CCX4 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
CCX4 antibody; CAX10 antibody; At1g54115 antibody; F15I1.21Cation/calcium exchanger 4 antibody; AtCCX4 antibody; Protein CATION CALCIUM EXCHANGER 4 antibody; Protein CATION EXCHANGER 10 antibody
Target Names
CCX4
Uniprot No.

Target Background

Function
The antibody targets a membrane-localized H⁺-dependent K⁺ and Na⁺ transporter.
Database Links

KEGG: ath:AT1G54115

STRING: 3702.AT1G54115.1

UniGene: At.37278

Protein Families
Ca(2+):cation antiporter (CaCA) (TC 2.A.19) family, Cation/calcium exchanger (CCX) subfamily
Subcellular Location
Membrane; Multi-pass membrane protein.
Tissue Specificity
Expressed in roots, leaves, stems, flowers and pollen.

Q&A

What is CXCR4 and why is it a significant target for cancer therapy?

CXCR4 is a G protein-coupled receptor that functions as a receptor for the chemokine CXCL12 (also known as stromal cell-derived factor 1 or SDF-1). It plays pivotal roles during ontogenesis, including chemotaxis of neural and vascular progenitors, migration of hematopoietic precursors from fetal liver to bone marrow, and B-lymphocyte and myeloid cell development . In cancer biology, CXCR4 overexpression contributes to tumor growth, progression, and metastasis, making it an attractive therapeutic target . It has been identified as a prognostic marker for acute myeloid leukemia (AML) and other malignancies .

How is CXCR4 expression detected in tumor samples?

CXCR4 expression can be detected through several methodologies:

  • Flow cytometry: Cells are detached using a non-enzymatic cocktail, washed with flow cytometry buffer, and immunostained with allophycoerythrin (APC)-conjugated anti-human CXCR4 antibody (such as clone 12G5). Analysis is typically performed on a flow cytometer, with data processing through software like FlowJo .

  • Immunohistochemistry: Tissue sections can be stained with anti-CXCR4 antibodies to visualize receptor expression in tumor and stromal cells .

  • In vitro binding assays: These assays evaluate antibody binding to CXCR4-expressing cells, typically conducted at 37°C using one million cells. Blocking studies can be performed using an excess of unmodified CXCR4 antibody. After incubation, cells are rinsed with cold PBS and pellets counted in an automated gamma counter to determine immunoreactive fractions .

What are the main mechanisms of action for anti-CXCR4 antibodies?

Anti-CXCR4 antibodies exert their anti-tumor effects through multiple mechanisms:

  • Blocking CXCL12-CXCR4 interaction: They prevent the binding of CXCL12 to CXCR4, thereby inhibiting downstream signaling pathways that promote tumor growth and metastasis .

  • Inhibition of chemotaxis: CXCR4 antibodies inhibit CXCL12-induced migration and calcium flux, disrupting the homing of cancer cells to supportive microenvironments like bone marrow .

  • Direct induction of apoptosis: Studies show that antibodies like MDX-1338 can directly induce programmed cell death in CXCR4-expressing cancer cells, independent of their ability to block CXCL12 binding .

  • Disruption of tumor-stromal interactions: By interfering with CXCR4 signaling, these antibodies can disrupt interactions between cancer cells and the tumor microenvironment that support survival and drug resistance .

How does antibody affinity impact the efficacy and safety profile of anti-CXCR4 therapeutics?

The relationship between antibody affinity and therapeutic index is complex, particularly for targets like CXCR4 that are expressed in both malignant and normal tissues. Research into antibody-drug conjugates (ADCs) targeting CXCR4 has revealed that:

What strategies can overcome the challenge of CXCR4 expression in normal tissues when developing targeted therapies?

Targeting CXCR4 presents a challenge due to its expression in normal tissues, particularly hematopoietic cells. Several strategies have been developed to improve the therapeutic window:

  • ADC design optimization: Through empirical ADC design, researchers have created anti-CXCR4 ADCs with favorable therapeutic indices. The optimal configuration includes:

    • Non-cleavable linkers

    • Auristatin as payload

    • Drug-to-antibody ratio (DAR) of 4

    • Low-affinity antibody with effector-reduced Fc

  • Selective delivery systems: These systems can enhance tumor targeting while minimizing exposure to normal tissues. For example, 89Zr-labeled CXCR4 antibodies have demonstrated the ability to selectively identify CXCR4-overexpressing tumors for imaging and potential therapeutic applications .

  • Combinatorial approaches: Combining CXCR4 inhibitors with chemotherapeutics has shown synergistic therapeutic effects. While chemotherapeutics like gemcitabine may induce CXCR4 expression, their combination with CXCR4 inhibitors can enhance efficacy while potentially reducing the doses needed for each agent .

How can radioisotope-labeled anti-CXCR4 antibodies be optimized for diagnostic imaging?

Radioisotope-labeled anti-CXCR4 antibodies represent promising tools for non-invasive tumor phenotyping. Key considerations for optimizing these diagnostic agents include:

  • Isotope selection: Zirconium-89 (89Zr) with its half-life of 78.4 hours is well-suited for antibody labeling, allowing sufficient time for the labeled antibody to reach the target and clear from non-target tissues .

  • Antibody characteristics:

    • The fully human anti-hCXCR4 antibody MDX-1338 has demonstrated high affinity for CXCR4 (EC50 = 2 nM for inhibition of 125I-CXCL12)

    • Immunoreactive fraction determination is crucial for ensuring the labeled antibody maintains target binding capacity

  • Correlation with expression levels: Studies have shown that 89Zr-CXCR4-mAb uptake correlates with CXCR4 expression levels in tumors, enabling effective stratification of patients who might benefit from CXCR4-targeted therapies .

What are the recommended protocols for evaluating anti-CXCR4 antibody efficacy in vitro?

Several standardized assays are employed to evaluate the efficacy of anti-CXCR4 antibodies:

  • Binding assays:

    • Cell-based binding assays using CXCR4-expressing cell lines

    • Competition assays with labeled CXCL12 to assess the antibody's ability to block ligand binding

    • Scatchard analysis to determine binding affinity (Kd)

  • Functional assays:

    • Migration inhibition assays: Measuring the antibody's ability to block CXCL12-induced cell migration in Transwell systems

    • Calcium flux assays: Quantifying the inhibition of CXCL12-induced calcium mobilization

    • Apoptosis assays: Flow cytometry with Annexin V and propidium iodide staining to assess antibody-induced cell death

  • Signaling pathway analysis:

    • Western blotting to evaluate inhibition of CXCR4 downstream signaling (e.g., ERK phosphorylation)

    • Reporter gene assays to measure pathway activation

What experimental design is optimal for evaluating anti-CXCR4 antibodies in xenograft models?

The optimal experimental design for in vivo evaluation involves:

  • Model selection:

    • For hematological malignancies: AML, NHL, and multiple myeloma xenograft models in severe combined immunodeficient mice

    • For solid tumors: NSCLC (non-small cell lung cancer) and TNBC (triple-negative breast cancer) xenografts with varying CXCR4 expression levels

  • Treatment regimen optimization:

    • Dosing: 10 mg/kg of CXCR4-mAb administered intraperitoneally twice weekly has shown efficacy in multiple xenograft models

    • Duration: Treatment continuation until control tumors reach predetermined size limits (typically 3-4 weeks)

  • Efficacy measurements:

    • Tumor volume measurements using calipers

    • Final tumor weight determination

    • Proliferation assessment via BrdU staining

    • Apoptosis quantification through TUNEL assays

  • Experimental controls:

    • Vehicle control

    • Isotype-matched control antibody (control-mAb)

    • Positive control with established therapeutic (if available)

How can antibody-drug conjugates targeting CXCR4 be optimized through systematic design?

The optimization of anti-CXCR4 ADCs involves systematic variation of key parameters:

  • Linker-payload selection:

    • Non-cleavable linkers have demonstrated superior performance compared to cleavable alternatives for CXCR4-targeting ADCs

    • Auristatin derivatives (such as MMAE) have proven effective as cytotoxic payloads

  • Drug-to-antibody ratio (DAR) optimization:

    • Empirical testing of various DARs (2, 4, 8) has identified DAR = 4 as optimal, balancing cytotoxic potency with pharmacokinetic properties

  • Antibody engineering:

    • Affinity modulation: Lower affinity variants may provide better discrimination between high-expressing tumor cells and lower-expressing normal cells

    • Fc engineering: Effector-reduced formats minimize unwanted immune activation against normal CXCR4-expressing cells

  • Systematic screening workflow:

    • Initial in vitro screening across multiple cell lines with varying CXCR4 expression

    • PK/PD studies in non-tumor-bearing animals to assess circulation time and normal tissue toxicity

    • Efficacy and safety evaluation in relevant tumor models

How can researchers distinguish between CXCR4-dependent and independent effects of anti-CXCR4 antibodies?

To distinguish between specific and non-specific effects:

  • Knockdown/knockout validation:

    • Generate CXCR4 knockdown/knockout cell lines using siRNA or CRISPR-Cas9

    • Compare antibody effects on parental vs. CXCR4-deficient cells

    • Rescue experiments with exogenous CXCR4 expression

  • Competitive binding studies:

    • Pre-block cells with excess unlabeled CXCR4 antibody or small molecule inhibitors

    • Assess whether pre-blocking eliminates the observed effects

  • Domain-specific mutations:

    • Introduce mutations in specific CXCR4 domains to identify binding regions critical for antibody activity

    • Evaluate antibody effects on cells expressing mutant CXCR4 variants

  • Cross-reactivity assessment:

    • Test antibody effects on cells expressing related chemokine receptors

    • Ensure observed effects correlate with CXCR4 expression levels across multiple cell lines

What are the potential mechanisms of resistance to anti-CXCR4 therapies, and how can they be addressed?

Potential resistance mechanisms include:

  • Target downregulation or mutation:

    • Mutations in the CXCR4 epitope recognized by the antibody

    • Downregulation of CXCR4 surface expression

    • Alternative splicing leading to isoforms with reduced antibody binding

  • Compensatory pathway activation:

    • Upregulation of alternative chemokine receptors (e.g., CXCR7)

    • Activation of downstream signaling pathways independent of receptor engagement

    • Increased expression of anti-apoptotic proteins

  • Strategies to overcome resistance:

    • Combination therapies targeting multiple pathways

    • Development of bispecific antibodies recognizing CXCR4 and alternative targets

    • Sequential therapy approaches to prevent or delay resistance emergence

How does CXCR4 heterogeneity within tumors affect antibody efficacy, and what methodologies can address this challenge?

Intratumoral heterogeneity presents significant challenges:

What emerging combinations of anti-CXCR4 antibodies with other therapies show the most promise?

Promising combination approaches include:

  • With conventional chemotherapeutics:

    • While chemotherapeutics like gemcitabine can induce CXCR4 expression, their combination with CXCR4 inhibitors has shown synergistic therapeutic effects

    • The use of CXCR4 antibodies in combinatorial therapeutic approaches may be particularly beneficial due to fewer off-target effects compared to small molecule inhibitors

  • With immune checkpoint inhibitors:

    • CXCR4 blockade can potentially enhance infiltration of immune cells into tumors

    • Preliminary studies suggest potential synergy between anti-CXCR4 antibodies and immune checkpoint inhibitors in increasing anti-tumor immune responses

  • With targeted therapies:

    • Combining CXCR4 antibodies with kinase inhibitors targeting oncogenic drivers

    • Dual inhibition of CXCR4 and other chemokine receptors that may provide compensatory signaling

How can multimodal imaging approaches incorporating anti-CXCR4 antibodies advance personalized medicine?

Multimodal imaging with CXCR4 antibodies offers several advantages:

  • Patient stratification applications:

    • 89Zr-CXCR4-mAb PET imaging can identify tumors with high CXCR4 expression that are likely to respond to CXCR4-targeted therapies

    • This approach enables non-invasive, whole-body assessment of CXCR4 expression across primary tumors and metastases

  • Treatment monitoring capabilities:

    • Sequential imaging to assess changes in CXCR4 expression during treatment

    • Early identification of developing resistance mechanisms

    • Evaluation of combination therapy effects on CXCR4 expression and signaling

  • Technical considerations for implementation:

    • Optimization of tracer pharmacokinetics and imaging protocols

    • Development of quantitative metrics for standardized interpretation

    • Integration with other molecular imaging modalities for comprehensive tumor characterization

What novel antibody engineering approaches might enhance the therapeutic index of anti-CXCR4 antibodies?

Advanced antibody engineering strategies include:

  • Format innovations:

    • Bispecific antibodies targeting CXCR4 and tumor-specific antigens to enhance selectivity

    • Conditionally active antibodies that become fully functional only in the tumor microenvironment

    • pH-sensitive binding to preferentially target the acidic tumor microenvironment

  • Payload delivery optimization:

    • Site-specific conjugation technologies for more homogeneous ADCs

    • Novel payloads with improved therapeutic windows

    • Cleavable linkers responsive to tumor-specific conditions

  • Immunological engagement strategies:

    • Fc engineering to modulate interaction with immune effector cells

    • Combination with immune-stimulating payloads

    • Development of tri-functional antibodies that simultaneously block CXCR4 signaling, deliver cytotoxic payloads, and engage immune effectors

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.