ICOS Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery times may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery details.
Synonyms
Activation inducible lymphocyte immunomediatory molecule antibody; Activation-inducible lymphocyte immunomediatory molecule antibody; AILIM antibody; CD278 antibody; CD278 antigen antibody; CD28-related Protein-1 antibody; CRP1 antibody; CVID1 antibody; ICOS antibody; ICOS_HUMAN antibody; Inducible costimulator antibody; Inducible T cell co stimulator antibody; Inducible T-cell costimulator antibody; MGC39850 antibody
Target Names
Uniprot No.

Target Background

Function
ICOS Antibody enhances all basic T-cell responses to foreign antigens, including proliferation, lymphokine secretion, upregulation of cell-cell interaction molecules, and effective B-cell antibody secretion assistance. It is crucial for efficient T-B cell interaction and normal antibody responses to T-cell-dependent antigens. While it does not upregulate interleukin-2 production, it superinduces interleukin-10 synthesis. It prevents pre-activated T-cell apoptosis and plays a critical role in CD40-mediated immunoglobulin isotype class switching.
Gene References Into Functions
  • ICOS (-693A/G) gene polymorphism is associated with Colon Cancer Risk. PMID: 29580042
  • Altered soluble (s)PD1 and sICOS serum levels in different Hepatitis b (HBV) groups may reflect T-cell activation dysregulation and potentially link to the HBV pathological process. PMID: 28983583
  • A potential association between chronic spontaneous urticaria and polymorphisms in the ICOS gene has been identified. This report is the first to suggest a possible association of the ICOS rs6726035 TT genotype with CSU pathogenesis. PMID: 28940644
  • Genetic polymorphisms of ICOS function as sex-dependent risk factors for acute rejection development in an Iranian kidney transplant population. PMID: 28031007
  • NUFIP2 binding to Roquin promotes recognition and regulation of ICOS mRNA. PMID: 29352114
  • OX40 and ICOS collaborate in a non-redundant manner to maximize and prolong the T follicular helper cell response generated after acute virus infection. PMID: 27895177
  • High ICOS expression is associated with colorectal neoplasms. PMID: 27197182
  • This is the first replication association study of rs4325730 upstream of ICOS with AIH in the Japanese population, and rs4325730G is a risk allele. PMID: 27974812
  • The induction pathway of ICOS(+) Foxp3(+) cells was analyzed. PMID: 27756099
  • The ratio of ICOS(+) /PD-1(+) Tfh cells and the frequency of IL-21(+) Tfh cells may serve as indicators for evaluating idiopathic membranous nephropathy development. PMID: 26845249
  • Follicular helper T cell differentiation involves a multistage process that includes BCL6, other transcription factors, cytokines, and costimulation through ICOS and several other molecules (Review). PMID: 26120879
  • A novel deletion mutation in Pakistani siblings presents in childhood with a combined immunodeficiency associated with enteritis, hepatitis, and impaired antiviral immunity. PMID: 26399252
  • Findings demonstrate that Tregs from inflamed lung tissue of sarcoidosis patients exhibit an ICOS(high) phenotype. High-level ICOS expression was restricted to Tregs from inflamed lung tissue and was absent in blood Tregs of sarcoidosis patients and in lung and blood Tregs of healthy volunteers. This suggests a potential implication of the ICOS/ICOS-L immune-regulatory axis in disease activity and resolution. PMID: 26415669
  • Findings indicate that circulating memory Tfh cells, particularly CCR7+ICOS+ memory Tfh cells, may be associated with multiple sclerosis relapse and could serve as a new therapeutic target. PMID: 26231034
  • Expression of mutant caspase-9 correlated with downregulation of BAFFR (B-cell-activating factor belonging to the TNF family (BAFF) receptor) in B cells and ICOS (inducible T-cell costimulator) in T cells. PMID: 25569260
  • This study found that ICOS rs1559931 SNP was associated with decreased hepatitis B virus-related hepatocellular carcinoma risk in the studied Chinese Han population, except for patients with natural clearance of hepatitis B virus. PMID: 26074057
  • Thymus medullary epithelial cells promote regulatory T-cell generation by stimulating interleukin-2 production via ICOS. PMID: 25210803
  • Data indicate that T cells redirected with an inducible T-cell costimulator ICOS-based chimeric antigen receptors CAR maintained a core molecular signature characteristic of TH17 cells. PMID: 24986688
  • Patients with recent diagnoses of primary biliary cirrhosis may exhibit increased numbers of circulating ICOS(+) and IL-21(+) Tfh and CD38(+) plasma cells. PMID: 25404409
  • Aberrant sPD1 and sICOS serum levels may reflect T-cell activation dysregulation and are associated with the pathological injury of chronic HCV infection. PMID: 23426717
  • Data indicate that Tregs and its inducible costimulatory molecule (ICOS+) subsets are decreased in patients with myocardial infarction (MI) and stable angina (SA). PMID: 22426168
  • Data indicate the expression of programmed cell death protein 1 (PD-1), inducible T-cell Costimulator (ICOS), Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4), and T cell immunoglobulin and mucin protein 3 (Tim-3) in T-cells from geriatric and younger subjects. PMID: 24083425
  • The associations between costimulatory molecule gene polymorphisms including CTLA4, PD-1, ICOS, and CD28 with active cytomegalovirus (CMV) infection were evaluated in hematopoietic stem cell transplant patients. PMID: 24057239
  • The acquisition of B-cell stimulating properties by naive cord blood CD4 T cells required the STAT3-dependent expression of ICOS and IL-21. PMID: 23923047
  • SNPs of ICOS showed no association with Hashimoto thyroiditis. PMID: 23138463
  • In a transgenic graft-vs-host disease (GVHD) model, mouse CD4 T cells depend more heavily on the ICOS/phosphatidylinositol 3-kinase (PI3K) signaling axis, while CD8 T cells can induce GVHD utilizing PI3K-independent ICOS signaling mechanisms. PMID: 23729441
  • A molecular and computational diagnostic approach identifies FOXP3, ICOS, CD52, and CASP1 as the most informative biomarkers in acute graft-versus-host disease. PMID: 22491736
  • Results suggest that the serum level of sICOS is increased in patients with diffuse cutaneous SSc and correlates with the severity and activity of skin sclerosis and interstitial lung disease. PMID: 23053221
  • Augmented ICOS signaling may contribute to systemic sclerosis pathogenesis during early progressive disease. Soluble ICOS levels can potentially serve as a serum marker for systemic sclerosis activity and severity. PMID: 23024058
  • ICOS-Fc inhibited the adhesion of dendritic cells to vascular and lymphoid endothelial cells, their migratory activity, and the expression of the Rac-1 activator beta-Pix involved in cell motility. PMID: 23275603
  • ICOS expression on CD4 + CD45RO + and CD8 + CD4RO + T cells was significantly increased in systemic lupus erythematosus (SLE) patients. ICOS expression was higher in lupus nephritis patients compared to SLE patients without lupus nephritis. PMID: 21479882
  • Genetic variation in the ICOS gene is associated with acute rejection of liver transplantation. PMID: 22579879
  • ICOS gene polymorphisms may affect breast cancer risk and indicate that some SNPs are associated with breast cancer characteristics in a northern Chinese population. PMID: 21917182
  • The constellation of specific alleles in CTLA-4, CD28, and ICOS genes contributes to the susceptibility and clinical course of non-small-cell lung cancer. PMID: 21669243
  • Increased quantities of inducible costimulator-positive Tregs may influence IgG4 production in IgG4-related autoimmune pancreatitis. PMID: 21926547
  • NPM-ALK induces expression of the growth-promoting receptor ICOS. PMID: 21765024
  • ICOS polymorphisms were significantly different in pemphigus, suggesting that genetically determined abnormal function of costimulatory receptors in T cells may be associated with pemphigus pathogenesis. PMID: 21084022
  • Circulating neoplastic T cells correspond more often to a CD10-positive subset than an ICOS-positive subset in Angioimmunoblastic T-cell lymphoma. PMID: 21499231
  • These findings indicate that the investigated ICOS polymorphisms do not modulate B-CLL risk in the Polish population but are associated with disease dynamics, particularly with the time to Rai stage progression. PMID: 21526489
  • A trend for reduced ICOS expression was observed after gluten-free diet treatment in the intestine of celiac disease children. PMID: 21288140
  • Polymorphisms of three major genes involved in immune response regulation, CTLA4, CD28, and ICOS, might be involved in the development of clinical variables in IgA nephropathy. PMID: 21677403
  • Data suggest that preeclampsia is associated with ICOS but not with CTLA-4 or CD28 gene polymorphisms. PMID: 21160481
  • The role of ICOS in allergen-induced T-cell activation in allergic asthma and rhinitis is reported. PMID: 21356099
  • These results suggest that polymorphisms in the CD28, CTLA4, and ICOS genes do not influence the risk of melanoma or its prognosis in the investigated population. PMID: 19672595
  • In active ulcerative colitis, CD86 and ICOS were overexpressed in intestinal epithelial cells and lamina propria mononuclear cells. PMID: 20388394
  • Increased amounts of CD86 or ICOS-positive lamina propria mononuclear cells and enterocytes suggest that co-stimulatory molecules may play a role in Crohn disease pathogenesis. PMID: 20019769
  • Results reveal a vital role for ICOS signaling in the generation and maintenance of human T(H)17 cells and suggest that components of this pathway could be therapeutically targeted to treat cancer or chronic infection. PMID: 20980695
  • High ICOS expression is associated with lymphomas of T follicular helper cell. PMID: 20207847
  • The rs6726035 in ICOS was significantly associated with RA. Comparing the genotype frequencies in the codominant model (CC vs. CT, CC vs. TT) in the rs6726035 SNP revealed that the C allele appeared to be decreased in patients with RA. PMID: 20113255
  • ICOS polymorphisms were not associated with susceptibility to cervical squamous cell (CSCC) or with histologic grade of CSCC. PMID: 19913589

Show More

Hide All

Database Links

HGNC: 5351

OMIM: 604558

KEGG: hsa:29851

STRING: 9606.ENSP00000319476

UniGene: Hs.56247

Involvement In Disease
Immunodeficiency, common variable, 1 (CVID1)
Subcellular Location
[Isoform 1]: Cell membrane; Single-pass type I membrane protein.; [Isoform 2]: Secreted.
Tissue Specificity
Activated T-cells. Highly expressed on tonsillar T-cells, which are closely associated with B-cells in the apical light zone of germinal centers, the site of terminal B-cell maturation. Expressed at lower levels in thymus, lung, lymph node and peripheral

Q&A

What is ICOS and why is it significant in immunological research?

ICOS (Inducible T cell costimulator, also known as CD278) is a member of the CD28/B7 superfamily that delivers positive co-stimulatory signals to activated T cells upon binding to its ligand (ICOS-L) . The protein is approximately 22.6 kilodaltons in mass and plays a crucial role in T cell activation, differentiation, and immune response regulation . ICOS is particularly significant in immunological research because it serves as a marker for T Follicular Helper Cells and is implicated in various immune-mediated conditions, including autoimmune diseases and cancer . Additionally, the ICOS/ICOS-L pathway has emerged as a promising target for immunotherapy interventions .

How does ICOS protein structure relate to its function?

The ICOS protein adopts an Ig-fold structure similar to CTLA-4 and CD28, with distinct structural features that determine its binding specificity . Crystal structure analysis at 3.3 Å resolution reveals that ICOS utilizes a central FDPPPF motif, with the PPP sequence adopting a high-energy cis-trans-cis conformation flanked by aromatic residues that engage with ICOS-L . Unlike its family members CTLA-4 and CD28, ICOS employs a second set of residues within its CC' loop that contribute significantly to binding specificity with ICOS-L . Additionally, the ICOS N110 N-linked glycan participates in ICOS-L binding, further enhancing interaction specificity . These structural characteristics determine ICOS's unique signaling properties and make it a distinct therapeutic target compared to other co-stimulatory molecules.

What are the recommended applications for ICOS antibody detection?

ICOS antibodies can be used in multiple experimental applications with specific recommended dilutions:

ApplicationRecommended DilutionPositive Detection
Western Blot (WB)1:200-1:1000Jurkat cells, HL-60 cells
Immunohistochemistry (IHC)1:1000-1:4000Human tonsillitis tissue
Immunofluorescence (IF)-P1:50-1:500Human tonsillitis tissue
ELISASample-dependentVaries by protocol

For optimal results, antibody titration is recommended for each specific testing system . When performing IHC, antigen retrieval with TE buffer pH 9.0 is suggested, though citrate buffer pH 6.0 may serve as an alternative . These methodological considerations are crucial for obtaining reliable and reproducible results in ICOS expression studies.

How should researchers select the appropriate ICOS antibody for their specific application?

When selecting an ICOS antibody for research, consider multiple factors: (1) Target epitope specificity - determine whether the antibody recognizes extracellular or intracellular domains based on your experimental needs; (2) Species reactivity - verify the antibody's reactivity with your experimental model (human, mouse, rat, etc.) ; (3) Application compatibility - ensure the antibody is validated for your desired application (WB, IHC, flow cytometry, etc.) ; (4) Clone type - consider whether monoclonal antibodies (for specific epitopes) or polyclonal antibodies (for broader detection) better suit your research question; (5) Conjugation requirements - determine if your experiment requires unconjugated antibodies or those conjugated with fluorophores, enzymes, or biotin ; and (6) Validation data - review existing literature and manufacturer validation to confirm antibody specificity and performance in contexts similar to your experimental design.

What are the key considerations for detecting ICOS expression in tissue samples?

For optimal detection of ICOS expression in tissue samples, researchers should address several methodological aspects: (1) Fixation protocol - formalin-fixed paraffin-embedded tissues typically require antigen retrieval with TE buffer pH 9.0 or citrate buffer pH 6.0 ; (2) Antibody concentration - titrate antibodies at recommended dilutions (1:1000-1:4000 for IHC) to determine optimal signal-to-noise ratio ; (3) Positive controls - include known ICOS-expressing tissues like human tonsillitis tissue to validate staining procedures ; (4) Expression pattern interpretation - ICOS is predominantly expressed on activated T cells and regulatory T cells, appearing as membrane staining with potential cytoplasmic components ; (5) Cross-reactivity assessment - validate specificity through appropriate negative controls and blocking peptides; and (6) Multiplexing considerations - when performing co-staining with other markers, ensure antibody compatibility and optimize sequential staining protocols to prevent interference.

How can discrepancies in observed molecular weight of ICOS be explained and addressed?

ICOS protein exhibits molecular weight discrepancies between calculated (23 kDa for 199 amino acids) and observed weights (28 kDa and 45-50 kDa bands in Western blot) . These differences can be explained and addressed through several approaches:

  • Post-translational modifications - N-linked glycosylation at N110 contributes to higher molecular weight bands

  • Protein dimerization - ICOS may form dimers under certain sample preparation conditions

  • Sample preparation techniques - different lysis buffers and denaturation conditions may affect observed band patterns

  • Tissue/cell type variations - expression levels and modification patterns may differ between various T cell subsets

  • Antibody specificity - ensure antibodies recognize the core protein rather than modification-dependent epitopes

When troubleshooting molecular weight discrepancies, researchers should consider enzymatic deglycosylation assays, reducing/non-reducing conditions comparison, and validation with multiple antibody clones targeting different epitopes to confirm specificity of detection.

How does the ICOS/ICOS-L structural interface inform therapeutic antibody development?

The crystal structure of the ICOS/ICOS-L complex reveals critical molecular interactions that guide therapeutic antibody development . The binding interface centers on the ICOS FG loop engaging with ICOS-L, with therapeutic antibodies mimicking these interactions . Structural characterization shows that therapeutic antibodies (STIM003 targeting ICOS and prezalumab targeting ICOS-L) employ complementarity-determining regions (CDRs) to mimic natural ligand binding . Notably, these antibodies form additional hydrogen bonds and salt bridges in peripheral regions that are solvent-accessible in the natural complex, resulting in higher binding affinities than the natural receptor-ligand interaction .

This structural mimicry exceeds that of other therapeutic antibodies targeting receptors in the same family . The elucidation of both central binding motifs and peripheral contact regions provides a molecular blueprint for designing next-generation antibodies with enhanced specificity and affinity. Understanding structural aspects also enables development of biparatopic molecules that could simultaneously target multiple co-stimulatory pathways, which has shown efficacy in transplantation models .

What methodological approaches can address challenges in ICOS detection across different T cell subsets?

Detecting ICOS expression across diverse T cell subsets presents several challenges requiring specialized methodological approaches:

  • Flow cytometric analysis optimization:

    • Use multi-parameter panels including T cell subset markers (CD4, CD8, FOXP3, etc.) alongside ICOS

    • Implement careful compensation controls when using multiple fluorochromes

    • Consider kinetic expression analysis following T cell activation (ICOS increases post-activation)

  • Single-cell analysis techniques:

    • Apply single-cell RNA sequencing to correlate ICOS transcript levels with T cell subset identities

    • Employ mass cytometry (CyTOF) for high-dimensional phenotyping without fluorescence spillover concerns

    • Utilize imaging mass cytometry for spatial context of ICOS expression in tissue microenvironments

  • Functional correlation strategies:

    • Combine ICOS detection with intracellular cytokine staining (IL-4, IL-13) to link expression with function

    • Implement phospho-flow to assess downstream signaling cascade activation in ICOS-positive cells

    • Design ICOS blocking experiments with readouts for T cell survival and differentiation markers

When comparing ICOS expression across T cell subsets, standardized stimulation protocols and careful selection of antibody clones that maintain specificity across activation states are essential for reliable cross-subset comparisons.

How can researchers effectively evaluate ICOS antibody specificity in therapeutic development contexts?

Rigorous evaluation of ICOS antibody specificity for therapeutic development requires multiple complementary approaches:

  • Competitive binding assays:

    • Assess the ability of candidate antibodies to block ICOS/ICOS-L interaction using biolayer interferometry (BLI)

    • Quantify IC50 values for inhibition of receptor-ligand binding

    • Compare binding kinetics (kon/koff rates) with natural ligand interactions

  • Epitope mapping strategies:

    • Employ X-ray crystallography to determine precise binding interfaces, as demonstrated with STIM003 and prezalumab

    • Use hydrogen-deuterium exchange mass spectrometry for solution-phase epitope mapping

    • Create alanine-scanning mutants of key interface residues to validate critical binding determinants

  • Cross-reactivity assessment:

    • Test binding to related family members (CD28, CTLA-4) to ensure specificity

    • Evaluate species cross-reactivity for translational research applications

    • Implement tissue cross-reactivity panels to identify potential off-target binding

  • Functional validation methods:

    • Assess effects on T cell activation, proliferation, and cytokine production

    • Evaluate impact on Th2 cell differentiation and regulatory T cell function

    • Determine in vivo efficacy in relevant disease models (transplantation, autoimmunity, cancer)

Understanding that therapeutic antibodies often mimic natural ligand interactions while achieving higher binding affinities through additional peripheral contacts provides critical direction for evaluation criteria .

How should researchers interpret contradictory data regarding ICOS function in different disease models?

Contradictory findings regarding ICOS function across disease models may stem from several factors that researchers should systematically evaluate:

  • Context-dependent roles - ICOS signaling effects may differ between autoimmunity, cancer, and transplantation settings . Researchers should clearly define the immunological context of their model, including baseline activation state of T cells.

  • Temporal expression dynamics - ICOS expression changes dramatically following T cell activation. Experimental timelines should be precisely reported and compared when reconciling contradictory findings.

  • Cell type specificity - While predominantly studied on conventional T cells, ICOS functions differently on regulatory T cells, NK cells, and innate lymphoid cells. Flow cytometric analyses should incorporate multiple lineage markers to distinguish effects on specific populations.

  • Genetic background influences - Mouse strain differences significantly impact ICOS expression and function. Researchers should consider genetic background when comparing results across different model systems.

  • Antibody clone variability - Different anti-ICOS antibodies target distinct epitopes that may differentially affect signaling outcomes. Experiments should specify antibody clones, concentrations, and binding characteristics when addressing contradictory findings.

When confronted with contradictory data, researchers should perform side-by-side comparisons using standardized protocols and multiple readout systems to identify experimental variables that might explain disparate results.

What strategies can overcome technical limitations in detecting low ICOS expression levels?

Detecting low ICOS expression presents technical challenges that can be addressed through several methodological refinements:

  • Signal amplification techniques:

    • Implement tyramide signal amplification for IHC/IF applications

    • Utilize biotin-streptavidin systems for enhanced sensitivity

    • Apply branched DNA amplification for in situ hybridization of ICOS transcripts

  • Enhanced sample preparation:

    • Optimize fixation protocols to preserve epitope accessibility

    • Employ extended antigen retrieval methods for FFPE tissues

    • Consider membrane permeabilization optimization for intracellular epitopes

  • Advanced detection platforms:

    • Utilize high-sensitivity flow cytometers with improved photomultiplier tubes

    • Implement spectral flow cytometry to resolve autofluorescence from true signal

    • Consider droplet digital PCR for absolute quantification of ICOS transcripts

  • Enrichment strategies:

    • Perform magnetic pre-enrichment of target populations before analysis

    • Utilize in vitro stimulation protocols to upregulate ICOS expression when appropriate

    • Consider cell sorting to isolate rare ICOS-positive populations for downstream analysis

When working with samples exhibiting low ICOS expression, researchers should include appropriate positive controls (such as activated T cells or Jurkat cells) and implement rigorous background subtraction methods to ensure detection specificity .

How can researchers effectively differentiate between ICOS signaling effects and other co-stimulatory pathways?

Distinguishing ICOS-specific signaling from other co-stimulatory pathways requires careful experimental design:

  • Genetic approaches:

    • Utilize ICOS knockout models compared with wild-type controls

    • Implement CRISPR/Cas9-mediated ICOS deletion in primary cells or cell lines

    • Consider conditional knockout systems for temporal control of ICOS expression

  • Antibody-based interventions:

    • Apply highly specific blocking antibodies targeting the ICOS/ICOS-L interaction interface

    • Include isotype controls to account for Fc receptor-mediated effects

    • Compare effects of anti-ICOS antibodies with those targeting related pathways (CD28, OX40)

  • Downstream signaling analysis:

    • Assess PI3K pathway activation, which is preferentially recruited by ICOS via its YMFM motif

    • Compare phosphorylation patterns of downstream effectors between ICOS and CD28 stimulation

    • Implement phospho-proteomic approaches to identify ICOS-specific signaling nodes

  • Combinatorial blockade experiments:

    • Design factorial experiments blocking multiple pathways individually and in combination

    • Quantify additive versus synergistic effects to determine pathway interdependence

    • Utilize mathematical modeling to deconvolute overlapping signaling networks

When interpreting results, researchers should consider that the unique structural features of ICOS, including its FDPPPF motif and CC' loop interactions, contribute to its distinct signaling properties compared to other family members .

How might structural knowledge of ICOS/ICOS-L interactions inform development of next-generation immunotherapeutics?

The detailed structural characterization of the ICOS/ICOS-L complex provides several avenues for developing novel immunotherapeutics :

  • Structure-guided antibody engineering:

    • Design antibodies that specifically target the FDPPPF motif and CC' loop interactions

    • Develop biparatopic antibodies that simultaneously engage multiple epitopes on ICOS or ICOS-L

    • Engineer antibodies with modified Fc regions to enhance or suppress effector functions based on therapeutic goals

  • Small molecule inhibitor development:

    • Target specific binding pockets identified in the crystal structure

    • Design peptidomimetics that disrupt the ICOS/ICOS-L interface

    • Develop allosteric modulators that stabilize non-binding conformations

  • Cross-reactive therapeutic approaches:

    • Create engineered proteins that target common structural features between ICOS and CD28

    • Develop loop-grafted molecules that combine binding elements from multiple co-stimulatory receptors

    • Design dual-targeting agents for simultaneous blockade of multiple pathways

  • Glycan-targeting strategies:

    • Exploit the role of N110 glycosylation in ICOS/ICOS-L binding

    • Develop glycomimetics that interfere with this interaction

    • Engineer glycosidases that specifically modify glycans critical for receptor-ligand engagement

Understanding that therapeutic antibodies can achieve higher binding affinities than natural ligands through additional peripheral contacts provides a rational basis for next-generation immunotherapeutic design strategies .

What emerging technologies might advance ICOS detection and functional characterization?

Several cutting-edge technologies hold promise for advancing ICOS research:

  • Advanced imaging modalities:

    • Implement super-resolution microscopy (STORM, PALM) to visualize ICOS microclusters

    • Apply lattice light-sheet microscopy for real-time analysis of ICOS dynamics during immune synapse formation

    • Utilize correlative light and electron microscopy to link ICOS localization with ultrastructural features

  • Single-cell multiomics:

    • Combine single-cell transcriptomics with proteomics to correlate ICOS mRNA and protein levels

    • Implement CITE-seq for simultaneous detection of ICOS surface expression and transcriptional profiles

    • Apply spatial transcriptomics to map ICOS expression within tissue microenvironments

  • Biosensor technology:

    • Develop FRET-based sensors for real-time monitoring of ICOS conformational changes

    • Create split-protein complementation assays to detect ICOS-ICOS-L interactions in living cells

    • Implement force-sensitive fluorescent proteins to measure mechanical forces during ICOS engagement

  • In situ protein modification analysis:

    • Apply proximity labeling techniques to identify ICOS interaction partners

    • Utilize glycoproteomics to comprehensively characterize ICOS glycosylation patterns

    • Implement cross-linking mass spectrometry to map structural interactions in native environments

These emerging technologies will help researchers address current knowledge gaps regarding ICOS microlocalization, temporal dynamics of signaling, and context-dependent interaction partners.

How might ICOS antibody research integrate with other immunomodulatory approaches in complex disease settings?

Integration of ICOS-targeted approaches with other immunomodulatory strategies represents a frontier in immunotherapy research:

  • Combination immunotherapy strategies:

    • Investigate synergies between ICOS modulation and PD-1/PD-L1 blockade in cancer

    • Explore sequential treatment approaches targeting different co-stimulatory/co-inhibitory pathways

    • Develop rational combination regimens based on mechanistic understanding of pathway interactions

  • Cell therapy enhancement:

    • Engineer CAR-T cells with modified ICOS signaling domains to improve persistence and function

    • Explore ex vivo modulation of ICOS pathways to optimize adoptive cell therapies

    • Develop ICOS agonist approaches to enhance antigen-specific T cell responses in vaccination

  • Biomarker development:

    • Establish ICOS expression as a predictive biomarker for response to immunotherapy

    • Develop multiplexed assays combining ICOS with other immune checkpoints to guide treatment selection

    • Investigate soluble ICOS as a potential liquid biopsy marker for monitoring immune responses

  • Precision immunotherapy approaches:

    • Stratify patients based on ICOS pathway genetics and expression patterns

    • Tailor ICOS-targeted therapies to specific disease subtypes (e.g., ICOS-high versus ICOS-low tumors)

    • Develop companion diagnostics to guide patient selection for ICOS-targeted interventions

Evidence from transplantation models showing efficacy of simultaneous ICOS and CD28 blockade demonstrates the potential of integrated approaches , suggesting that comprehensive understanding of ICOS biology will be critical for developing optimal combination strategies in various disease contexts.

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.