COAD Antibody

Shipped with Ice Packs
In Stock

Description

Terminology Validation

  • The term "COAD Antibody" does not appear in PubMed, ClinicalTrials.gov, or the CAS Content Collection.

  • No entries in antibody databases such as AbDb (Antibody Structure Database) or AACDB (Antigen-Antibody Complex Database) reference this term .

  • The acronym "COAD" is not recognized by the International Nonproprietary Names (INN) system for antibodies or related biologics .

Potential Misinterpretations

  • COAD may refer to Chronic Obstructive Airway Disease, but this is unrelated to antibody nomenclature.

  • COAD could be a typographical error for:

    • COV2-2130: A monoclonal antibody targeting SARS-CoV-2 (discussed in sources 6, 8, 11) .

    • ADCs (Antibody-Drug Conjugates): A therapeutic platform described in source 10 .

    • Anti-Chemokine Antibodies: Autoantibodies linked to COVID-19 outcomes (source 6) .

Recommendations

  1. Verify the spelling or context of "COAD Antibody."

  2. Explore related antibodies with similar nomenclature:

Antibody NameTarget/FunctionSource Reference
COV2-2130SARS-CoV-2 spike protein (Class 3 RBD)
Anti-Amyloid AntibodiesAlzheimer’s disease (e.g., Aducanumab)
AZD7442Long-acting COVID-19 prophylaxis/therapy
Anti-CXCL13 AntibodiesLinked to reduced long COVID risk

Research Gaps

No peer-reviewed studies, clinical trials, or regulatory filings (e.g., FDA, EMA) mention "COAD Antibody." This suggests the term is either:

  • A proprietary compound not yet publicly disclosed.

  • A misinterpretation of established antibody nomenclature.

Next Steps

To resolve ambiguity, consider:

  • Consulting internal proprietary databases or unpublished research.

  • Reviewing patents (e.g., USPTO, WIPO) for potential matches.

  • Clarifying the target antigen or therapeutic area associated with "COAD."

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 weeks lead time (made-to-order)
Synonyms
COAD antibody; PPAT antibody; At2g18250 antibody; T30D6.24Phosphopantetheine adenylyltransferase antibody; EC 2.7.7.3 antibody; AtCoaD antibody; Dephospho-CoA pyrophosphorylase antibody; Pantetheine-phosphate adenylyltransferase antibody
Target Names
COAD
Uniprot No.

Target Background

Function
This enzyme reversibly transfers an adenylyl group from ATP to 4'-phosphopantetheine, producing dephospho-CoA (dPCoA) and pyrophosphate. It does not utilize 4'-phosphopantothenoylcysteine as a substrate.
Gene References Into Functions
This enzyme catalyzes a key regulatory step in the coenzyme A (CoA) biosynthetic pathway. This pathway is crucial for plant growth, stress resistance, and seed lipid storage. [PMID: 18621975](https://www.ncbi.nlm.nih.gov/pubmed/18621975)
Database Links

KEGG: ath:AT2G18250

STRING: 3702.AT2G18250.1

UniGene: At.40020

Protein Families
Eukaryotic CoaD family
Subcellular Location
Cytoplasm.

Q&A

What is the relationship between immune cell infiltration and COAD prognosis?

Available evidence indicates that immune cell infiltration patterns strongly correlate with cancer prognosis in colorectal adenocarcinoma. Studies analyzing data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets have demonstrated a beneficial effect of certain immune cell populations, particularly Th17 cells, on COAD prognosis . This relationship suggests that immune system activity within the tumor microenvironment plays a critical role in determining disease outcomes. Research approaches typically involve bioinformatic analysis of large genomic databases to identify correlations between immune cell presence and survival metrics, followed by experimental validation using immunohistochemistry on patient tissue samples.

Which hub genes are associated with antibody responses in COAD?

Six key hub genes have been identified that correlate with immune cell function (particularly Th17 cells) and COAD prognosis: KRT23, ULBP2, ASRGL1, SERPINA1, SCIN, and SLC28A2 . These genes appear to modulate immune responses within the tumor microenvironment, including antibody production and efficacy. When investigating these genes, researchers typically employ RNA sequencing data analysis, followed by protein-protein interaction network construction to identify functional relationships. Validation experiments often include immunohistochemical staining of tumor samples to confirm expression patterns and correlation with infiltrating immune cells.

How are antibody responses characterized in COAD research?

Antibody responses in COAD are typically characterized using immunohistochemistry on biopsy specimens, which enables classification of immune responses with high sensitivity and specificity. This approach allows researchers to visualize antibody distribution within the tumor microenvironment and quantify expression levels . Modern protocols often combine immunohistochemistry with clinical examination, laboratory tests, and genotyping to provide comprehensive characterization. For optimal results, tissue samples should be properly fixed in formalin, embedded in paraffin, and stained with appropriate antibodies using standardized detection systems like the ultraView Universal Alkaline Phosphatase Red Detection Kit or NOVADetect DAB-Substrat Kit .

How can researchers reconcile conflicting antibody expression data in COAD studies?

When researchers encounter conflicting antibody expression data in COAD studies, a multi-faceted approach is necessary. First, evaluate methodological differences between studies, including antibody specificity, sample processing protocols, and detection systems. Cohort heterogeneity must also be considered, as differences in patient demographics, tumor stage, and treatment history can significantly impact results.

A recommended approach is to perform a meta-analysis of available data using strict inclusion criteria, followed by validation experiments using multiple antibodies against the same target on a well-characterized tissue microarray. Including tissue microarrays containing multiple known positive and negative controls (as described in the immunohistochemistry literature) can help standardize results across different laboratories . Finally, complementary techniques such as mass spectroscopy-based proteomics can resolve persistent discrepancies by providing antibody-independent protein identification.

What are the most effective experimental designs for studying the role of antibodies in COAD progression?

The most effective experimental designs combine in vitro, in vivo, and clinical approaches. Begin with mechanistic studies using colorectal cancer cell lines to establish baseline antibody effects. These should be followed by patient-derived xenograft models that better recapitulate the tumor microenvironment.

For clinical studies, a prospective cohort design with longitudinal sampling offers the strongest evidence for antibody dynamics during disease progression. Tissue samples should be collected at multiple timepoints and analyzed using a combination of immunohistochemistry and multiplexed immunofluorescence to capture spatial relationships between antibodies and other immune components . Including control tissues from healthy margins and conducting parallel analyses of circulating antibodies in blood samples strengthens the experimental framework. Statistical analysis should account for patient heterogeneity using multivariate models that adjust for known prognostic factors.

How can researchers distinguish between causative and correlative relationships in antibody-COAD associations?

Distinguishing causative from correlative relationships requires rigorous experimental approaches. Initial correlative findings from bioinformatic analyses should be validated through functional studies. This typically involves antibody depletion or supplementation experiments in cell culture and animal models to establish direct effects on cancer cell behavior.

CRISPR/Cas9-mediated gene editing of antibody-producing cells or target genes can provide strong mechanistic evidence. For instance, knocking out the six hub genes identified in COAD research (KRT23, ULBP2, ASRGL1, SERPINA1, SCIN, and SLC28A2) and observing the impact on antibody production and tumor progression can help establish causality . Additionally, time-course studies tracking antibody levels before and after interventions can establish temporal relationships necessary for causal inference. Finally, mendelian randomization approaches using genetic instruments as proxies for antibody levels can help overcome confounding in observational studies.

What are the optimal protocols for antibody detection in COAD tissue samples?

Optimal protocols for antibody detection in COAD tissue samples involve a systematic approach to tissue processing and staining. Samples should be fixed in formalin and embedded in paraffin, with serial sections prepared for comprehensive analysis. Congo red staining viewed under cross-polarized light can be used for initial amyloid detection if relevant.

For immunohistochemistry, pretreatment conditions significantly impact staining quality. Using Cell Conditioning 1 or sodium citrate pretreatment (4 times, 5 minutes, 600W) optimizes antigen retrieval for most antibodies . Commercial monoclonal and polyclonal antibodies should be used at validated dilutions (ranging from 1:600 to 1:160,000 depending on the specific antibody) . Automated immunostainers such as the BenchMark XT with standardized detection kits ensure reproducibility.

Quality control measures should include on-slide positive controls (tissue microarrays containing known samples), negative controls (omission of primary antibody), and validation using specimens with confirmed antibody profiles. All antibodies should be extensively validated before use in research studies to ensure they accurately detect the proteins of interest .

How should researchers integrate antibody data with genomic and transcriptomic profiles in COAD?

Integration of antibody data with genomic and transcriptomic profiles requires a multi-omics approach. Begin by establishing standardized protocols for simultaneous extraction of proteins, RNA, and DNA from the same tissue sample to minimize variability. Spatial transcriptomics and multiplexed immunofluorescence on sequential sections can preserve spatial context when comparing genomic and antibody data.

For data integration, researchers should employ computational methods such as multi-omics factor analysis (MOFA) or similarity network fusion (SNF) to identify patterns across data types. Correlation networks between antibody levels and gene expression can reveal regulatory relationships, particularly for the six hub genes identified in COAD research . Validation of these integrative findings should include single-cell approaches that directly link gene expression to protein production within specific cell populations.

Finally, pathway enrichment analysis should be performed to contextualize findings within biological processes, potentially revealing mechanisms by which antibodies influence COAD progression through modulation of specific signaling pathways.

What statistical approaches best capture the relationship between antibody markers and COAD outcomes?

The complex relationship between antibody markers and COAD outcomes requires sophisticated statistical approaches. Cox proportional hazards models remain the foundation for survival analysis, but should be enhanced with time-dependent coefficients to account for changing antibody effects over the disease course. Competing risk analysis is essential when multiple outcome events (recurrence, metastasis, death) may occur.

Machine learning approaches, particularly regularized regression methods like LASSO or elastic net, can identify the most predictive antibody markers from high-dimensional datasets. These should be combined with unsupervised clustering to identify patient subgroups with distinct antibody profiles and outcomes. For validation, both internal (cross-validation, bootstrapping) and external (independent cohort) approaches are necessary to ensure generalizability.

Bayesian hierarchical models offer advantages when integrating multiple data types, allowing researchers to incorporate prior knowledge about antibody-gene relationships. Regardless of the approach, researchers should report comprehensive model diagnostics and effect sizes with confidence intervals rather than p-values alone, enabling better interpretation of the clinical significance of antibody markers in COAD prognosis.

How has the understanding of COAD antibody significance evolved with emerging single-cell technologies?

Single-cell technologies have revolutionized our understanding of antibody significance in COAD by revealing previously unappreciated heterogeneity in immune cell populations. Traditional bulk analysis methods often masked the diverse cellular sources of antibodies within the tumor microenvironment. Single-cell RNA sequencing now allows researchers to identify specific B cell subpopulations responsible for antibody production and characterize their spatial distribution relative to tumor cells.

These technologies have also revealed complex interactions between antibody-producing cells and other immune components, particularly Th17 cells, which have been implicated in favorable COAD prognosis . By combining single-cell transcriptomics with spatial technologies like Visium or CODEX, researchers can now map the entire antibody production network within COAD tissues, revealing localized immune responses that correlate with tumor behavior.

Furthermore, single-cell B cell receptor (BCR) sequencing has enabled tracking of clonal evolution in antibody-producing cells, providing insights into how the antibody repertoire adapts during COAD progression and in response to therapy.

What role do antibodies play in the context of immunotherapy response in COAD patients?

Antibodies play multifaceted roles in immunotherapy response among COAD patients. Endogenous antibody production can enhance or inhibit checkpoint inhibitor efficacy through various mechanisms. First, antibodies can form immune complexes that activate Fc receptor-bearing cells, promoting antigen presentation and T cell activation. Conversely, certain antibody isotypes may induce immunosuppressive signals that counteract immunotherapy effects.

The presence of specific antibodies against tumor-associated antigens before treatment initiation has been associated with improved responses to immune checkpoint inhibitors. This suggests that pre-existing humoral immunity may identify patients most likely to benefit from immunotherapy. Monitoring antibody levels and specificity during treatment can provide early indicators of response or resistance.

Recent research has also focused on how the expression of the six hub genes (KRT23, ULBP2, ASRGL1, SERPINA1, SCIN, and SLC28A2) influences antibody production and immunotherapy efficacy . Understanding these relationships may lead to new combination strategies that leverage antibody responses to enhance immunotherapy outcomes in COAD patients.

How can researchers utilize antibody signatures for COAD patient stratification?

Antibody signatures offer powerful tools for COAD patient stratification beyond traditional TNM staging. Implementation requires a systematic approach beginning with discovery cohorts to identify candidate antibody patterns, followed by validation in independent patient populations. Multiplexed assays measuring multiple antibodies simultaneously provide more robust signatures than single markers.

Prospective studies should validate these signatures in real-world clinical settings before implementation. For maximum utility, researchers should develop standardized assays that can be performed on readily available specimens (blood or routine diagnostic biopsies) and establish clear thresholds for risk categorization based on antibody profiles. Integration with the six hub genes identified in COAD research can further refine patient stratification approaches .

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.