Biomarker Analysis Services for Colon Cancer
Drug R&D Solutions

Biomarker Analysis Services for Colon Cancer

Inquiry

Alfa Cytology offers specialized biomarker analysis services dedicated to advancing Colon Cancer research and therapeutic development. Leveraging a comprehensive biomarker panel, our services are designed to elucidate the complex pathophysiology of Colon Cancer, supporting the identification and characterization of molecular targets relevant to drug discovery. All services are strictly focused on preclinical research and drug development, and do not include clinical diagnostic offerings.

Biomarker Discovery and Identification

Effective therapeutic intervention in Colon Cancer begins with the identification of robust and disease-relevant biomarkers. Alfa Cytology's biomarker discovery services provide a systematic approach to uncovering novel molecular indicators by integrating high-throughput screening and rigorous validation. Our process encompasses initial candidate identification through literature mining, in silico analyses, and experimental screening, followed by multi-stage validation to ensure relevance and reproducibility for drug development applications.

Multi Omics: Our multi-omics platform integrates cutting-edge genomics, transcriptomics, and proteomics technologies to enable comprehensive study of biological systems implicated in Colon Cancer. Through next-generation sequencing, quantitative PCR, RNA-Seq, mass spectrometry, and multiplex immunoassays, we identify and characterize biomarkers at the DNA, RNA, protein, and metabolite levels. This approach allows us to interrogate key disease pathways, including those involved in cell cycle regulation, immune modulation, angiogenesis, and DNA repair, which are central to Colon Cancer pathogenesis.

Candidate Validation: Alfa Cytology employs robust validation strategies to prioritize candidate biomarkers with strong associations to Colon Cancer pathophysiology. Preliminary screening is conducted using both in vitro and ex vivo models, with candidates evaluated for expression specificity, functional relevance, and mechanistic linkage to disease processes. Criteria for advancement include reproducibility, differential expression in disease versus control samples, and involvement in clinically actionable pathways.

Biomarker Assay Development and Validation

Diverse Technological Platforms: Our laboratory infrastructure supports custom assay development across a range of technological platforms, including immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology. We tailor platform selection and adaptation to the specific requirements of each biomarker and project, ensuring optimal sensitivity, specificity, and throughput for preclinical research applications.

Immunoassays: We offer enzyme-linked immunosorbent assays (ELISA), chemiluminescent immunoassays, and multiplex bead-based assays for quantitative protein detection.

Mass Spectrometry: Our LC-MS/MS workflows enable precise quantification and characterization of proteins, peptides, and metabolites relevant to Colon Cancer.

Flow Cytometry: High-parameter flow cytometry is utilized for the analysis of cell surface and intracellular markers, enabling single-cell resolution and population profiling.

Molecular Diagnostics: We provide PCR-based assays, next-generation sequencing, and digital PCR for the detection of gene mutations, copy number variations, and expression profiling.

Histopathology And Imaging: Advanced immunohistochemistry and digital imaging platforms support spatial analysis and quantification of biomarker expression in tissue sections.

Rigorous Method Validation: All analytical methods developed and applied at Alfa Cytology undergo rigorous validation in accordance with established regulatory guidelines. Validation parameters include specificity, sensitivity, accuracy, precision, linearity, and reproducibility. Comprehensive quality control measures are implemented at every stage to ensure data integrity and reliability for preclinical research purposes.

Biomarker Quantitative and Qualitative Analysis

Our quantitative analysis capabilities encompass absolute and relative quantification of biomarker levels in diverse biological matrices. We implement standardized calibration, normalization, and data analysis protocols to ensure robust and reproducible measurement of candidate biomarkers, supporting data-driven decision-making in Colon Cancer drug development.

Sample Analysis: Alfa Cytology processes a broad range of sample types, including tissue lysates, plasma, serum, cell lines, and formalin-fixed paraffin-embedded (FFPE) specimens. Each sample undergoes standardized preparation and analytical protocols, with strict adherence to quality control procedures to ensure sample integrity, traceability, and data accuracy throughout the analysis workflow.

High Throughput Capabilities: Our high-throughput analytical platforms enable multiplexed biomarker analysis, facilitating simultaneous evaluation of multiple targets from limited sample volumes. This approach enhances efficiency, conserves valuable specimens, and accelerates the generation of comprehensive data sets essential for preclinical Colon Cancer research.

Key Biomarkers for Colon Cancer Drug Development

Gene Target Biological Function Application as a Biomarker
CD274 molecule (CD274) The CD274 molecule, also known as programmed death-ligand 1 (PD-L1), is a transmembrane protein that plays a key role in the regulation of immune responses. It is primarily expressed on antigen-presenting cells, such as dendritic cells and macrophages, as well as on some non-hematopoietic cells and various tumor cells. CD274 binds to its receptor, programmed death-1 (PD-1), on T cells and other immune cells, leading to the inhibition of T cell activation, proliferation, and cytokine production. This interaction serves as an immune checkpoint that helps maintain self-tolerance and prevent autoimmunity by downregulating immune responses. In the context of cancer, increased expression of CD274 on tumor cells can contribute to immune evasion by suppressing anti-tumor T cell activity. CD274 (PD-L1) expression is used as a biomarker in oncology, particularly in the context of immunotherapy. Immunohistochemical assessment of CD274 levels in tumor tissue can provide information relevant to patient stratification for immune checkpoint inhibitor therapies targeting the PD-1/PD-L1 axis. Higher levels of CD274 expression in certain tumor types have been correlated with response rates to these therapies. Additionally, CD274 expression may be evaluated to inform prognosis and disease characterization in various malignancies.
KRAS proto-oncogene, GTPase (KRAS) KRAS (Kirsten rat sarcoma viral oncogene homolog) encodes a small GTPase that functions as a molecular switch in signal transduction pathways. It cycles between an active GTP-bound state and an inactive GDP-bound state, thereby regulating signaling cascades such as the MAPK/ERK and PI3K/AKT pathways. These pathways control key cellular processes including proliferation, differentiation, and survival. KRAS is activated by upstream signals from receptor tyrosine kinases and relays these signals to downstream effectors. Mutations in KRAS can lead to constitutive activation, resulting in uncontrolled cell growth and oncogenesis. KRAS is widely used as a biomarker in oncology, particularly in colorectal, lung, and pancreatic cancers. Mutation status of KRAS is assessed to inform prognosis and guide therapeutic decisions, as certain KRAS mutations are associated with resistance to specific targeted therapies, such as anti-EGFR monoclonal antibodies in colorectal cancer. Detection of KRAS mutations in tumor tissue or circulating tumor DNA is also utilized for molecular characterization of tumors.
cyclin D1 (CCND1) Cyclin D1, encoded by the CCND1 gene, is a regulatory protein that plays a critical role in cell cycle progression. It functions as a regulatory subunit of cyclin-dependent kinases CDK4 and CDK6, forming active holoenzymes that phosphorylate the retinoblastoma protein (pRB). This phosphorylation event facilitates the transition from the G1 to the S phase of the cell cycle by promoting the release of E2F transcription factors, which activate genes necessary for DNA synthesis. Cyclin D1 expression is tightly regulated by mitogenic signals, and its accumulation is a key event in the control of cell proliferation. Cyclin D1 is frequently assessed as a biomarker in various cancers, particularly in breast cancer and mantle cell lymphoma. Overexpression or gene amplification of CCND1 has been observed in several tumor types and is associated with altered cell cycle control and increased cellular proliferation. In clinical practice, cyclin D1 immunohistochemistry is used to help distinguish mantle cell lymphoma from other B-cell lymphomas and to provide prognostic information in certain cancers. Its expression profile can also aid in tumor classification and may inform therapeutic decision-making.
epidermal growth factor receptor (EGFR) The epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein and a member of the ErbB family of receptor tyrosine kinases. EGFR is activated by binding of specific ligands, such as epidermal growth factor (EGF) and transforming growth factor-alpha (TGF-α), leading to receptor dimerization and autophosphorylation of intracellular tyrosine residues. This activation initiates multiple downstream signaling pathways, including the RAS-RAF-MEK-ERK and PI3K-AKT cascades, which regulate cellular processes such as proliferation, differentiation, migration, and survival. EGFR signaling is tightly regulated under physiological conditions, and aberrant activation due to overexpression or mutations has been implicated in oncogenesis. EGFR is used as a biomarker in several clinical contexts, particularly in oncology. Its expression levels, gene amplification, and specific activating mutations are assessed in various cancers, including non-small cell lung cancer (NSCLC), colorectal cancer, and head and neck squamous cell carcinoma. EGFR status can inform prognosis, guide the selection of targeted therapies (such as tyrosine kinase inhibitors and monoclonal antibodies), and predict therapeutic response. Molecular testing for EGFR mutations is commonly performed to identify patients who may benefit from EGFR-targeted treatments.
interleukin 6 (IL6) Interleukin 6 (IL6) is a multifunctional cytokine produced by various cell types, including T cells, B cells, macrophages, endothelial cells, and fibroblasts. It plays a central role in the regulation of immune responses, inflammation, hematopoiesis, and the acute phase reaction. IL6 mediates its effects by binding to the IL6 receptor (IL6R), which then associates with the signal-transducing component gp130, leading to activation of downstream signaling pathways such as JAK/STAT, MAPK, and PI3K. IL6 stimulates the differentiation of B cells into antibody-producing plasma cells, promotes T cell proliferation and differentiation, and induces hepatic synthesis of acute-phase proteins. It also influences metabolic, regenerative, and neural processes. IL6 is frequently measured as a biomarker of inflammation and immune activation. Elevated levels of IL6 in blood or other body fluids have been observed in a variety of conditions, including infections, autoimmune diseases, and inflammatory disorders. It is also used in clinical and research settings to assess disease activity or severity in conditions such as rheumatoid arthritis, sepsis, and certain cancers. Additionally, IL6 concentrations are monitored in some cases to evaluate response to therapy targeting inflammatory pathways.
microRNA 21 (MIR21) MicroRNA 21 (MIR21) is a small non-coding RNA molecule that plays a regulatory role in gene expression at the post-transcriptional level. It primarily functions by binding to complementary sequences in the 3' untranslated regions (UTRs) of target messenger RNAs (mRNAs), leading to mRNA degradation or inhibition of translation. MIR21 has been shown to modulate various cellular processes, including cell proliferation, apoptosis, differentiation, and migration. It is involved in the regulation of multiple signaling pathways, such as the PTEN/PI3K/AKT and TGF-β pathways, by targeting genes like PTEN, PDCD4, and TPM1. MIR21 is widely expressed in various tissues and is often upregulated in response to cellular stress and injury. MIR21 has been widely studied as a potential biomarker in oncology and other disease contexts. Its expression is frequently elevated in a variety of solid tumors, including those of the lung, breast, colon, pancreas, and liver, as well as in some hematological malignancies. MIR21 can be detected in tissue samples and in body fluids such as blood and urine, which has facilitated its investigation as a non-invasive biomarker. Research has focused on its association with disease diagnosis, prognosis, and monitoring of treatment response, particularly in cancer. Additionally, altered MIR21 expression has been observed in non-malignant conditions such as cardiovascular and fibrotic diseases.
mucin 1, cell surface associated (MUC1) Mucin 1 (MUC1) is a transmembrane glycoprotein predominantly expressed on the apical surface of epithelial cells in various tissues, including the respiratory tract, gastrointestinal tract, and mammary glands. Its extracellular domain is heavily glycosylated, contributing to the formation of protective mucous barriers and playing a role in cell signaling, cell adhesion modulation, and protection against pathogens. MUC1 can interact with intracellular signaling pathways, influencing cellular proliferation, differentiation, and immune responses. MUC1 has been utilized as a biomarker in several clinical contexts, most notably in oncology. Elevated expression and altered glycosylation patterns of MUC1 have been observed in various carcinomas, including breast, ovarian, pancreatic, and lung cancers. These changes can be detected in tissue samples and, in some cases, in circulating blood, aiding in cancer diagnosis, prognosis, and monitoring. MUC1 is also the target of several immunohistochemical assays and has been explored as a target for therapeutic interventions.
mutS homolog 2 (MSH2) MutS homolog 2 (MSH2) is a key component of the DNA mismatch repair (MMR) system. It forms heterodimers primarily with MSH6 (to form MutSα) or MSH3 (to form MutSβ), which recognize and bind to base-base mismatches and insertion-deletion loops that arise during DNA replication. Upon binding to mismatched DNA, the MSH2-containing complexes initiate repair by recruiting additional MMR proteins, leading to excision of the erroneous DNA strand segment and subsequent resynthesis. This process maintains genomic stability by correcting replication errors and preventing accumulation of mutations. MSH2 is used as a biomarker in the assessment of DNA mismatch repair deficiency. Loss or reduction of MSH2 protein expression, often detected by immunohistochemistry, is observed in certain cancers, including colorectal and endometrial carcinomas. Such deficiency is associated with microsatellite instability (MSI), which has diagnostic, prognostic, and therapeutic implications. MSH2 status is also evaluated in the context of hereditary cancer syndromes, such as Lynch syndrome, where germline mutations in the MSH2 gene contribute to increased cancer risk.
tumor protein p53 (TP53) Tumor protein p53 (TP53) encodes a sequence-specific transcription factor that plays a central role in regulating the cell cycle, maintaining genomic stability, and inducing apoptosis in response to cellular stress or DNA damage. p53 activates the expression of a variety of genes involved in cell cycle arrest, DNA repair, senescence, and programmed cell death. By mediating these processes, p53 acts as a critical tumor suppressor, preventing the propagation of cells with genomic abnormalities. TP53 is widely used as a biomarker in oncology due to its frequent mutation in a variety of human cancers. Detection of TP53 mutations or abnormal p53 protein accumulation can provide information regarding tumor presence, subtype, prognosis, and potential response to therapy. Assessment of TP53 status is performed in diagnostic, prognostic, and predictive contexts across several cancer types, including breast, colorectal, lung, and ovarian cancers.
vascular endothelial growth factor A (VEGFA) Vascular endothelial growth factor A (VEGFA) is a key signaling protein involved in vasculogenesis and angiogenesis. It primarily stimulates the proliferation, migration, and survival of endothelial cells, which line the interior surface of blood vessels. VEGFA binds to specific tyrosine kinase receptors, mainly VEGFR-1 and VEGFR-2, on the surface of endothelial cells, activating downstream signaling pathways that promote the formation of new blood vessels from pre-existing vasculature. This process is essential during embryonic development, tissue repair, and in response to hypoxic conditions. Additionally, VEGFA increases vascular permeability and plays a role in inflammatory responses. VEGFA levels have been measured in various biological fluids and tissues as an indicator of angiogenic activity. Elevated VEGFA expression has been observed in multiple pathological conditions characterized by abnormal blood vessel formation, such as certain cancers, age-related macular degeneration, diabetic retinopathy, and rheumatoid arthritis. In oncology, VEGFA has been used to assess tumor angiogenesis and may provide information regarding disease progression or response to anti-angiogenic therapies. In ophthalmology, VEGFA measurement has been applied in the context of neovascular eye diseases to guide therapeutic decisions.

Partner with Alfa Cytology to Advance Innovation

Explore Research Opportunities with Alfa Cytology. Our biomarker research services and technological capabilities are designed to support exploratory and preclinical research in Colon Cancer. The biomarkers described herein are research targets only and are presented for scientific exploration; we do not claim any biomarker as validated or mandatory for Colon Cancer research or therapy. Our focus is exclusively on preclinical research stages, and we maintain a commitment to scientific objectivity and rigor throughout all projects.

We invite you to engage with Alfa Cytology for collaborative discussions on biomarker research in Colon Cancer. Our approach is exploratory, fostering scientific collaboration and knowledge exchange without making claims regarding biomarker validation or necessity. Connect with us to advance preclinical research through objective and innovative biomarker discovery.

HOW WE WORK

Make Order

Make Order

Experimental Scheme

Experimental Scheme

Implementation

Implementation

Conclusion

Conclusion
Make an Inquiry