificity of the first. Upon T-cell activation, it recruits and dephosphorylates SKAP1 and FYN. It dephosphorylates LYN, thereby modulating LYN activity. In the context of microbial infection, it acts as a receptor for human cytomegalovirus protein UL11 and mediates UL11 binding to T-cells. This binding leads to reduced induction of tyrosine phosphorylation of multiple signaling proteins upon T-cell receptor stimulation and impaired T-cell proliferation.
Gene References Into Functions
  1. This research reveals the heterogeneity of CD4 effector memory T cells expressing CD45RA and provides insights into T-cell responses against dengue virus and other viral pathogens. PMID: 29133794
  2. Five-year disease-free survival for patients with high transcriptional expression of CD45 (n = 107) was 62.4%, while for patients with low expression (n = 53) it was 36.2% (P = 0.003). Patients exhibiting high expression of CD45 demonstrated better local recurrence-free survival and disease-specific survival. PMID: 29177949
  3. CD45 acts as a regulator of IL-2 synergy in the NKG2D-mediated activation of immature human NK cells. PMID: 28655861
  4. TCR phosphorylation exhibits a negative correlation with TCR-CD45 separation. PMID: 29467364
  5. Our findings demonstrate that the expression of CD163 and CD206 in monocytes is modulated by LPS in vitro. LPS induces CD163 expression and downregulates the spontaneously increased expression of CD206. PMID: 25914252
  6. The no-wash, no-lyse staining protocol with CD45-KO and CD14-PB allows for clear differentiation and gating of the monocyte population under near-physiological conditions. PMID: 25758468
  7. The regulatory effect of the mannose receptor (MR) is mediated by a direct interaction with CD45 on the T cell, inhibiting its phosphatase activity. This results in up-regulation of CTLA-4 and the induction of T-cell tolerance. Inhibition of CD45 prevents expression of B-cell lymphoma 6 (Bcl-6), a transcriptional inhibitor that directly binds the CTLA-4 promoter and regulates its activity. PMID: 27601670
  8. pUL11 induces IL-10 producing T cells through its binding to the CD45 phosphatase on T cells. PMID: 28628650
  9. Expression of IL10R subunits within the leukocyte population (CD45(+) cells) was significantly higher in primary brain tumors compared to metastases. PMID: 28982901
  10. CD45 expression is routinely measured in the diagnostics of acute leukemias in relation to SSc. PMID: 26415521
  11. A phosphosite within the SH2 Domain of Lck regulates its activation by CD45. This leads to a negative feedback loop that responds to signaling events, fine-tuning active Lck amounts and TCR sensitivity. PMID: 28735895
  12. The C77G variant is not associated with ovarian cancer in the Norwegian population. However, it may be linked to a less aggressive cancer type. PMID: 28759630
  13. Our findings suggest that CD45 is a key regulator of BCR-signaling thresholds mediated by T-cell help. PMID: 27056269
  14. We demonstrate for the first time the physiological existence of ct-CD45 in human plasma and show that it may be an extrinsic factor contributing to the maintenance of human T-cell quiescence. PMID: 27718235
  15. Our findings indicate that if the w/h ratio on SSC versus CD45 plot is less than 1.6, AML may be considered. Conversely, if it is more than 1.6, ALL may be diagnosed. Using morphometric analysis of the blast cluster on SSC versus CD45, it was possible to distinguish between ALL and AML, and their subtypes. PMID: 27748273
  16. The use of the common leukocyte marker CD45 enhances the sensitivity of the diagnosis of lymphocytic myocarditis. PMID: 28025077
  17. Patients with the PTPRC rs10919563 A allele exhibit a poor response to anti-TNF therapy in rheumatoid arthritis. PMID: 27074847
  18. CD41 and CD45 expression marks the onset of haemangioblastoma (HB) neovascularization and the stepwise development of the angioformative period. These markers also represent potential therapeutic targets for anti-vascular treatment. PMID: 26468019
  19. PTPRC has become the most replicated genetic biomarker of response to TNF inhibitors. PMID: 25896535
  20. CD45RO in tumor-infiltrating lymphocytes was found to be a positive prognostic factor in squamous non-small cell lung cancer. PMID: 26678911
  21. CD45RO+ memory T-cells produce IL-17 in patients with atherosclerosis. PMID: 26667768
  22. Data highlight the structural basis for, and potent signaling effects of, local CD45 antigen and kinase segregation. PMID: 26998761
  23. A CD45+/CD19 - cell population in bone marrow aspirates correlated with the clinical outcome of patients with mantle cell lymphoma. PMID: 25739938
  24. High CD45 expression is associated with multiple myeloma. PMID: 26994849
  25. C77G T(reg) exhibited diminished upregulation of activation markers, lower phosphorylation of p56(lck)(Y505), and a reduced proliferative potential when stimulated with anti-TcR or anti-TcR plus CD28 mAb, suggesting decreased responsiveness to activating stimuli. These findings indicate that alterations in CD45 isoform combination resulting from the C77G mutation influence responsiveness. PMID: 26355564
  26. Findings demonstrate that CD45 antigen(+) and c-Kit protein(+) hematopoietic cells were more abundant in muscle than in bone marrow between embryonic 14.5 and 17.5 days. PMID: 26389592
  27. The ratio of CD4+CD45RO+CD25-/lowCD127+: CD4+CD45RO+CD25hiCD127-/low in peripheral blood identifies heart transplant recipients at risk for cardiac allograft vasculopathy. PMID: 25539460
  28. CD45+ cells were abundant in the stroma of physiologically immature placental villi and decreased as pregnancy progressed. PMID: 25043745
  29. This study demonstrated a correlation between copy number variations of PTPRC and opioid dependence. PMID: 25345593
  30. This study did not replicate the association between PTPRC and the response to anti-TNF treatment in a Southern European population. However, it found that TRAF1/C5 risk RA variants potentially influence anti-TNF treatment response. PMID: 25834819
  31. Long noncoding RNA encoded by the natural antisense gene of CD45 contributes to the expressional regulation of the CD45RO splicing variant via recruitment of DNA methyltransferase and histone modification modulators specific to the sense gene CD45. PMID: 25381328
  32. In T-cells, cholesterol-dependent domains function in the regulation of the Src family kinase Lck (p56lck) by sequestering Lck from its activator CD45. (Review) PMID: 25658353
  33. Patients with the presence of CD8- and CD45RO-positive T cells in bone marrow exhibited better survival of gastric cancer patients compared to those without these cells in bone marrow. PMID: 25804232
  34. Low expression of CD39(+) /CD45RA(+) on regulatory T cells (Treg ) cells in type 1 diabetic children in contrast to high expression of CD101(+) /CD129(+) on Treg cells in children with coeliac disease. PMID: 25421756
  35. Late-outgrowth CD45 negative endothelial progenitor cells express markers associated with pluripotency and can directly express an osteogenic phenotype under bone differentiation conditions. PMID: 25531767
  36. SLAMF7-triggered inhibition is mediated by a mechanism involving Src kinases, CD45, and SHIP-1, which is defective in MM cells. PMID: 25312647
  37. Phosphatase CD45 both positively and negatively regulates T cell receptor phosphorylation in reconstituted membrane protein clusters, depending on LCK activity. PMID: 25128530
  38. Results show that CD45 PTP activity is inhibited by galectin-1 in the anaplastic large cell lymphoma cell line H-ALCL. PMID: 24589677
  39. Expressing CD45 promoters containing these regions and tethered to green fluorescent protein (GFP) in a primary B-cell differentiation assay and a transplantation model resulted in high levels of GFP in lymphoid, myeloid, and nucleated erythroid cells. PMID: 24852660
  40. The rare sub-population of CD45(-)/Lin(-)/SSEA-4(+) VSEL stem cells survived after Hespan sedimentation. PMID: 24364909
  41. Hematopoietic cell marker CD45 is expressed in hepatic progenitor cells. PMID: 24396288
  42. CD45RA-Foxp3high Tregs increase in the peripheral circulation of head and neck squamous cell carcinoma patients. PMID: 24761979
  43. The regulation of alternative splicing in CD45 by IkappaBL was independent from the kinase activity of CLK1. PMID: 23953137
  44. High CD45 surface expression is associated with a poor prognosis in both BCP-ALL and T-ALL. PMID: 23911702
  45. PTPRC/CD45 is down-regulated in leukemogenic tyrosine kinase expressing cells. PMID: 23997015
  46. [review] Circulating CD34+/KDR+/CD45dim endothelial progenitor cells hold great potential as biomarkers of vasculogenesis and endothelial repair when research protocols of in vitro culture and flow cytometry are included. PMID: 23171577
  47. Heterogeneity within the Lin(-)CD45(-) cell fraction is the likely explanation for differences in the hUCB cell populations. PMID: 23840798
  48. Galectin-3-induced apoptosis of Jurkat cells is regulated by both O-glycans and N-glycans on CD45. PMID: 24211831
  49. Spatial regulation of Lck by CD45 and GM1 ganglioside determines the outcome of the apoptotic response to Gal-1. This local regulation likely occurs only upon intimate effector (Gal-1 expressing) cell-T-cell attachment. PMID: 24231767
  50. A glycosylation-dependent CD45RB epitope defines previously unacknowledged CD27-(IgM high) B cell subpopulations enriched in young children and after hematopoietic stem cell transplantation. PMID: 24211716

Show More

Hide All

Database Links

HGNC: 9666

OMIM: 126200

KEGG: hsa:5788

STRING: 9606.ENSP00000356346

UniGene: Hs.654514

Involvement In Disease
Severe combined immunodeficiency autosomal recessive T-cell-negative/B-cell-positive/NK-cell-positive (T(-)B(+)NK(+) SCID); Multiple sclerosis (MS)
Protein Families
Protein-tyrosine phosphatase family, Receptor class 1/6 subfamily
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Membrane raft.
Tissue Specificity
Isoform 1: Detected in thymocytes. Isoform 2: Detected in thymocytes. Isoform 3: Detected in thymocytes. Isoform 4: Not detected in thymocytes. Isoform 5: Detected in thymocytes. Isoform 6: Not detected in thymocytes. Isoform 7: Detected in thymocytes. Is

Q&A

What is CD45 and why is it an important target for monoclonal antibodies?

CD45 is a heavily glycosylated protein that functions as a common leukocyte antigen. It is expressed on at least 90% of myeloid leukemias and is not found in tissues of non-hematopoietic origin, making it an attractive target for both diagnostic and therapeutic applications in hematological research . CD45 is commonly used as a marker to differentiate leukocytes from circulating tumor cells (CTCs) in cancer research, particularly in studies involving liquid biopsies and rare cell detection . The extensive post-translational modifications of CD45, including heavy N- and O-linked glycosylation, make it a challenging target for antibody generation, which has driven the development of specialized recombinant approaches .

How do recombinant CD45 monoclonal antibodies differ from conventional antibodies?

Recombinant CD45 monoclonal antibodies are generated using molecular biology techniques that allow for precise control over antibody structure and properties. Unlike conventional antibodies produced through traditional hybridoma technology alone, recombinant antibodies can be engineered for specific characteristics such as improved affinity, reduced immunogenicity, or enhanced stability. When targeting heavily glycosylated proteins like CD45, recombinant approaches can offer superior recognition of native epitopes compared to antibodies generated against prokaryotic expression systems or peptide fragments . This is because the recombinant proteins can be expressed in eukaryotic systems that maintain appropriate post-translational modifications, particularly the extensive glycosylation that is critical for CD45 structure and function .

What are the primary research applications for CD45 monoclonal antibodies?

CD45 monoclonal antibodies serve multiple research purposes:

  • Immunophenotyping: Identification and characterization of leukocyte populations in flow cytometry and immunohistochemistry

  • CTC Detection: Differentiation between white blood cells and circulating tumor cells in cancer research and diagnostics

  • Radioimmunotherapy (RIT): Targeted delivery of radionuclides to CD45-expressing cells for treatment of hematological malignancies

  • Cell Sorting: Isolation of specific leukocyte populations for downstream applications

  • Functional Studies: Investigation of CD45's role in immune cell signaling and activation

These applications leverage the specificity of anti-CD45 antibodies to enable precise identification or targeting of leukocytes in complex biological samples .

What validation methods should be employed to confirm CD45 antibody specificity?

Rigorous validation of CD45 antibody specificity is essential for reliable research outcomes. Recommended validation approaches include:

  • Flow Cytometry: Testing against CD45-positive cell lines (e.g., THP-1) in comparison with isotype controls

  • Knockout Controls: Verification of antibody specificity using CD45 knockout cell lines, which should show no staining with a specific anti-CD45 antibody

  • Cross-Reactivity Testing: Evaluation against irrelevant his-tagged proteins or non-CD45 expressing cells to exclude non-specific binding

  • Multiple Detection Methods: Confirmation of specificity using orthogonal techniques such as Western blotting, immunohistochemistry, and ELISA

  • Epitope Mapping: Determination of the specific recognition site on CD45 to ensure the antibody targets the intended region

These validation steps are particularly important for CD45 due to its complex glycosylation pattern, which can affect antibody recognition .

How can researchers overcome challenges in generating monoclonal antibodies against heavily glycosylated regions of CD45?

Generating high-affinity monoclonal antibodies against heavily glycosylated proteins like CD45 presents significant challenges due to the complex post-translational modifications that affect epitope accessibility and recognition. To overcome these challenges, researchers should consider:

  • Eukaryotic Expression Systems: Use of HEK293T or similar eukaryotic cell lines for expressing recombinant CD45 with native-like glycosylation patterns. This approach has been shown to generate antibodies with superior affinity and specificity compared to those raised against prokaryotic proteins or synthetic peptides .

  • Extracellular Domain Focus: Cloning and expressing the extracellular portion of CD45 with appropriate glycosylation sites intact. In published protocols, this has been achieved by amplifying CD45 cDNA from cells naturally expressing CD45 (such as HL60 cells) and inserting the extracellular domain sequence into eukaryotic expression vectors .

  • Purification Strategy: Implementation of affinity chromatography using his-tag purification methods to obtain highly purified glycosylated CD45 protein for immunization. The addition of a his-tag enables efficient protein purification while minimally affecting the native structure .

  • Immunization Protocol: Multiple immunizations (typically three at two-week intervals) with properly folded glycosylated CD45 protein in combination with appropriate adjuvants to elicit robust immune responses .

  • Hybridoma Screening: Sequential screening using both ELISA (against recombinant protein) and flow cytometry (against native CD45-expressing cells) to identify clones that recognize the naturally glycosylated protein on cell surfaces .

This integrated approach has been demonstrated to generate antibodies with robust affinity and specificity with as few as one cell fusion and two cyclic sub-cloning steps .

What are the considerations for developing CD45-targeted radioimmunotherapy approaches?

Radioimmunotherapy (RIT) using CD45-targeted antibodies represents a promising approach for treating hematological malignancies, but requires careful optimization. Key considerations include:

  • Conventional vs. Pretargeted Approaches: Pretargeted radioimmunotherapy (PRIT) using anti-CD45 antibody-streptavidin conjugates followed by biotinylated clearing agents and radiolabeled-DOTA-biotin has demonstrated superior tumor-to-blood ratios (20:1) compared to conventional RIT (<1:1) at 24 hours .

  • Radionuclide Selection: Choice of appropriate radionuclides (e.g., Yttrium-90, Iodine-131) based on half-life, energy emission, and tissue penetration characteristics to match the specific disease distribution pattern .

  • Dosimetry Calculations: Careful determination of radiation absorbed dose to target tissues versus normal organs to maximize therapeutic efficacy while minimizing toxicity. PRIT approaches have shown the ability to deliver at least twice as much radiation to bone marrow and five times more to spleen compared to conventional RIT .

  • Antibody Engineering: Modification of antibody structure to optimize pharmacokinetics, biodistribution, and radionuclide conjugation without compromising CD45 binding affinity.

  • Combinatorial Approaches: Integration with conditioning regimens for hematopoietic cell transplantation to enhance disease eradication while enabling hematopoietic recovery .

The development of these approaches requires balancing the delivery of myeloablative doses to disease sites while managing radiation exposure to normal tissues, particularly in cases where CD45 is expressed on both malignant and normal hematopoietic cells .

How do glycosylation patterns of CD45 vary across different cell types and disease states, and how does this impact antibody recognition?

CD45 exhibits remarkable heterogeneity in glycosylation patterns that varies by cell lineage, differentiation stage, and disease state. These variations have significant implications for antibody recognition:

  • Lineage-Specific Isoforms: Different hematopoietic cell types express distinct CD45 isoforms (CD45RA, CD45RB, CD45RC, etc.) due to alternative splicing of exons 4, 5, and 6, which affects the protein's extracellular domain structure and glycosylation pattern .

  • Aberrant Glycosylation in Malignancy: Leukemic cells often display altered glycosylation profiles compared to their normal counterparts, potentially creating unique epitopes or masking conserved ones .

  • Antibody Epitope Accessibility: Changes in glycosylation can dramatically affect the accessibility of protein epitopes, potentially reducing binding affinity of antibodies that target regions adjacent to or containing glycosylation sites .

  • Cross-Reactivity Concerns: Antibodies generated against recombinant CD45 may exhibit differential binding to native CD45 from various cell types or disease states due to glycosylation differences .

  • Validation Requirements: These variations necessitate comprehensive validation of antibody performance across multiple cell types and conditions to ensure consistent recognition .

To address these challenges, researchers should consider using eukaryotic expression systems that most closely mimic the glycosylation patterns of target cells and validate antibody performance against primary cells from relevant disease states rather than relying solely on cell line data .

What methodological approaches can enhance the specificity of CD45 antibodies for circulating tumor cell (CTC) detection?

Enhancing specificity for CTC detection requires strategies that maximize the distinction between CD45-positive leukocytes and CD45-negative tumor cells:

  • Antibody Generation Strategy: Using eukaryotic-expressed recombinant CD45 as immunogen produces antibodies that recognize native CD45 glycoforms present on patient leukocytes with higher specificity than antibodies generated against prokaryotic proteins or peptides .

  • Multi-Parameter Approach: Combining CD45 negativity with positive markers such as EpCAM and cytokeratins (e.g., CK19) in a multi-color immunofluorescence assay improves discrimination between CTCs and leukocytes .

  • Sample Processing Optimization: Implementing negative selection with anti-CD45 after initial enrichment with tumor-specific markers (e.g., EpCAM) can reduce false positives. This sequential approach first enriches for EpCAM-positive cells using magnetic separation, followed by CD45 staining to exclude remaining leukocytes .

  • Validation Against Patient Samples: Testing antibody performance on blood samples from patients with known disease rather than relying solely on cell lines ensures applicability to clinical specimens where glycosylation patterns may differ .

  • Image-Based Cytometry: Using high-resolution microscopy combined with automated image analysis algorithms that assess multiple parameters (morphology, marker intensity, nuclear features) can further enhance discrimination .

These approaches have been shown to improve the reliability of CTC enumeration results, which is crucial for clinical applications in cancer diagnosis and monitoring .

What are the optimal protocols for generating CD45 recombinant monoclonal antibodies?

The generation of high-quality CD45 recombinant monoclonal antibodies involves several critical steps:

  • Antigen Preparation:

    • Clone CD45 cDNA from CD45-expressing cells (e.g., HL60)

    • Insert the extracellular domain sequence into a eukaryotic expression vector (e.g., pCDNA3.1)

    • Transfect HEK293T cells to express the recombinant CD45 protein with native-like glycosylation

    • Purify using nickel-affinity chromatography for his-tagged proteins

  • Immunization Strategy:

    • Primary immunization: 50 μg glycosylated rhCD45-his protein with complete Freund's adjuvant

    • Subsequent immunizations: Same protein amount with incomplete Freund's adjuvant

    • Three immunizations at two-week intervals

    • Monitor antiserum titer via ELISA (target titer >1:32,000)

  • Hybridoma Generation and Screening:

    • Fusion of splenocytes from high-titer mice with Sp2/0-Ag14 myeloma cells

    • Culture in HAT medium to select for hybridomas

    • Initial screening via ELISA against rhCD45-his protein

    • Secondary screening against irrelevant his-tagged proteins to exclude his-specific antibodies

    • Tertiary screening via flow cytometry using CD45+ cells (e.g., HL60) to confirm recognition of native CD45

  • Clone Selection and Antibody Production:

    • Two cycles of sub-cloning to establish stable hybridoma cell lines

    • Scale-up production using hollow fiber bioreactor systems

    • Purification via protein A/G affinity chromatography

  • Validation:

    • Flow cytometry against CD45+ and CD45- cells

    • Comparison with commercial antibodies for specificity and sensitivity

    • Testing against patient samples to confirm clinical utility

This methodical approach has been demonstrated to yield antibodies with superior performance characteristics compared to commercially available alternatives, particularly for applications requiring recognition of heavily glycosylated native CD45 .

What characterization methods should be employed to evaluate CD45 antibody performance for specific applications?

Comprehensive characterization of CD45 antibodies is essential to ensure their suitability for specific applications:

  • Binding Affinity Assessment:

    • Surface Plasmon Resonance (SPR) to determine kon, koff, and KD values

    • Flow cytometry titration to establish EC50 on relevant cell types

    • Comparative analysis with established commercial antibodies

  • Epitope Mapping:

    • Peptide array analysis to identify linear epitopes

    • Competition assays with known epitope-specific antibodies

    • Cross-reactivity testing against CD45 isoforms to determine isoform specificity

  • Glycoform Recognition:

    • Testing against enzymatically deglycosylated CD45 to determine dependence on glycosylation

    • Evaluation across multiple cell types with different natural CD45 glycosylation patterns

    • Performance assessment in patient samples where glycosylation may be altered by disease

  • Functional Characterization:

    • Evaluation of interference with CD45 phosphatase activity

    • Assessment of impact on cellular signaling pathways

    • Determination of internalization rates if relevant for therapeutic applications

  • Application-Specific Validation:

    • For Flow Cytometry: Resolution of positive/negative populations, signal-to-noise ratio, stability of fluorochrome conjugates

    • For Immunohistochemistry: Optimization of antigen retrieval, background staining, tissue penetration

    • For Therapeutic Applications: In vivo biodistribution, pharmacokinetics, immunogenicity

    • For CTC Detection: Sensitivity and specificity in spiked samples, comparison with established methods

Each characterization method should be selected based on the intended application, with more extensive characterization required for therapeutic or diagnostic applications compared to basic research use .

How should researchers design experimental controls when using CD45 antibodies in multi-parameter flow cytometry?

Proper experimental design with appropriate controls is critical for accurate interpretation of CD45 antibody performance in multi-parameter flow cytometry:

  • Isotype Controls:

    • Include matched isotype control antibodies (same species, isotype, and fluorochrome) to assess non-specific binding

    • Use the same concentration as the CD45 antibody to enable direct comparison

  • Fluorescence Minus One (FMO) Controls:

    • Include samples stained with all fluorochromes except anti-CD45 to establish accurate gating boundaries

    • Particularly important in multi-parameter panels where spectral overlap can complicate interpretation

  • Positive and Negative Cell Controls:

    • Include known CD45+ cells (e.g., THP-1, peripheral blood mononuclear cells) as positive controls

    • Include CD45- cells (e.g., epithelial cell lines) as negative controls

    • Ideally, include CD45 knockout cell lines derived from CD45+ parental lines to demonstrate specificity

  • Blocking Controls:

    • Pre-block with unlabeled anti-CD45 antibody before adding fluorochrome-conjugated anti-CD45 to confirm epitope specificity

    • Include competitive blocking with recombinant CD45 protein

  • Technical Validation:

    • Titrate antibody to determine optimal concentration (highest signal-to-noise ratio)

    • Assess stability over time, particularly for tandem dyes that may degrade

    • Include compensation controls for each fluorochrome to correct for spectral overlap

What are the critical factors in optimizing CD45 antibodies for radioimmunotherapy applications?

Optimization of CD45 antibodies for radioimmunotherapy requires attention to several critical factors:

  • Antibody Modification Strategies:

    • Direct radiolabeling approaches using various chelators (DOTA, DTPA) for metallic radionuclides or direct iodination methods

    • Pretargeting approaches using CD45 antibody-streptavidin conjugates followed by radiolabeled biotin, which have demonstrated superior tumor-to-blood ratios (20:1 vs. <1:1) compared to conventional approaches

  • Biodistribution Optimization:

    • Selection of antibody fragments (F(ab')2, Fab) or engineered formats to adjust circulation time

    • Use of clearing agents in pretargeted approaches to remove unbound antibody from circulation before administering the radionuclide

    • Dose fractionation strategies to improve therapeutic index

  • Radionuclide Selection Criteria:

    • Physical half-life matched to biological half-life of the antibody construct

    • Radiation characteristics (particle range, energy) appropriate for the size and distribution of target lesions

    • Chemistry compatible with selected conjugation strategy

  • Dosimetry Considerations:

    • Careful calculation of radiation absorbed dose to target tissues vs. critical normal organs

    • Biodistribution studies to determine optimal timing for therapeutic administration

    • Patient-specific dosing based on tracer studies with imaging radionuclides

  • Combination Strategies:

    • Integration with conditioning regimens for stem cell transplantation

    • Combination with other targeted agents or immunotherapies

    • Dose escalation protocols to determine maximum tolerated dose

These optimizations are essential for maximizing therapeutic efficacy while minimizing toxicity, particularly when targeting CD45, which is expressed on both malignant and normal hematopoietic cells .

How does antibody glycosylation influence the pharmacokinetics and immunogenicity of CD45-targeted therapeutics?

The glycosylation pattern of therapeutic antibodies targeting CD45 significantly impacts their in vivo behavior:

  • Fc Glycosylation Effects:

    • Core fucosylation reduces antibody-dependent cellular cytotoxicity (ADCC)

    • Terminal sialylation extends serum half-life but may reduce Fc receptor binding

    • Mannose-rich glycans increase clearance through mannose receptors in the liver

  • Immunogenicity Considerations:

    • Non-human glycoforms (e.g., α-Gal, Neu5Gc) from certain expression systems can trigger immune responses

    • Glycosylation heterogeneity may expose or mask immunogenic epitopes

    • Changes in glycosylation during storage can create neo-epitopes

  • Expression System Selection:

    • Human cell lines (e.g., HEK293) provide glycosylation most similar to natural human antibodies

    • CHO cells produce glycoforms with minor differences from human patterns

    • Plant and insect cell systems produce distinctly different glycoforms that may alter pharmacokinetics

  • Glycoengineering Approaches:

    • Knockout of specific glycosyltransferases to eliminate unwanted glycoforms

    • Expression in engineered cell lines to produce homogeneous glycosylation

    • Enzymatic remodeling of purified antibodies to optimize glycan structures

  • Analytical Characterization:

    • Mass spectrometry to define glycan composition and site occupancy

    • Lectin binding assays to identify specific glycan structures

    • In vitro functional assays to correlate glycoform with biological activity

These factors must be carefully considered when developing CD45-targeted therapeutics to ensure consistent efficacy and safety profiles across manufacturing batches and patient populations .

What strategies can improve the detection sensitivity of circulating tumor cells using CD45 antibodies?

Enhancing CTC detection sensitivity while maintaining specificity requires integrated optimization strategies:

  • Sample Preparation Optimization:

    • Red blood cell lysis buffers that preserve CTC viability and antigen expression

    • Density gradient centrifugation to enrich for mononuclear cells and CTCs

    • Immunomagnetic negative selection using anti-CD45 to deplete leukocytes before positive selection for CTCs

  • Antibody Cocktail Approach:

    • Use of multiple anti-CD45 antibody clones recognizing different epitopes to ensure detection of all leukocyte populations

    • Combination with positive selection markers (EpCAM, cytokeratins) in multiplexed assays

    • Inclusion of additional leukocyte markers (CD66b for granulocytes) to identify potential false negatives

  • Signal Amplification Methods:

    • Tyramide signal amplification for immunofluorescence applications

    • Quantum dot conjugates for improved signal-to-noise ratio

    • Multi-layer detection systems to enhance fluorescence intensity

  • Advanced Imaging Techniques:

    • Confocal microscopy with z-stacking to improve discrimination of cell boundaries

    • Spectral unmixing to resolve overlapping fluorophores

    • Artificial intelligence-based image analysis to distinguish CTCs from leukocytes based on subtle morphological differences

  • Workflow Integration:

    • Automated sample processing systems to minimize cell loss and variability

    • Standardized fixation protocols to preserve antigen expression

    • Quality control procedures including spike-in controls with known numbers of tumor cells

Implementation of these strategies has been shown to significantly improve the reliability of CTC enumeration in patient samples, which is critical for clinical applications in cancer diagnosis and monitoring .

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.