CXCR4 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Production

CXCR4 recombinant monoclonal antibodies are synthesized through recombinant DNA techniques. The process involves:

  1. Immunization: Animals are immunized with a human CXCR4-derived peptide to generate antigen-specific B cells .

  2. Hybridoma Formation: B cells are fused with myeloma cells to create hybridomas capable of continuous antibody production .

  3. Gene Cloning: The cDNA encoding the antibody is sequenced, synthesized, and cloned into expression vectors .

  4. Expression and Purification: Vectors are transfected into host cells (e.g., CHO or HEK293), followed by antibody purification via affinity chromatography .

This method ensures high specificity and batch-to-batch consistency, critical for research and therapeutic applications.

Key Applications

CXCR4 recombinant mAbs are utilized across diverse fields:

ApplicationDetailsReference
Immunofluorescence (IF)Detects CXCR4 expression in cell lines and tissues (Recommended dilution: 1:20–1:200)
Cancer ResearchIdentifies CXCR4 overexpression in invasive ductal carcinoma (moderate to strong membranous/cytoplasmic staining)
HIV-1 InhibitionBlocks X4-tropic HIV-1 entry by competing with gp120 for CXCR4 binding
Therapeutic UseInduces apoptosis in hematologic cancers (e.g., AML, multiple myeloma) and inhibits metastasis

Mechanisms of Action

CXCR4 recombinant mAbs exert effects through multiple pathways:

  • Receptor Blockade: Competes with CXCL12 (SDF-1) or HIV-1 gp120 for CXCR4 binding, inhibiting chemotaxis and viral entry .

  • Apoptosis Induction: Cross-linking CXCR4 triggers caspase-dependent apoptosis in cancer cells (e.g., AML cell lines) .

  • Immune Modulation: Enhances β-chemokine (MIP-1α, RANTES) secretion, downregulating CCR5 and reducing R5-tropic HIV-1 infection .

For example, the A120 mAb targets extracellular loops 1/2 (ECL1/2), suppressing both X4 and R5 HIV-1 strains by upregulating endogenous antiviral chemokines . In contrast, MDX-1338 (BMS-936564), a fully human IgG4 mAb, blocks CXCL12-induced calcium flux and migration while inducing tumor cell apoptosis .

Comparative Analysis with Other Anti-CXCR4 Agents

CXCR4 recombinant mAbs exhibit distinct advantages over small-molecule antagonists or peptide inhibitors:

FeatureRecombinant mAbsSmall Molecules (e.g., AMD3100)
Target SpecificityHigh (binds conformational epitopes)Moderate (binds transmembrane pockets)
HIV-1 InhibitionBlocks X4 and R5 strains via multiple mechanismsPrimarily targets X4 strains
Therapeutic EfficacyInduces apoptosis; effective as monotherapyRequires combination therapies
Safety ProfileReduced leukocytosis and hematopoietic toxicityRisk of cardiac arrhythmias

Notably, recombinant mAbs like UMB-2 enable reliable CXCR4 detection in formalin-fixed tissues, unlike older antibodies (e.g., 12G5), which fail in fixed samples .

Research and Clinical Findings

  • Cancer Therapy: Anti-CXCR4 antibody-drug conjugates (ADCs) with auristatin payloads show efficacy in xenograft models of therapy-resistant multiple myeloma and AML, achieving tumor regression at 10 mg/kg doses .

  • HIV-1 Prevention: A120 mAb reduces R5 HIV-1 infection in PBMCs by 80% through CCR5 downregulation and β-chemokine induction .

  • Safety: Low-affinity recombinant mAbs minimize off-target effects on normal CXCR4+ tissues (e.g., hematopoietic stem cells), enhancing therapeutic index .

Future Directions

Ongoing research focuses on optimizing drug-to-antibody ratios (DAR) and linker-payload configurations to improve ADC efficacy . Additionally, bispecific antibodies targeting CXCR4 and PD-1/PD-L1 are under exploration to enhance antitumor immune responses .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

The production of CXCR4 recombinant monoclonal antibody involves a multi-step process. Initially, the CXCR4 monoclonal antibody is harvested, and its gene sequence is meticulously analyzed. Subsequently, the CXCR4 monoclonal antibody gene is integrated into a plasmid vector and then transfected into a host cell line for cultivation. The immunogen employed for CXCR4 monoclonal antibody production is a recombinant human CXCR4 protein. The CXCR4 recombinant monoclonal is purified using affinity chromatography from cell culture supernatant, and its specificity is rigorously evaluated through ELISA. This antibody exhibits reactivity towards human CXCR4 protein.

CXCR4 is a G protein-coupled receptor protein that binds to the chemokine CXCL12. It plays a pivotal role in cell migration, proliferation, and survival, particularly in immune cells and cancer cells. CXCR4 is involved in various physiological processes, including hematopoiesis, angiogenesis, organogenesis, and immune surveillance. Dysregulation of CXCR4 signaling has been implicated in a range of diseases, including cancer, HIV infection, and inflammatory disorders.

Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery timelines may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery estimates.
Synonyms
CXCR4; C-X-C chemokine receptor type 4; CXC-R4; CXCR-4; FB22; Fusin; HM89; LCR1; Leukocyte-derived seven transmembrane domain receptor; LESTR; Lipopolysaccharide-associated protein 3; LAP-3; LPS-associated protein 3; NPYRL; Stromal cell-derived factor 1 receptor; SDF-1 receptor; CD antigen CD184
Target Names
Uniprot No.

Target Background

Function
CXCR4 serves as a receptor for the C-X-C chemokine CXCL12/SDF-1, mediating signal transduction by increasing intracellular calcium ion levels and enhancing MAPK1/MAPK3 activation. It is also implicated in the AKT signaling cascade. CXCR4 plays a role in regulating cell migration, notably during wound healing. It acts as a receptor for extracellular ubiquitin, leading to enhanced intracellular calcium ions and reduced cellular cAMP levels. CXCR4 binds bacterial lipopolysaccharide (LPS) and mediates LPS-induced inflammatory responses, including TNF secretion by monocytes. This receptor is involved in hematopoiesis and cardiac ventricular septum formation. It also plays an essential role in the vascularization of the gastrointestinal tract, likely by regulating vascular branching and/or remodeling processes in endothelial cells. CXCR4 is involved in cerebellar development. In the CNS, it potentially mediates hippocampal-neuron survival. Notably, in microbial infections, CXCR4 acts as a coreceptor (with CD4 being the primary receptor) for human immunodeficiency virus-1/HIV-1 X4 isolates and as a primary receptor for certain HIV-2 isolates. It facilitates Env-mediated fusion of the virus.
Gene References Into Functions
  1. Functional analysis in human breast cancer cells revealed that LL-37 induced the internalization of CXCR4 through interaction with Glu268, a residue of CXCR4, independent of the binding pocket (Asp171, Asp262, and Glu288) for CXCR4 inhibitor AMD3100. This finding suggests that LL-37 acts as a distinct agonist of CXCR4. PMID: 30251699
  2. Collectively, these data indicate that the S18-2 protein induces epithelial to mesenchymal cell transition through the TWIST2/E-cadherin signaling pathway and, consequently, CXCR4-mediated migration of prostate cancer cells. PMID: 29396484
  3. A study identified a variant near the chemokine receptor CXCR4 that was jointly associated with an increased risk for progressive supranuclear palsy and Parkinson's disease. Additionally, in a mouse model of tauopathy, expression of CXCR4 and functionally associated genes was significantly altered in regions of the mouse brain that accumulate neurofibrillary tangles most prominently. PMID: 29636460
  4. The expression of CXCR4 and mTOR was found to be negatively correlated with remission. Kaplan-Meier analysis indicated a significant decrease in the rate of progression-free survival (PFS) and overall survival (OS) in patients exhibiting positive CXCR4 and mTOR expression. PMID: 28952842
  5. Findings demonstrate that the CXCL12-CXCR4 axis promotes migration, invasion, and EMT processes in B-CPAP cells, at least partially, by activating the NF-kappaB signaling pathway. PMID: 29316404
  6. Results indicate that non-oxidizable HMGB1 induces sustained cardiac fibroblasts migration despite the redox state of the environment and by altering the CXCL12/CXCR4 axis. This effect influences proper cardiac remodeling following an infarction. PMID: 28716707
  7. CXCR4 is highly abundant in the zona glomerulosa and in aldosterone-producing adenomas, suggesting a significant role in adrenocortical physiology. This finding further presents CXCR4 as a potential target for molecular imaging of aldosterone-producing tissue. PMID: 29279316
  8. High CXCR4 expression is associated with bladder cancer progression. PMID: 30015971
  9. The overexpression of CXCR4 increased sVCAM1, and the sVCAM1 secreted from CXCR4-overexpressing non-small cell lung carcinoma cells recruited and arrested additional osteoclast progenitors to promote osteoclastogenesis. PMID: 30355915
  10. MiR-125b functions as an important downstream mediator upon the activation of the CXCL12/CXCR4 axis. PMID: 28176874
  11. Data suggest that CXCL12 and its receptor CXCR4 are critical in maintaining homeostasis, specifically during hematopoiesis. Current clinical trials (especially in hematological tumors) are investigating whether adding CXCR4 inhibitors to impair tumor dissemination will enhance the effectiveness of ongoing anti-cancer treatments. (CXCL12 = C-X-C motif chemokine ligand 12; CXCR4 = C-X-C motif chemokine receptor-4) [REVIEW] PMID: 29288743
  12. Hypoxia-induced expression of CXCR4 promoted trophoblast cell migration and invasion via the activation of HIF1alpha, which is crucial during placentation. PMID: 29786753
  13. CXCR4 expression was up-regulated in NSCLC cell lines. Inhibition of CXCR4 may reduce EMT, invasion, and migration of NSCLC cells. PMID: 29972256
  14. Results suggest that BCP-ALL cells create a leukemic niche that attracts leukemic cells in a CXCR4/CXCL12-independent manner. PMID: 28619846
  15. Serum CXCR4 and CXCL12 levels increase significantly in septic neonates, making them valuable markers in the diagnosis of neonatal sepsis. Serum concentrations of both chemokines represent promising novel biomarkers for neonatal sepsis. PMID: 28562124
  16. This study provides an atomistic-level description of the activation dynamics of the C-X-C chemokine receptor type 4 (CXCR4), a class A GPCR and significant drug target. PMID: 30238101
  17. CXCL12 and CXCR4 polymorphisms may be risk factors for hepatocellular carcinoma (HCC) and may serve as potential HCC markers. PMID: 29741398
  18. The results suggest that CXCR4 is a predictor of poor prognosis and may serve as a biomarker of the mesenchymal subtype in patients with Glioblastoma multiforme (GBM). Additionally, CXCR4 mediated the mitogen-activated protein kinase signaling pathway, which was identified specifically in patients with mesenchymal GBM. PMID: 29767255
  19. Stromal cell-derived factor-1/C-X-C chemokine receptor type 4 axis induces human dental pulp stem cell migration through FAK/PI3K/Akt and GSK3beta/beta-catenin pathways. PMID: 28067275
  20. EGFR overexpression and mutations lead to changes in the biological characteristics of human lung adenocarcinoma cells through the CXCR4/CXCL12 signaling pathway. PMID: 30037369
  21. BACH1 may inhibit the progression of colorectal cancer through the BACH1/CXCR4 pathway. PMID: 29481800
  22. High CXCR4 expression is associated with differential expression patterns in adenocarcinoma and squamous cell carcinoma of the lung relative to small cell lung cancer. PMID: 30076481
  23. No significant associations were found between mean plasma levels of either CXCL12 or CXCR4 with age, gender, tumor site, tumor size, lymph-node involvement, or tumor stage. PMID: 29693336
  24. The aim of the present study was to assess whether fibrosis markers, estrogen receptor (ER)alpha, and the stromal derived factor (SDF)1/CXC chemokine receptor type 4 (CXCR4) axis are abnormally expressed in the endometrium of patients with intrauterine adhesions. PMID: 29568895
  25. Daily oral administration of AMD070 significantly inhibited lung metastasis of B88SDF1 cells in nude mice. These results indicated that AMD070 could be a novel orally bioavailable inhibitor of oral cancer metastasis. PMID: 29749473
  26. These data demonstrated that JWA suppressed the migration/invasion of breast carcinoma cells by downregulating the expression of CXCR4, suggesting that JWA may hold prognostic and therapeutic potential for patients with breast cancer. PMID: 29658570
  27. These findings suggest that SDF1 (e.g., presented on proteoglycans) can rapidly activate integrins in an allosteric manner by binding to site 2 in the absence of CXCR4. The allosteric integrin activation by SDF1 represents a novel target for drug discovery. PMID: 29301984
  28. High CXCR4 expression is associated with lymph node metastasis in colorectal cancer. PMID: 29719205
  29. This effect can be suppressed by miR-613 through direct downregulation of CXCR4. PMID: 29845707
  30. These results underscore a key role for the CXCR4-CXCL12 chemokine axis in breast cancer progression and highlight the prognostic significance of this chemokine axis for breast cancer survival. PMID: 29516917
  31. CXCR4 can induce PI3Kdelta inhibitor resistance in ABC DLBCL. PMID: 29472546
  32. Our results demonstrated greater expression of pRET and CXCR4 in cisplatin-resistant neuroblastomas (NBs). Vandetanib significantly inhibited SHSY5YR cell proliferation, colony formation, and invasion, while downregulating pRET and CXCR4 expression. PMID: 29436676
  33. Disruption of the CXCR4/CXCL12 axis by CXCR4 antagonist AMD3100 blocked the contribution of both cancer and stromal cells to the metastatic cascade in the liver. PMID: 29436696
  34. LncRNA PRNCR1 up-regulates CXCR4 through targeting miR-211-5p, which affects osteogenic differentiation and thus contributes to osteolysis after hip replacement. PMID: 29775758
  35. Results demonstrated that miR-1246 inhibited cell invasion and EMT process by targeting CXCR4 and blocking JAK/STAT and PI3K/AKT signal pathways in lung cancer cells. PMID: 29171984
  36. High CXCR4 expression is associated with hepatocellular and cholangiocellular carcinomas in tumor capillaries. PMID: 29282035
  37. Each of the CXCR4-derived peptides exhibited high affinity for GroEL with a binding stoichiometry near seven. It was observed that the peptides interact with the paired alpha helices in the apical domain of the chaperonin. Each of the two chaperonin rings is capable of accommodating all seven CXCR4 peptides bound to GroEL under saturation conditions. ATP alone or combined with GroES promoted the peptide release from... PMID: 29627450
  38. Down-regulation of CXCR4 significantly reduced cell proliferation, while remarkably increased cell apoptosis and apoptotic protein expression levels in osteosarcoma cells. PMID: 29734183
  39. Quercetin suppressed breast cancer stem cell proliferation, self-renewal, and invasiveness. It also lowered the expression levels of proteins related to tumorigenesis and cancer progression, such as aldehyde dehydrogenase 1A1, C-X-C chemokine receptor type 4, mucin 1, and epithelial cell adhesion molecules. PMID: 29353288
  40. Icaritin enhances MSC proliferation, chemotaxis to stromal cell-derived factor-1, and osteogenic differentiation through STAT-3 activation, with a consequential up-regulation in the expression and activity of CXCR4. Phosphorylated STAT-3 binds the CXCR4 promoter, upregulating its expression. PMID: 29679717
  41. CXCL11 did not significantly alter the (13)C-(1)H-HSQC spectrum of CXCR4. Our findings point towards ubiquitin as a biased agonist of CXCR4. PMID: 28455789
  42. High CXCR4 expression may define a specific subtype of sporadic malignant peripheral nerve sheath tumor with a favorable prognosis. PMID: 29020982
  43. Data support the importance of SDF-1 and CXCR4 expression for loco-regional control and overall survival in HNSCC after primary radiochemotherapy. PMID: 29061496
  44. Presence of SST5, CXCR4, and ETA on tumor cells and of SST3, CXCR4, and ETA on microvessels gradually increased from grade II to grade IV tumors. PMID: 29696364
  45. These data revealed that CXCR4 is a novel hepatocellular carcinoma (HCC) vascular marker for vessel sprouting and could serve as a potential therapeutic target and a predictive factor for sorafenib treatment in patients with HCC. PMID: 28223275
  46. Hetero-oligomerization of a1B/D-adrenergic receptor with the chemokine (C-X-C motif) receptor 4:atypical chemokine receptor 3 heteromeric complex is required for a1B/Dadrenergic receptor function. PMID: 28862946
  47. CXCR4+ cells were increased in response to DOXO, mainly in human cardiac mesenchymal progenitor cells (CmPC), a subpopulation with regenerative potential. PMID: 28837147
  48. This work demonstrates distinct roles for the SDF-1/CXCR4 or CXCR7 network in human induced pluripotent stem cell-derived ventricular cardiomyocyte specification, maturation, and function. PMID: 28711757
  49. Implantation of IGF1R(+) human dental pulp mesenchymal stem cells exerted enhanced neuroplasticity via integrating inputs from both CXCR4 and IGF1R signaling pathways. PMID: 27586516
  50. CXCR4 was overexpressed on systemic lupus erythematosus B cells, positively correlating with disease activity and kidney involvement. PMID: 27665947

Show More

Hide All

Database Links

HGNC: 2561

OMIM: 162643

KEGG: hsa:7852

STRING: 9606.ENSP00000386884

UniGene: Hs.593413

Involvement In Disease
WHIM syndrome (WHIMS)
Protein Families
G-protein coupled receptor 1 family
Subcellular Location
Cell membrane; Multi-pass membrane protein. Cell junction. Early endosome. Late endosome. Lysosome. Note=In unstimulated cells, diffuse pattern on plasma membrane. On agonist stimulation, colocalizes with ITCH at the plasma membrane where it becomes ubiquitinated. In the presence of antigen, distributes to the immunological synapse forming at the T-cell-APC contact area, where it localizes at the peripheral and distal supramolecular activation cluster (SMAC).
Tissue Specificity
Expressed in numerous tissues, such as peripheral blood leukocytes, spleen, thymus, spinal cord, heart, placenta, lung, liver, skeletal muscle, kidney, pancreas, cerebellum, cerebral cortex and medulla (in microglia as well as in astrocytes), brain microv

Q&A

What is CXCR4 and why is it important in research?

CXCR4 is a G-protein coupled receptor belonging to the chemokine receptor family with a molecular mass of 39.7 kDa comprising 352 amino acid residues in humans. It functions primarily as a receptor for stromal cell-derived factor 1 (SDF-1/CXCL12), transducing signals by increasing intracellular calcium levels and enhancing MAPK1/MAPK3 activation . CXCR4 plays crucial roles in multiple biological processes including embryonic development, immune cell trafficking, and hematopoiesis. The receptor is also implicated in various pathological conditions, serving as a co-receptor for HIV-1 entry into CD4+ cells and as a prognostic marker in cancer progression . Its broad expression on immune cells, stem cells, and cancer cells makes it a significant target for both basic science investigation and therapeutic development.

How do recombinant monoclonal antibodies against CXCR4 differ from traditional antibodies?

Recombinant CXCR4 monoclonal antibodies are produced using recombinant DNA technology, which offers several advantages over traditional hybridoma-derived antibodies. The production process involves sequencing the cDNA of CXCR4 antibody-producing hybridomas, synthesizing the gene that codes for the monoclonal antibody, cloning it into a vector, and transfecting it into cells for cultivation . The recombinant antibodies are then purified from cell culture supernatant using affinity chromatography. This approach allows for precise control over antibody characteristics, improved batch-to-batch consistency, and the ability to introduce specific modifications to enhance functionality. Unlike traditional methods that rely solely on hybridoma stability, recombinant technology enables production of fully human antibodies with reduced immunogenicity and tailored effector functions, making them particularly valuable for both research and therapeutic applications.

What are the common experimental applications for CXCR4 recombinant monoclonal antibodies?

CXCR4 recombinant monoclonal antibodies serve multiple experimental purposes across various research disciplines. The most common applications include:

ApplicationDilution RangePurposeDetection Method
Flow Cytometry1:20-1:100Cell surface expression quantificationFluorophore-conjugated secondary antibodies
Immunohistochemistry (IHC)1:20-1:200Tissue localization studiesChromogenic or fluorescent detection
Western Blot1:500-1:2000Protein expression analysisChemiluminescence
ELISA1:100-1:1000Quantitative detection in solutionColorimetric or fluorometric readout
Immunofluorescence (IF)1:20-1:200Subcellular localizationFluorescence microscopy

These antibodies are particularly valuable in research on HIV infection mechanisms, cancer cell migration and metastasis, and hematopoietic stem cell mobilization . They can be used to block the CXCL12-CXCR4 interaction, inducing functional changes that permit detailed mechanistic studies of downstream signaling events and cellular responses.

How should researchers evaluate epitope specificity when selecting CXCR4 antibodies?

Selecting CXCR4 antibodies with appropriate epitope specificity is crucial for experimental success. Studies have demonstrated that antibodies targeting different regions of CXCR4 can elicit diverse and sometimes opposing biological effects. For example, the A80 monoclonal antibody, which binds to the third extracellular loop (ECL3) of CXCR4, enhances syncytium formation in HIV-infected cells, while antibodies A145 and A120, which target the N-terminal domain and a conformational epitope involving ECL1 and ECL2 respectively, inhibit HIV-1 infection .

To systematically evaluate epitope specificity, researchers should:

  • Conduct competitive binding assays with known ligands (CXCL12/SDF-1) to determine if the antibody competes for the ligand binding site

  • Perform cross-blocking studies with other characterized anti-CXCR4 antibodies

  • Use cells expressing CXCR4 mutants with alterations in specific domains to map binding regions

  • Consider using epitope binning techniques to classify antibodies into groups with similar binding characteristics

  • Validate functional consequences of binding through downstream assays like calcium flux or cell migration

This comprehensive approach ensures selection of antibodies with epitope specificities appropriate for the intended experimental or therapeutic application.

What protocols are most effective for measuring receptor occupancy in CXCR4 studies?

Effective receptor occupancy measurement is critical when evaluating anti-CXCR4 antibodies, particularly in preclinical and clinical development. Based on research with monoclonal antibodies like MEDI3185, a systematic approach should include both free and total surface CXCR4 assays .

For optimal receptor occupancy analysis:

  • Develop displacement interference controls during assay development to ensure accuracy

  • Implement a dual-parameter flow cytometry approach to simultaneously measure:

    • Free CXCR4 (using a competing fluorescently-labeled antibody)

    • Total surface CXCR4 (using a non-competing antibody targeting a different epitope)

  • Monitor time-dependent and dose-dependent changes in receptor expression

  • Incorporate controls for anti-drug antibody (ADA) interference

  • Perform parallel pharmacokinetic analysis to correlate serum antibody levels with receptor occupancy

Research has shown that surface CXCR4 expression can increase following antibody dosing, with different cell populations (lymphocytes, monocytes, granulocytes) showing varying magnitude of upregulation . This dynamic expression pattern must be accounted for when interpreting receptor occupancy data.

How can researchers troubleshoot assay interference issues in CXCR4 antibody studies?

Assay interference is a significant challenge in CXCR4 antibody research, potentially leading to misleading results and interpretation. Studies with MEDI3185 have revealed several sources of interference and strategies to address them :

Interference TypeManifestationTroubleshooting Approach
Displacement interferenceCompeting antibodies causing false negative resultsUse non-competing detection antibodies; validate with displacement controls
Anti-drug antibody (ADA) neutralizationParadoxical results where surface CXCR4 appears occupied without detectable antibody in serumCompare pharmacokinetic, ADA, and total CXCR4 results; implement ADA screening assays
Target modulation effectsChanges in surface expression altering interpretationMonitor total CXCR4 as a reliable marker unaffected by ADA
Epitope maskingSteric hindrance preventing detection antibody bindingTest multiple detection antibodies targeting different epitopes

Researchers should implement parallel assays measuring both free and total receptor, and validate findings through orthogonal methods. Additionally, inclusion of appropriate controls for each potential interference mechanism is essential for accurate data interpretation. When apparent discrepancies arise, comparative analysis of multiple parameters often reveals the underlying cause of interference .

How does CXCR4 antibody-induced signaling differ from natural ligand activation?

CXCR4 antibodies can induce signaling cascades that both overlap with and diverge from natural ligand (CXCL12/SDF-1) activation, revealing complex receptor biology. The A80 monoclonal antibody demonstrates this phenomenon clearly - it uniquely induces agglutination of peripheral blood mononuclear cells (PBMC) and CEM cells without activating calcium mobilization, which is typically observed with SDF-1 stimulation . This suggests antibodies can selectively activate certain downstream pathways while bypassing others.

Key differences include:

  • Calcium flux: SDF-1 reliably induces calcium mobilization, while antibodies like A80 do not trigger this response

  • Cell adhesion: Some antibodies (A80) promote homologous lymphocyte adhesion in a ligand-independent manner

  • Receptor internalization: SDF-1 rapidly induces CXCR4 internalization, whereas antibody effects on internalization vary by epitope

  • Apoptosis induction: BMS-936564/MDX-1338 induces apoptosis in various cell lines, representing a mechanism distinct from competitive antagonism

  • Signaling kinetics: Antibodies typically induce more prolonged signaling compared to the transient nature of ligand-induced activation

Understanding these mechanistic differences has significant implications for developing CXCR4-targeted therapeutics with precise functional outcomes. This knowledge allows researchers to design antibodies that selectively modulate specific aspects of CXCR4 biology.

What role do CXCR4 antibodies play in understanding cancer progression and potential therapeutics?

CXCR4 antibodies have emerged as powerful tools for investigating cancer biology and developing targeted therapies, particularly for hematological malignancies. The BMS-936564/MDX-1338 antibody exemplifies this application, showing efficacy against acute myeloid leukemia (AML), non-Hodgkin lymphoma (NHL), chronic lymphoid leukemia (CLL), and multiple myeloma in both in vitro studies and xenograft models .

Research has revealed several mechanisms through which CXCR4 antibodies affect cancer progression:

  • Direct induction of apoptosis: BMS-936564/MDX-1338 induces programmed cell death in cancer cell lines, independent of its blocking activity

  • Disruption of stromal interactions: By blocking CXCL12-CXCR4 signaling, antibodies can mobilize cancer cells from protective niches in the bone marrow

  • Inhibition of metastasis: Preventing CXCR4-mediated homing to CXCL12-expressing tissues reduces metastatic spread

  • Modulation of tumor microenvironment: CXCR4 blockade affects immune cell trafficking, potentially enhancing anti-tumor immunity

Recent studies also indicate that CXCR4 inhibition can enhance the efficacy of CD19-targeted therapies in B-cell malignancies, suggesting valuable combination approaches . The therapeutic potential is further supported by immunohistochemical studies showing variable CXCR4 expression in invasive ductal carcinoma, with cytoplasmic and membranous staining patterns that correlate with disease progression .

How can researchers design experiments to evaluate the efficacy of CXCR4 antibodies in different disease models?

Designing robust experiments to evaluate CXCR4 antibody efficacy requires careful consideration of disease-specific mechanisms and appropriate models. Based on successful approaches with antibodies like BMS-936564/MDX-1338, researchers should implement a multi-tiered experimental strategy :

In vitro evaluation:

  • Affinity determination: Measure binding affinity to CXCR4-expressing cells (typically low nanomolar range is effective)

  • Functional antagonism: Assess the antibody's ability to block CXCL12 binding and inhibit downstream functions:

    • Migration inhibition assays (EC₅₀ values)

    • Calcium flux measurements

  • Direct cellular effects: Quantify apoptosis induction using flow cytometry with Annexin V/PI staining

  • Cell line panel testing: Evaluate efficacy across multiple cell lines representing the disease of interest

In vivo evaluation:

  • Pharmacokinetic/pharmacodynamic studies: Determine dosing regimens that achieve target receptor occupancy

  • Xenograft models: Establish tumors from relevant cell lines or patient-derived samples

  • Efficacy endpoints: Monitor tumor growth inhibition, survival, and biomarker changes

  • Combination studies: Test with standard-of-care therapeutics for potential synergistic effects

Important considerations include:

  • Monitoring receptor modulation (as CXCR4 surface levels can increase after antibody treatment)

  • Accounting for potential anti-drug antibody responses

  • Including appropriate control antibodies (isotype-matched)

  • Implementing clinically relevant dosing schedules

This comprehensive approach allows for thorough assessment of antibody efficacy across multiple dimensions of disease biology.

How are CXCR4 antibodies being integrated with other immunotherapeutic approaches?

CXCR4 antibodies are increasingly being investigated as complementary agents in combination immunotherapy strategies. Recent research suggests that CXCR4 inhibition can significantly enhance the efficacy of other immunotherapeutic approaches through several mechanisms:

  • Combination with CD19-targeted therapies: Studies indicate that CXCR4 inhibition enhances the efficacy of CD19 monoclonal antibodies in B-cell lymphomas, suggesting synergistic potential for treating hematological malignancies

  • Overcoming stromal protection: By disrupting the CXCR4-CXCL12 axis, these antibodies can mobilize malignant cells from protective niches, rendering them more susceptible to conventional chemotherapies and targeted agents

  • Immune checkpoint inhibitor combinations: Emerging research suggests CXCR4 blockade may alter the tumor microenvironment in ways that enhance T-cell infiltration and activation, potentially improving responses to PD-1/PD-L1 inhibitors

  • CAR-T cell therapy enhancement: CXCR4 inhibition may improve CAR-T cell trafficking to tumor sites and counteract immune suppression mechanisms

To effectively integrate CXCR4 antibodies with other immunotherapies, researchers must carefully consider timing and sequencing of combination treatments, potential for overlapping toxicities, and disease-specific factors that may influence response rates. The field is advancing toward more rational design of combination approaches based on mechanistic understanding of how CXCR4 signaling interfaces with other immune pathways.

What novel modifications to CXCR4 antibodies are being explored to enhance their research and therapeutic utility?

Researchers are actively exploring innovative modifications to enhance the functionality and versatility of CXCR4 antibodies:

Modification TypePurposeResearch Applications
Fc engineeringModulation of effector functionsEnhanced ADCC or CDC for cancer therapy; reduced effector functions for pure antagonism
Bispecific formatsSimultaneous targeting of CXCR4 and complementary targetsRedirecting immune cells to CXCR4+ tumors; dual pathway inhibition
Antibody-drug conjugatesTargeted delivery of cytotoxic payloadsSelective elimination of CXCR4-expressing cells with reduced systemic toxicity
pH-dependent bindingEnhanced tumor tissue selectivityPreferential binding at tumor microenvironment pH for improved specificity
Fragment-based approachesImproved tissue penetrationEnhanced access to solid tumors and biological barriers

The BMS-936564/MDX-1338 antibody exemplifies some of these approaches, being developed as a fully human IgG4 monoclonal antibody that specifically recognizes human CXCR4 . Similarly, MEDI3185 incorporates three amino acid mutations in the Fc region, designated as a "triple mutant" (TM) molecule, resulting in the ablation of antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity .

These modifications allow researchers to develop antibodies with precisely tuned properties for specific applications, whether focused on pure antagonism, selective cell killing, or complex immunomodulatory functions.

How do epigenetic and post-translational modifications of CXCR4 affect antibody binding and function?

The interaction between CXCR4 antibodies and their target is significantly influenced by the receptor's complex regulation through epigenetic and post-translational modifications. Though not extensively covered in the search results, this emerging research area merits attention for several reasons:

  • Glycosylation status: CXCR4 undergoes N-glycosylation at its N-terminal domain, which can alter antibody accessibility to certain epitopes. Researchers should consider how differential glycosylation patterns across cell types might affect antibody binding kinetics and epitope recognition.

  • Receptor phosphorylation: Upon activation, CXCR4 undergoes phosphorylation at serine residues in its C-terminal domain, triggering β-arrestin recruitment and receptor internalization. This conformational change can significantly impact the binding of antibodies targeting C-terminal or conformational epitopes.

  • Sulfation of tyrosine residues: The N-terminal domain of CXCR4 contains sulfated tyrosines that play a crucial role in CXCL12 binding. Antibodies targeting this region may show variable binding depending on sulfation status.

  • Ubiquitination and receptor degradation: CXCR4 can undergo ubiquitin-mediated degradation, affecting surface expression levels and potentially confounding receptor occupancy measurements.

  • Epigenetic regulation of expression: Researchers should account for how epigenetic mechanisms influence CXCR4 expression levels when designing experiments, as hypoxia and inflammatory conditions can upregulate CXCR4 through epigenetic mechanisms.

These modifications create heterogeneity in CXCR4 presentation that may not be apparent in simplified experimental systems but can significantly impact antibody performance in complex biological contexts. Future research should systematically characterize how these modifications affect antibody binding parameters and downstream functional consequences.

What are the key considerations for researchers beginning work with CXCR4 recombinant monoclonal antibodies?

Researchers entering the field of CXCR4 antibody research should prioritize several critical factors to ensure experimental validity and interpretability. First, epitope specificity is paramount, as antibodies targeting different regions of CXCR4 (N-terminus, extracellular loops) exhibit dramatically different functional effects . Second, careful validation of antibody performance in the specific cellular context of interest is essential, as CXCR4 expression and regulation vary substantially across cell types . Third, researchers should implement robust controls for potential assay interference, particularly when measuring receptor occupancy or functional outcomes .

The dynamic nature of CXCR4 expression following antibody treatment presents a particular challenge, as surface levels can increase significantly after exposure to anti-CXCR4 antibodies . This phenomenon must be accounted for in experimental design and data interpretation. Additionally, researchers should carefully select application-appropriate antibody formats and detection methods, considering factors such as internalization kinetics and potential steric hindrances.

Finally, interdisciplinary collaboration is increasingly valuable as CXCR4 research spans immunology, oncology, virology, and developmental biology. Integrating insights across these disciplines may reveal novel applications and understanding of CXCR4 biology.

How might advances in CXCR4 antibody technology contribute to personalized medicine approaches?

Advances in CXCR4 antibody technology are poised to make significant contributions to personalized medicine, particularly in oncology and immunological disorders. The varying expression patterns of CXCR4 across patients and disease states provide an opportunity for biomarker-guided therapeutic approaches. Immunohistochemical studies already demonstrate heterogeneous CXCR4 expression in tumors like invasive ductal carcinoma, with cytoplasmic and membranous staining patterns that could potentially predict treatment response .

Future personalized approaches may include:

  • Expression-based patient stratification: Selecting patients with high CXCR4 expression for targeted therapy based on immunohistochemistry or molecular diagnostics

  • Functional diagnostics: Developing ex vivo assays to predict individual patient response to various CXCR4-targeted antibodies

  • Combination therapy optimization: Using molecular profiles to determine optimal combinations of CXCR4 antibodies with other targeted agents

  • Monitoring tools: Employing circulating tumor cell CXCR4 expression as a liquid biopsy approach for real-time therapy adjustment

  • Antibody engineering: Creating patient-specific antibody variants optimized for individual disease characteristics

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.