ICAM1 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Mechanisms of Action

ICAM1 antibodies exert therapeutic effects through:

  • Immune Modulation: Blocking LFA-1/ICAM1 interaction reduces leukocyte adhesion to endothelial cells, delaying graft rejection and inhibiting tumor-associated macrophage recruitment .

  • Antibody-Drug Conjugates (ADCs): Deliver cytotoxic payloads (e.g., MMAF, MMAE) to cancer cells via ICAM1 internalization, inducing microtubule disruption and apoptosis .

  • Fc-Dependent Functions: Enhanced Fc-engineered variants recruit NK cells/macrophages for ADCC/ADCP, particularly in multiple myeloma .

Preclinical Applications

Table 1: ICAM1 Monoclonal Antibodies and Their Applications

Antibody CloneTarget EpitopeKey ApplicationsSources
7F7ICAM1 epitopeBlocks LFA-1 interaction, inhibits TNF-α production in T cells/monocytes
Wehi-CAM-1 (1H4)85–115 kDa ICAM1Inhibits T-cell adhesion to endothelial cells, blocks lymphocyte aggregation
YN1/1.7.4Mouse CD54Neutralizes ICAM1 in vivo, reduces cytokine production (CXCL1, IFNγ, IL-17)
MSH-TP15Domain 1-2Fc-engineered variant enhances ADCC/ADCP in myeloma models
IC1-MMAF/MMAEICAM1 (human)Eradicates myeloma cells in xenografts; IC50: 7.3–68.7 pM in TNBC lines

Therapeutic Targets:

  • Cancer: ICAM1 is overexpressed in multiple myeloma, triple-negative breast cancer (TNBC), and certain lymphomas . ADCs (e.g., ICAM1-MMAF) achieve 100% survival in myeloma xenografts .

  • Transplantation: Anti-ICAM1 delays graft rejection by inhibiting leukocyte adhesion (e.g., rat small bowel models) .

  • Inflammation/Atherosclerosis: ICAM1 antibodies reduce leukocyte infiltration in inflammatory lesions and vascular plaques .

Clinical and Therapeutic Potential

Table 2: ICAM1 ADCs: Payloads and Efficacy

ADCPayloadTarget CancerEfficacy (In Vivo)Sources
ICAM1-MMAFMonomethyl auristatin FMultiple myelomaComplete tumor eradication in xenografts
ICAM1-MMAEMonomethyl auristatin ETNBCIC50: 13.1 pM in MDA-MB-436
ICAM1-Topoisomerase INovel topoisomerase ILung/liver cancersTumor regression at 5–6 mg/kg
ICAM1-DXdDeruxtecan (Dxd)Hematologic/solidComparable/better efficacy vs. ICAM1-MMAF

Challenges:

  • Naked Antibodies: Limited clinical efficacy due to transient ICAM1 blockade (e.g., bersanlimab) .

  • Toxicity: Off-target effects on endothelial cells/macrophages, though ADCs show selective tumor targeting .

Comparative Efficacy

ADC Payload Comparison:

  • MMAF/MMAE: Superior cytotoxicity in myeloma/TNBC (IC50: sub-nanomolar) .

  • Topoisomerase I Inhibitors: Novel payloads match/beat deruxtecan in preclinical models .

  • Fc-Engineered Variants: MSH-TP15 Fc-eng. enhances ADCC/ADCP, outperforming wild-type in myeloma .

Challenges and Future Directions

  • Resistance: Tumor antigen downregulation (e.g., CD38 in daratumumab-refractory myeloma) .

  • Combination Therapies: Pairing ICAM1 ADCs with checkpoint inhibitors or CAR-T cells to overcome resistance .

  • Novel Targets: Exploring ICAM1 in other cancers (e.g., melanoma, NSCLC) and non-oncologic applications .

Product Specs

Buffer
Liquid in phosphate-buffered saline (PBS) containing 50% glycerol, 0.5% bovine serum albumin (BSA), and 0.02% sodium azide.
Description

This monoclonal antibody against human ICAM1 is generated using a unique process involving immunization of mice with a synthesized peptide derived from the human CD54 protein. After immunization, B cells are isolated from the splenocytes of these mice and fused with myeloma cells to create hybridoma cells. These hybridomas are screened for production of the desired ICAM1 antibody. Once identified, the hybridoma cells are cultured in the mouse abdominal cavity to produce ascites fluid containing the antibody. The ICAM1 monoclonal antibody is then purified from the ascites fluid through affinity chromatography using a specific immunogen. This purified antibody is highly specific for human ICAM1 and suitable for applications such as ELISA and immunohistochemistry (IHC).

ICAM1, also known as CD54, is a cell surface glycoprotein that plays a critical role in various biological processes, particularly in immune responses. As a ligand for the leukocyte adhesion protein LFA-1 (integrin alpha-L/beta-2), ICAM1 facilitates leukocyte trans-endothelial migration by promoting the assembly of endothelial apical cups. This process is mediated by the activation of ARHGEF26/SGEF and RHOG. ICAM1 also interacts with other leukocyte integrins, such as Mac-1, contributing to leukocyte trafficking, antigen presentation, and T-cell activation.

Beyond its role in immune responses, ICAM1 has been implicated in the pathogenesis of various diseases, including inflammatory and autoimmune diseases, atherosclerosis, and cancer. Its involvement in these conditions highlights the importance of ICAM1 in cell-cell interactions and inflammatory processes.

Form
Liquid
Lead Time
We typically ship orders within 1-3 business days of receiving them. Delivery times may vary depending on the shipping method and location. For specific delivery timelines, please consult with your local distributor.
Synonyms
Antigen identified by monoclonal antibody BB2 antibody; BB 2 antibody; BB2 antibody; CD 54 antibody; CD_antigen=CD54 antibody; CD54 antibody; Cell surface glycoprotein P3.58 antibody; Human rhinovirus receptor antibody; ICAM 1 antibody; ICAM-1 antibody; ICAM1 antibody; ICAM1_HUMAN antibody; intercellular adhesion molecule 1 (CD54) antibody; intercellular adhesion molecule 1 (CD54), human rhinovirus receptor antibody; Intercellular adhesion molecule 1 antibody; Major group rhinovirus receptor antibody; MALA 2 antibody; MALA2 antibody; MyD 10 antibody; MyD10 antibody; P3.58 antibody; Surface antigen of activated B cells antibody; Surface antigen of activated B cells, BB2 antibody
Target Names
Uniprot No.

Target Background

Function

ICAM proteins serve as ligands for the leukocyte adhesion protein LFA-1 (integrin alpha-L/beta-2). During leukocyte trans-endothelial migration, engagement of ICAM1 promotes the assembly of endothelial apical cups through the activation of ARHGEF26/SGEF and RHOG.

ICAM1 also plays a significant role in microbial infections. It acts as a receptor for major receptor group rhinovirus A-B capsid proteins and Coxsackievirus A21 capsid proteins. Furthermore, upon Kaposi's sarcoma-associated herpesvirus/HHV-8 infection, ICAM1 is degraded by the viral E3 ubiquitin ligase MIR2. This degradation is believed to prevent lysis of infected cells by cytotoxic T-lymphocytes and NK cells.

Gene References Into Functions
  1. miR-335-5p targets the 3'UTR of ICAM-1, inhibiting its expression and preventing invasion and metastasis of thyroid cancer cells. PMID: 30119270
  2. Advanced glycation end products (AGEs) increase IL-6 and ICAM-1 expression via the receptor for AGEs (RAGE), mitogen-activated protein kinase (MAPK), and nuclear factor kappa B (NF-kappaB) pathways in human gingival fibroblasts (HGFs). This upregulation may contribute to the progression of periodontal diseases. PMID: 29193068
  3. Meta-analyses have not found an association between the ICAM-1 rs5498 polymorphism and diabetic retinopathy in patients with type 2 diabetes. PMID: 30419874
  4. Carbon monoxide releasing molecule-2 (CORM-2) inhibits Pseudomonas aeruginosa-induced prostaglandin E2 (PGE2)/IL-6/ICAM-1 expression and lung inflammatory responses by reducing reactive oxygen species generation and attenuating inflammatory pathways. PMID: 30007888
  5. Soluble factors released from smooth muscle cells can stimulate ICAM-1 expression in co-cultured endothelial cells, potentially leading to leukocyte migration into the subendothelial space. PMID: 29852173
  6. The CNOT1 protein provides a platform for the recruitment of TTP (Tristetraprolin) and CNOT7, and is involved in TTP-mediated ICAM1 and IL8 mRNA decay. PMID: 29956766
  7. ICAM-1 expression is not significantly linked to metastatic disease in pancreatic ductal adenocarcinoma. PMID: 29355490
  8. Meta-analysis suggests that the ICAM-1 gene rs5498 polymorphism decreases the risk of coronary artery disease (CAD). PMID: 30290609
  9. Serum levels of soluble ICAM1 are higher in young adults with reduced physical activity compared to those who participate in optimal physical activity. This finding was observed in a study conducted in Bulgaria with medical and dental students aged 20 +/-2 years. PMID: 29183155
  10. Treatment with tumor necrosis factor-alpha (TNF-alpha) and interleukin-10 (IL-10) can affect the expression of ICAM-1 and CD31 in human coronary artery endothelial cells. PMID: 29949812
  11. ICAM-1 is not a specific screening marker for pulmonary arterial hypertension in systemic sclerosis. PMID: 29687288
  12. The expression level of ICAM-1 determines the susceptibility of human endothelial cells to simulated microgravity. PMID: 29080356
  13. The combination of IL-6 -572C/G and ICAM-1 K469E polymorphisms has a synergistic effect on the onset of sudden sensorineural hearing loss. PMID: 29695657
  14. Peripheral blood lymphocyte subsets in patients with lung cancer differ from those in healthy individuals. Circulating CD44+ and CD54+ lymphocytes appear to be a promising criterion for predicting survival in lung cancer patients undergoing chemotherapy. PMID: 29148014
  15. Serum ICAM-1 levels are associated with type 2 diabetes mellitus with microalbuminuria, contributing to the severity of diabetic kidney disease. PMID: 29310968
  16. Serum CCL2 and soluble ICAM-1 (sICAM-1) concentrations are significantly decreased in central nervous system (CNS) tumors compared to a control group. Among the proteins tested in the serum, CCL2 exhibits a higher area under the receiver operating characteristic curve (AUC) compared to sICAM-1 in differentiating subjects with CNS brain tumors from non-tumoral subjects. PMID: 29086194
  17. Patient-derived anaplastic thyroid cancer (ATC) cells overexpress ICAM-1 and are largely eliminated by autologous ICAM-1 chimeric antigen receptor (CAR) T cells in vitro and in animal models. This study represents the first demonstration of CAR T therapy against both a metastatic thyroid cancer cell line and advanced ATC patient-derived tumors, showing significant therapeutic efficacy and survival benefit in animal models. PMID: 29025766
  18. Data suggest that ICAM-1 is an essential receptor for both acute hemorrhagic conjunctivitis (AHC)-causing and non-AHC strains of enterovirus. PMID: 29284752
  19. Cell adhesion and western blotting assays have shown that arachidin-1 attenuates tumor necrosis factor (TNF)-alpha-induced monocyte/endothelial cell adhesion and intercellular adhesion molecule-1 (ICAM-1) expression. PMID: 29115410
  20. Anthropometric and physiological parameters do not affect the response of ICAM-1 to exercise in healthy men. PMID: 29696063
  21. 15-Lipoxygenase-1 (15-LOX-1) expression in colon and prostate cancer cells leads to reduced angiogenesis. This effect may be mediated by an increase in the expression of both ICAM-1 and the anti-angiogenic protein thrombospondin-1 (TSP-1). PMID: 28757355
  22. Single nucleotide polymorphisms (SNPs) in the ICAM1 (rs1799969) and SERPINB2 (rs6103) genes have been found to be protective against thalidomide-induced peripheral neuropathy (TiPN). In children with inflammatory bowel disease, TiPN is common but usually mild and generally reversible. Cumulative dose seems to be the most relevant risk factor, while polymorphisms in genes involved in neuronal inflammation may provide protection. PMID: 28817461
  23. Analysis of aberrant DNA methylation and hydroxymethylation of the ICAM1 gene promoter in the thyrocytes of patients with autoimmune thyroiditis. PMID: 28388873
  24. The membrane-bound ICAM-1 isoform is essential and sufficient to promote inflammation-dependent extracellular matrix contraction, which favors cancer cell invasion. ICAM-1 mediates the generation of acto-myosin contractility downstream of the Src kinases in stromal fibroblasts. PMID: 27901489
  25. Thrombin-activated platelets release exosomes that convey microRNA (miRNA) between cells. miRNA-223 regulates the expression of adhesion molecules, including ICAM-1. miRNA-223 downregulates ICAM-1 primarily by impacting the NF-kappaB and MAPK pathways. PMID: 28460288
  26. Data suggest that obese children and adolescents have increased circulating biomarkers of endothelial dysfunction (specifically ICAM1) and early signs of renal damage, similar to those with type 1 diabetes (T1D). This observation confirms that obesity, like T1D, is a cardiovascular risk factor. PMID: 27246625
  27. Polymorphisms in ICAM-1 (and IL-17) have shown significant association with Guillain-Barré syndrome. PMID: 27595159
  28. Airway ICAM-1 expression is markedly upregulated in patients with chronic obstructive pulmonary disease (COPD), which could be crucial in rhinoviral and nontypeable Haemophilus influenzae (NTHi) infections. Parenchymal ICAM-1 is affected by smoking, with no further enhancement in COPD subjects. PMID: 28056984
  29. Soluble vascular cell adhesion molecule-1 (sVCAM-1) reflects xerostomia in primary Sjogren's syndrome. sICAM-1 and soluble E-selectin (sE-selectin) may serve as additional parameters of secondary Sjogren's syndrome activity. PMID: 29068581
  30. Atorvastatin strengthens Skp2 binding to FOXO1 or ICAM1, leading to ubiquitination and degradation. Skp2-dependent ubiquitination of major pathogenic molecules is the key mechanism for statins' protective effect on endothelial function in diabetes. PMID: 28802579
  31. Augmented expression of endothelial adhesion molecules ICAM1/VCAM1 is involved in the pathophysiology of patients with antiphospholipid syndrome. PMID: 29096830
  32. The CD133(+) CD44(+) CD54(+) cellular subpopulation of circulating tumor cells has prognostic value in colorectal cancer (CRC) patients with liver metastasis, particularly in the survival of CRC patients with liver metastasis who did not undergo surgical treatment for metastasis. PMID: 29105339
  33. Data indicate that the residue volume at phenylalanine (Phe) in alpha1-helix is critical for alpha(L)/beta(2) integrin (CD49a/CD18) activation and binding with soluble/immobilized ICAM1. PMID: 29079572
  34. Elevated serum uric acid concentration is significantly associated with inflammation of maternal systemic vasculature, as indicated by increased TNF-alpha and ICAM-1 expression in women with preeclampsia. PMID: 26511169
  35. ICAM-1 is associated with hypertension and stroke risk in women. PMID: 27235695
  36. Data suggest that CDH11, ICAM1, and CLDN3 are overexpressed in tumors compared to normal esophagus, normal gastric tissue, and non-dysplastic Barrett's esophagus. PMID: 27363029
  37. High levels of serum ICAM-1 are associated with the development of multiple organ failure. High levels of VCAM-1 are associated with both multiple organ failure and in-hospital mortality. PMID: 27701021
  38. Matrix stiffness-dependent ICAM-1 clustering is an important regulator of vascular inflammation. PMID: 27444067
  39. Perindopril (PD) increased CKIP-1 and Nrf2 levels in the kidney tissues of diabetic mice, improving the anti-oxidative effect and renal dysfunction. This ultimately reversed the upregulation of fibronectin (FN) and ICAM-1. PMID: 28286065
  40. Protein tyrosine phosphatase non-receptor type 22 (PTPN22) colocalized with its substrates at the leading edge of cells migrating on surfaces coated with the LFA-1 ligand intercellular adhesion molecule-1 (ICAM-1). PMID: 27703032
  41. SHP-2, through its association with ICAM-1, mediates ICAM-1-induced Src activation and modulates vascular endothelial cadherin (VE-cadherin) switching association with ICAM-1 or actin. This process negatively regulates neutrophil adhesion to endothelial cells and enhances their transendothelial migration. PMID: 28701303
  42. While no association was found between sICAM-1 levels and affective temperament scores, sICAM-1 was related to the state severity of manic symptoms. PMID: 27693464
  43. ICAM-1 is a pro-inflammatory protein. PMID: 28390825
  44. Human pancreatic microvascular endothelial cells (HPMCs) are capable of inhibiting the growth of gastrointestinal tumors through a mechanism involving the anti-adhesive capabilities of sICAM-1. PMID: 28323210
  45. Following transepithelial migration, neutrophil adhesion to ICAM-1 results in the activation of Akt and beta-catenin signaling, increased epithelial cell proliferation, and wound healing. PMID: 26732677
  46. P-selectin and ICAM-1 play roles in mediating THP-1 monocyte adhesion. PMID: 28262902
  47. Dissecting the molecular mechanism revealed that the p38-Notch1 axis is the main downstream signaling pathway in CD54-mediated regulation of cancer stem cells in prostate cancers. PMID: 28042317
  48. Knockdown of the expression of mcircRasGEF1B reduces lipopolysaccharide (LPS)-induced ICAM-1 expression. Additionally, mcircRasGEF1B regulates the stability of mature ICAM-1 mRNAs. PMID: 27362560
  49. This study shows that two adhesion molecules, shed as soluble forms, are elevated during the acute phase of leptospirosis: E-selectin and s-ICAM1. These molecules may interfere with the process of immune cell recruitment to clear Leptospira at tissue levels. PMID: 28686648
  50. Data suggest that CD2AP acts as a negative regulator of ICAM-1 clustering, which limits the formation of ICAM-1 adhesion complexes to prevent uncontrolled neutrophil adhesion and transcellular transmigration. PMID: 28484055

Show More

Hide All

Database Links

HGNC: 5344

OMIM: 147840

KEGG: hsa:3383

STRING: 9606.ENSP00000264832

UniGene: Hs.643447

Protein Families
Immunoglobulin superfamily, ICAM family
Subcellular Location
Membrane; Single-pass type I membrane protein.

Q&A

What is ICAM1 and what cellular functions does it mediate?

ICAM1 (CD54) is an intercellular adhesion molecule that serves as a ligand for the leukocyte adhesion protein LFA-1 (integrin alpha-L/beta-2). It plays a critical role in facilitating leukocyte trans-endothelial migration by promoting the assembly of endothelial apical cups through ARHGEF26/SGEF and RHOG activation . Beyond its role in immune cell trafficking, ICAM1 functions as a receptor for several pathogens, including major receptor group rhinovirus A-B capsid proteins and Coxsackievirus A21 capsid proteins . Interestingly, during Kaposi's sarcoma-associated herpesvirus (HHV-8) infection, ICAM1 is degraded by viral E3 ubiquitin ligase MIR2, likely as a viral immune evasion mechanism to prevent infected cell lysis by cytotoxic T-lymphocytes and NK cells .

How do I select the appropriate ICAM1 monoclonal antibody clone for my research?

Selection of the optimal ICAM1 antibody clone depends on several experimental factors. First, consider species reactivity—some clones like 1A29 demonstrate cross-reactivity with mouse, human, and rat ICAM1, making them versatile for comparative studies . Second, evaluate application compatibility—determine whether your antibody has been validated for your specific application (IHC-P, ICC, Flow Cytometry, Western Blot). For instance, clone 1A29 has been validated across multiple applications . Third, review citation records—antibodies cited in numerous publications (e.g., 70+ citations for clone 1A29) typically indicate reliable performance . Finally, consider epitope location—for functional studies, antibodies targeting the LFA-1 binding domain may be preferred, while antibodies recognizing intracellular domains are suitable for detection but not functional blockade experiments.

What expression patterns of ICAM1 should I expect across different tissue and cell types?

ICAM1 exhibits a dynamic expression pattern that varies by tissue type, activation state, and disease context. In normal tissues, ICAM1 is constitutively expressed at low levels on vascular endothelium, type 1 alveolar epithelial cells, and certain hematopoietic progenitors . Expression significantly increases upon inflammatory stimulation, particularly on activated vascular endothelium, macrophages, T-cells, and B-cells . In pathological contexts, ICAM1 demonstrates elevated expression in multiple cancer types, with particularly high levels in multiple myeloma cells and triple-negative breast cancer (TNBC) . Within TNBC tumors, ICAM1 expression correlates with tumor differentiation status, showing significantly higher expression in poorly differentiated (grade 3) tumors compared to moderately differentiated (grade 2) tumors . Additionally, ICAM1 expression positively correlates with BRCA1/2 and TP53 mutations in TNBC, potentially serving as a biomarker for specific molecular subtypes .

What are the optimal fixation and staining protocols for ICAM1 detection in tissue sections?

For optimal ICAM1 detection in tissue sections, a carefully optimized immunohistochemistry protocol is essential. Begin with formalin-fixed, paraffin-embedded (FFPE) specimens sectioned at 4-5μm thickness. After deparaffinization and rehydration, perform heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) for 20 minutes. For membrane-bound ICAM1 preservation, avoid harsh detergents during permeabilization—use 0.1% Triton X-100 for 10 minutes at room temperature. When blocking, employ a dual blocking strategy with 5-10% normal serum from the secondary antibody host species plus 1% BSA to minimize background. For primary antibody incubation, a 1:100-1:500 dilution of anti-ICAM1 monoclonal antibody (such as clone 1A29) is typically effective when incubated overnight at 4°C . To enhance detection sensitivity while maintaining specificity, utilize a polymer-based detection system rather than the traditional avidin-biotin complex. For dual immunofluorescence studies combining ICAM1 with other markers, sequential staining with appropriate spectral separation is recommended to prevent cross-reactivity.

How should I optimize flow cytometry protocols for accurate quantification of ICAM1 surface expression?

Accurate quantification of ICAM1 surface expression by flow cytometry requires careful protocol optimization. Begin with gentle cell dissociation methods—enzymatic dissociation with collagenase IV (for solid tissues) or Accutase (for cell lines) preserves surface ICAM1 better than trypsin, which can cleave surface proteins. Maintain cells at 4°C during processing to prevent receptor internalization or shedding. For staining, use saturating concentrations of fluorochrome-conjugated anti-ICAM1 antibodies (typically 0.25-1μg per million cells) and include appropriate isotype controls matched for fluorochrome type and concentration . To enable absolute quantification rather than relative mean fluorescence intensity (MFI), incorporate quantitative flow cytometry using calibration beads with known antibody binding capacity to establish a standard curve. This approach has proven valuable in studies comparing ICAM1 and CD38 expression levels on multiple myeloma cells . For multiparameter analysis, include markers to identify specific cell populations (e.g., CD138 for plasma cells in multiple myeloma studies) alongside ICAM1. When analyzing samples from therapeutic antibody trials, include a secondary detection step to evaluate potential epitope masking by circulating therapeutic antibodies.

What controls should I include when evaluating ICAM1 antibody specificity and sensitivity?

Comprehensive controls are essential for validating ICAM1 antibody specificity and sensitivity. For positive controls, include cell lines with documented high ICAM1 expression, such as cytokine-stimulated endothelial cells (HUVECs treated with TNF-α) or multiple myeloma cell lines . Negative controls should include both isotype controls (matched to the ICAM1 antibody's isotype, host species, and concentration) and ICAM1-negative or low-expressing cell lines. For definitive validation, employ ICAM1 knockout or knockdown models created using CRISPR-Cas9 or shRNA approaches, respectively. When working with new antibody clones, perform side-by-side comparisons with previously validated anti-ICAM1 antibodies targeting different epitopes. For cross-reactivity assessment across species, test the antibody against recombinant ICAM1 proteins from multiple species and validate with cells from each target species. Pre-adsorption controls (pre-incubating the antibody with purified ICAM1 protein before staining) can confirm binding specificity by demonstrating signal elimination. Finally, establish clear detection thresholds by titrating the antibody across multiple concentrations to determine the optimal signal-to-noise ratio.

How can I utilize ICAM1 antibodies to study leukocyte trans-endothelial migration in vitro?

To investigate leukocyte trans-endothelial migration using ICAM1 antibodies, implement a transwell migration assay with primary human endothelial cells (HUVECs or HDMECs) grown to confluence on 3-5μm pore transwell inserts. Pre-activate endothelial cells with TNF-α (10ng/ml, 4-6 hours) to upregulate ICAM1 expression . Establish experimental conditions comparing control IgG treatment versus function-blocking anti-ICAM1 antibodies (10-50μg/ml). For mechanistic studies examining ICAM1's role in endothelial cup formation, combine this approach with confocal microscopy using fluorescently-labeled leukocytes and anti-ICAM1 antibodies to visualize ICAM1 clustering during migration . To quantify the specific contribution of ICAM1-mediated adhesion versus other adhesion molecules, design a comprehensive blocking antibody panel including anti-ICAM1, anti-VCAM1, and anti-selectins. For real-time dynamics, adapt this system to live-cell imaging platforms, adding fluorescently-labeled anti-ICAM1 antibodies (non-blocking epitopes) to visualize ICAM1 redistribution during leukocyte adhesion and transmigration. This approach reveals how ICAM1 engagement promotes endothelial apical cup assembly through ARHGEF26/SGEF and RHOG activation, a critical step in the trans-endothelial migration process .

What techniques can I use to evaluate ICAM1 antibody internalization kinetics for antibody-drug conjugate development?

For antibody-drug conjugate (ADC) development, precise characterization of ICAM1 antibody internalization kinetics is critical. Begin with flow cytometry-based internalization assays using dual-labeled antibodies: primary anti-ICAM1 antibody followed by secondary antibodies with pH-sensitive fluorophores (e.g., pHrodo) that brighten in acidic endosomal/lysosomal compartments . Establish a time-course experiment (5, 15, 30, 60, 120 minutes) at physiological temperature (37°C) with parallel 4°C controls to differentiate active internalization from passive surface binding. For high-resolution visualization, implement confocal microscopy with fluorescently-labeled anti-ICAM1 antibodies co-stained with organelle markers (early endosome: EEA1; late endosome: Rab7; lysosome: LAMP1) to track intracellular trafficking pathways. Quantify colocalization using Pearson's correlation coefficient or Manders' overlap coefficient. For ADC development specifically, employ a patient specimen-based phage library selection approach similar to that described for identifying human antibodies that are rapidly internalized by malignant cells . This approach has successfully identified antibodies against ICAM1 that are ideal for the ADC format. When conjugating the antibody to cytotoxic payloads like monomethyl auristatin F (MMAF), optimize the drug-to-antibody ratio (DAR) and linker chemistry based on internalization rates to ensure efficient intracellular drug release .

How should I design experiments to evaluate ICAM1 expression in the context of tumor microenvironment interactions?

To comprehensively evaluate ICAM1 expression in the tumor microenvironment context, employ a multi-modal experimental approach. First, establish 3D co-culture systems combining tumor cells (e.g., multiple myeloma cells) with bone marrow stromal cells, osteoblasts, and immune components to recapitulate the native microenvironment . Within this system, use flow cytometry with a comprehensive antibody panel to quantify ICAM1 expression on distinct cell populations and correlate with other surface markers. Implement spatial transcriptomics and multiplexed immunohistochemistry on patient-derived samples to map ICAM1 expression patterns relative to specific microenvironmental niches. For functional studies, develop reporter systems (e.g., luciferase under ICAM1 promoter control) to monitor real-time regulation of ICAM1 expression in response to microenvironmental signals. Research has shown that ICAM1 expression is further accentuated by bone marrow microenvironmental factors in multiple myeloma . To evaluate therapeutic implications, design experiments comparing anti-ICAM1 antibody efficacy in 2D monoculture versus 3D co-culture systems, as microenvironmental interactions may influence antibody accessibility and efficacy. For in vivo validation, utilize orthotopic xenograft models with humanized microenvironments to assess how tumor-stroma interactions affect ICAM1 targeting in a physiologically relevant context .

What is the evidence supporting ICAM1 as a therapeutic target in multiple myeloma?

ICAM1 has emerged as a promising therapeutic target in multiple myeloma based on several compelling lines of evidence. First, quantitative flow cytometry studies have demonstrated that ICAM1 is highly expressed on multiple myeloma cells at levels comparable to CD38, a validated therapeutic target . Importantly, this high expression is maintained across disease progression from diagnosis to advanced stages . Second, ICAM1 expression is further accentuated by bone marrow microenvironmental factors, making it an ideal target within the primary disease niche . Third, ICAM1 shows differential overexpression on multiple myeloma cells compared to normal cells, providing a therapeutic window for selective targeting . Fourth, ICAM1 expression remains high even in daratumumab-refractory patients who show decreased CD38 expression, suggesting potential utility in resistant disease settings . Fifth, preclinical studies with anti-ICAM1 antibody-drug conjugates have demonstrated potent anti-myeloma cytotoxicity both in vitro and in vivo . In orthotopic xenograft models, anti-ICAM1 ADC completely eliminated disease cells and resulted in 100% survival for the duration of experiments (~200 days), surpassing the efficacy of naked anti-ICAM1 antibodies . While naked anti-ICAM1 antibodies showed limited clinical efficacy in human trials despite preclinical promise, the enhanced potency of ADC formulations provides a compelling rationale for continued therapeutic development targeting ICAM1 in multiple myeloma .

How do ICAM1 antibody-drug conjugates compare to naked antibodies in preclinical cancer models?

ICAM1 antibody-drug conjugates demonstrate markedly superior efficacy compared to naked antibodies across multiple preclinical cancer models. In orthotopic multiple myeloma xenograft models, anti-ICAM1 ADC conjugated to monomethyl auristatin F (MMAF) achieved complete tumor elimination and 100% survival over ~200 days, while naked anti-ICAM1 antibody showed limited efficacy in the same experimental setting . This dramatic improvement reflects the ADC's ability to deliver potent cytotoxic payloads directly to cancer cells, inducing microtubular catastrophe and cell death . Mechanistically, the enhanced efficacy of ICAM1-ADCs stems from ICAM1's favorable internalization kinetics—upon binding, ICAM1-antibody complexes are rapidly internalized, facilitating efficient intracellular delivery of conjugated cytotoxins . In triple-negative breast cancer models, rationally designed ICAM1-ADCs have similarly demonstrated the ability to eradicate tumors while sparing healthy tissues, leveraging ICAM1's differential expression pattern . Importantly, the ADC format overcomes limitations of naked antibodies observed in clinical trials, where a human anti-ICAM1 antibody (BI-505) was well-tolerated but showed limited clinical activity despite achieving doses that saturated ICAM1 binding sites . This clinical-preclinical discordance highlights how conjugation to cytotoxic payloads transforms ICAM1 targeting from a primarily signaling-based to a direct cytotoxic mechanism of action.

What considerations are important when developing ICAM1 antibodies for therapeutic applications in relation to potential on-target toxicities?

Development of ICAM1-targeted therapeutics requires careful consideration of potential on-target toxicities due to ICAM1's expression beyond tumor tissues. First, perform comprehensive immunohistochemical mapping of ICAM1 expression across normal tissues, with particular attention to activated vascular endothelium, type 1 alveolar epithelial cells, hematopoietic progenitors, and activated immune cells including macrophages, T-cells, and B-cells . For antibody-drug conjugates, consider strategic linker chemistry design—utilize linkers requiring specific enzymatic cleavage found predominantly in tumor cells to minimize payload release in normal tissues. Select antibody clones recognizing tumor-enriched ICAM1 epitopes or post-translational modifications where possible. When advancing to preclinical toxicology, non-human primate studies are essential given ICAM1's roles in immune function . Monitor potential immune-related adverse events resulting from ICAM1 blockade on immune cells, alongside standard toxicology parameters. For antibodies blocking ICAM1-ligand interactions, evaluate effects on physiological immune functions dependent on those interactions . Importantly, clinical trial design should include careful dose escalation with extensive safety monitoring. Previous clinical experience with naked anti-ICAM1 antibody (BI-505) demonstrated good tolerability as a single agent, providing an encouraging safety foundation . For enhanced-potency formats like ADCs, consider additional biodistribution studies using imaging techniques to confirm tumor-selective accumulation prior to clinical translation.

How can I address issues with background staining when using ICAM1 antibodies in immunohistochemistry?

Background staining with ICAM1 antibodies in immunohistochemistry often stems from endogenous expression on vasculature and immune cells, alongside technical factors. Implement a multi-faceted optimization strategy beginning with sample preparation—use freshly prepared tissue sections as prolonged storage can increase non-specific binding. For FFPE tissues, extend deparaffinization time and use fresh xylene to ensure complete paraffin removal. Optimize antigen retrieval carefully—ICAM1 epitopes may be sensitive to over-retrieval, so test multiple buffer systems (citrate pH 6.0 versus EDTA pH 9.0) and retrieval times. When blocking, employ a sequential blocking approach starting with avidin-biotin blocking (if using biotin-based detection), followed by 5-10% normal serum matching the secondary antibody host species, plus 1% BSA and 0.1% cold fish skin gelatin to reduce hydrophobic interactions. For antibody incubation, determine the optimal concentration through titration experiments (typically 1:100-1:500) and extend primary antibody incubation to overnight at 4°C to improve signal-to-noise ratio . Include additional washing steps with PBS containing 0.05-0.1% Tween-20 to remove weakly bound antibodies. For tissues with high endogenous peroxidase activity, employ dual peroxidase blocking (3% H₂O₂ for 10 minutes, followed by commercial peroxidase blocking solution for 10 minutes). Finally, use highly cross-adsorbed secondary antibodies specifically tested for minimal cross-reactivity against the species being examined.

What strategies can help resolve contradictory findings when comparing different ICAM1 detection methods?

Contradictory findings across different ICAM1 detection methods can be systematically addressed through a comprehensive validation approach. First, evaluate epitope differences—map the binding sites of antibodies used in each method to determine if they recognize distinct ICAM1 domains or isoforms. Some epitopes may be masked in certain experimental conditions or detection methods. Second, compare sample preparation protocols—ICAM1 detection can be significantly affected by fixation methods (formaldehyde versus alcohol-based fixatives), with some epitopes being particularly fixation-sensitive. Third, implement multi-platform validation—when flow cytometry and immunohistochemistry yield discrepant results, add a third method such as Western blotting or RNA-seq/qPCR to triangulate actual expression levels. Fourth, evaluate detection sensitivity thresholds—quantitative flow cytometry using calibration beads can establish absolute receptor density measurements to compare with semi-quantitative immunohistochemistry scoring . Fifth, consider ICAM1 biology—its expression is dynamically regulated by cytokines and stress conditions, so documentation of exact experimental conditions and timing is critical. Finally, implement side-by-side technical replicates using identical samples processed simultaneously for each detection method to eliminate batch-related variables. For particularly challenging contradictions, develop reporter cell lines expressing fluorescently-tagged ICAM1 to directly correlate protein expression with antibody binding across different detection platforms.

How should I interpret changes in ICAM1 expression in response to therapeutic interventions?

Interpreting ICAM1 expression changes following therapeutic interventions requires careful consideration of multiple factors. First, establish reliable baseline measurements using quantitative approaches like flow cytometry with antibody binding capacity beads to determine absolute receptor numbers per cell . Second, distinguish between alterations in surface versus total ICAM1 by comparing flow cytometry (surface detection) with Western blotting or intracellular flow cytometry (total protein). Therapeutic stress can trigger ICAM1 internalization without changing total expression. Third, evaluate the kinetics of expression changes through time-course experiments, as ICAM1 regulation may show biphasic responses—initial upregulation due to stress followed by downregulation in responding cells. Fourth, correlate ICAM1 expression changes with functional outcomes using appropriate assays (cell viability, apoptosis, migration) to determine if expression changes are mechanistically linked to therapeutic efficacy. Fifth, analyze ICAM1 expression alongside other relevant markers (e.g., CD38 in multiple myeloma) to identify potential compensatory mechanisms or resistance patterns . Studies have shown that ICAM1 expression remains high in daratumumab-refractory multiple myeloma when CD38 expression decreases, suggesting distinct regulatory mechanisms . Sixth, consider cell heterogeneity—bulk measurements may mask significant subpopulation differences, so employ single-cell analysis methods where possible. Finally, for in vivo therapeutic studies, compare expression in responding versus non-responding lesions to identify if ICAM1 changes correlate with treatment sensitivity or resistance.

How can ICAM1 expression patterns be utilized for patient stratification in clinical trials?

ICAM1 expression patterns offer significant potential for patient stratification in clinical trials through several approaches. First, establish standardized quantitative assessment methods—implement digital pathology with automated scoring algorithms for immunohistochemistry or standardized flow cytometry with calibration beads to ensure consistent quantification across multiple trial sites . Second, define clinically relevant expression thresholds—correlate ICAM1 expression levels with response rates in early-phase trials to identify optimal cut-points for subsequent patient selection. Research has shown that ICAM1 is differentially overexpressed on multiple myeloma cells compared to normal cells, providing a basis for such thresholds . Third, develop companion diagnostic assays—create validated IHC or flow cytometry-based assays specifically designed to identify patients likely to benefit from ICAM1-targeted therapies. Fourth, incorporate ICAM1 into multi-marker panels—combine ICAM1 assessment with other relevant biomarkers (e.g., CD38 status in multiple myeloma) to create integrated predictive models . For triple-negative breast cancer, ICAM1 expression correlates with molecular subtypes and tumor differentiation status, with significantly higher expression in poorly differentiated (grade 3) versus moderately differentiated (grade 2) tumors . Fifth, implement serial monitoring—measure ICAM1 expression changes during treatment to identify adaptive resistance mechanisms and inform treatment adjustments. Finally, explore circulating soluble ICAM1 (sICAM1) as a less invasive biomarker that could complement tissue-based assessment for longitudinal monitoring during clinical trials.

What novel approaches are being developed to enhance the specificity and efficacy of ICAM1-targeted therapeutics?

Researchers are developing several innovative approaches to enhance ICAM1-targeted therapeutics. First, next-generation antibody-drug conjugates are being designed with optimized drug-to-antibody ratios and cleavable linkers specifically tuned to ICAM1's internalization kinetics . The conjugation of anti-ICAM1 antibodies to auristatin derivatives has demonstrated potent cytotoxicity through targeted delivery and microtubular catastrophe induction . Second, bispecific antibody platforms are being explored to engage both ICAM1 and immune effector cells, potentially overcoming limitations of naked antibodies observed in clinical trials . Third, researchers are developing ICAM1-targeted CAR-T cell therapies, building on successes seen in preclinical thyroid cancer models with anti-ICAM1 CAR-T cells that showed efficacy without significant toxicity . Fourth, combination strategies pairing ICAM1-targeted agents with immune checkpoint inhibitors are being investigated to leverage ICAM1's role in immune cell interactions. Fifth, viral-based immunotherapies exploiting ICAM1's function as a receptor for oncolytic viruses like coxsackievirus A21 represent another innovative approach . Sixth, structure-guided antibody engineering is being employed to develop antibodies targeting tumor-specific ICAM1 conformations or post-translational modifications. Finally, researchers are creating Fc-modified ICAM1 antibodies with enhanced ADCC activity, which have shown improved anti-tumor activity in preclinical studies compared to conventional antibody formats .

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.