Biomarker Analysis Services for Stomach Cancer
Drug R&D Solutions

Biomarker Analysis Services for Stomach Cancer

Inquiry

At Alfa Cytology, we provide specialized biomarker analysis services exclusively focused on advancing drug discovery and preclinical development for Stomach Cancer research. Our comprehensive biomarker panel is designed to facilitate a deep understanding of stomach cancer pathophysiology, supporting pharmaceutical and biotechnology partners in the early stages of therapeutic development. Please note that all our services are strictly limited to research and preclinical applications; we do not offer clinical diagnostic services.

Biomarker Discovery and Identification

The foundation of effective therapeutic intervention lies in the precise discovery and identification of disease-relevant biomarkers. At Alfa Cytology, our biomarker discovery services are integral to the drug development pipeline, enabling the identification of molecular signatures associated with stomach cancer. Our approach incorporates systematic screening of candidate biomarkers using high-throughput and targeted methodologies, followed by rigorous validation to ensure biological relevance. We employ a sequential process of discovery, initial screening, and orthogonal validation to build a robust panel of research biomarkers for further study.

Multi Omics: Utilizing cutting-edge -omics technologies, Alfa Cytology integrates genomics, transcriptomics, proteomics, and other molecular profiling strategies for a comprehensive study of biological systems in stomach cancer. Our platform enables the identification of DNA alterations (mutations, amplifications), RNA expression changes (mRNA, microRNA), protein abundance, and metabolite shifts relevant to disease pathways. Through this multi-layered approach, we interrogate key oncogenic drivers, immune evasion mechanisms, and signaling cascades such as the PI3K/AKT, MET, EGFR, and FGFR pathways, generating a holistic molecular landscape for stomach cancer research.

Candidate Validation: Our candidate validation strategies are designed to establish strong associations between biomarker candidates and stomach cancer pathophysiology. We employ a combination of preliminary in vitro and in vivo screening, cross-platform verification, and functional assays to prioritize biomarkers with the highest potential impact. Criteria for promising candidates include reproducibility, specificity to disease state, relevance to known oncogenic pathways, and feasibility for downstream assay development. This enables efficient prioritization of biomarkers for further preclinical investigation.

Biomarker Assay Development and Validation

Diverse Technological Platforms: Alfa Cytology offers custom assay development leveraging a diverse array of technological platforms. Our capabilities include adaptation and optimization of analytical platforms to meet specific research requirements, encompassing immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology and imaging techniques. Each platform is selected and tailored based on biomarker type, sample matrix, and analytical goals.

Immunoassays: We develop and implement a range of immunoassays, including ELISA, chemiluminescent, and multiplex bead-based platforms, for sensitive and specific quantification of protein biomarkers.

Mass Spectrometry: Our laboratory utilizes advanced LC-MS/MS workflows for precise quantitation and characterization of proteins, peptides, and metabolites associated with stomach cancer.

Flow Cytometry: We employ flow cytometry for multiparametric analysis of cell surface and intracellular biomarkers, enabling detailed phenotyping of tumor and immune cell populations.

Molecular Diagnostics: Our molecular diagnostics capabilities encompass PCR, qPCR, digital PCR, and next-generation sequencing for the detection of genetic and transcriptomic biomarkers, including mutations, amplifications, and microRNAs.

Histopathology And Imaging: We provide histopathological evaluation and advanced imaging, including immunohistochemistry and digital pathology, to localize and quantify biomarker expression within tissue context.

Rigorous Method Validation: All analytical methods undergo rigorous validation following established preclinical research guidelines. Our validation process assesses performance characteristics such as sensitivity, specificity, linearity, precision, and reproducibility. Comprehensive quality control measures are implemented throughout, including the use of reference standards, controls, and regular calibration to ensure data integrity and reliability.

Biomarker Quantitative and Qualitative Analysis

Our quantitative analysis capabilities are designed to deliver accurate, reproducible, and high-resolution measurement of biomarker levels across diverse sample types. We employ validated protocols and robust statistical methods to ensure the reliability of quantitative data, supporting data-driven decisions in preclinical research.

Sample Analysis: We handle a wide range of sample types, including tissue, blood, serum, plasma, and other relevant biological matrices. Our sample analysis protocols are standardized for consistency and include stringent pre-analytical and analytical quality measures. All samples are processed and analyzed using validated workflows to ensure the integrity and reproducibility of results.

High Throughput Capabilities: Alfa Cytology utilizes multiplexed analytical platforms to enable high-throughput biomarker analysis, allowing simultaneous quantification of multiple targets in limited sample volumes. This approach enhances efficiency, reduces turnaround times, and conserves precious research samples, making it ideal for large-scale preclinical studies.

Key Biomarkers for Stomach Cancer Drug Development

Gene Target Biological Function Application as a Biomarker
CD274 molecule (CD274) 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 various tissue and tumor cells. CD274 binds to its receptor, programmed death-1 (PD-1), on T cells, leading to the inhibition of T cell activation, proliferation, and cytokine production. This interaction contributes to the maintenance of immune tolerance and prevention of autoimmunity, but can also be exploited by tumor cells to evade immune surveillance. CD274 (PD-L1) expression is used as a biomarker in oncology, particularly for the selection of patients for immune checkpoint inhibitor therapies targeting the PD-1/PD-L1 pathway. Immunohistochemical assessment of PD-L1 levels in tumor tissue can inform therapeutic decision-making in several cancer types, including non-small cell lung cancer, urothelial carcinoma, and melanoma. CD274 expression may also be evaluated in research settings to study tumor immune microenvironments and immune evasion mechanisms.
CEA cell adhesion molecule 5 (CEACAM5) CEA cell adhesion molecule 5 (CEACAM5), also known as carcinoembryonic antigen (CEA), is a glycoprotein belonging to the immunoglobulin superfamily. It is primarily expressed on the surface of epithelial cells in the gastrointestinal tract and plays a role in cell adhesion by mediating homophilic and heterophilic interactions. CEACAM5 participates in intercellular binding, contributing to the maintenance of tissue architecture. It is also involved in modulating immune responses and has been implicated in inhibiting anoikis, a form of apoptosis induced by cell detachment, which may facilitate cell survival in certain contexts. CEACAM5 is widely utilized as a tumor marker in clinical practice, particularly for monitoring colorectal cancer. Its expression is elevated in several malignancies, including colorectal, pancreatic, gastric, and lung cancers, compared to normal adult tissues. Measurement of CEACAM5 levels in serum is commonly used to assist in the detection of cancer recurrence, assess treatment response, and monitor disease progression. It is also used as an adjunct in the differential diagnosis of tumors of epithelial origin.
MET proto-oncogene, receptor tyrosine kinase (MET) The MET proto-oncogene encodes a receptor tyrosine kinase known as the hepatocyte growth factor receptor (HGFR). MET is primarily expressed on the surface of epithelial and some mesenchymal cells. Upon binding its ligand, hepatocyte growth factor (HGF), MET undergoes dimerization and autophosphorylation, initiating multiple downstream signaling cascades. These pathways include the PI3K-AKT, RAS-MAPK, and STAT pathways, which regulate diverse cellular processes such as proliferation, survival, motility, morphogenesis, and angiogenesis. MET signaling plays an essential role during embryonic development, tissue regeneration, and wound healing. Dysregulation of MET activity, through gene amplification, mutation, or overexpression, has been implicated in oncogenic transformation and tumor progression. MET has been investigated as a biomarker in various cancers, including non-small cell lung cancer, gastric cancer, and renal cell carcinoma. Its expression levels, gene amplification, and mutations are assessed to provide prognostic information and to guide therapeutic decisions, particularly regarding the use of MET inhibitors or targeted therapies. Detection of MET alterations may be performed using immunohistochemistry, fluorescence in situ hybridization, or next-generation sequencing. The presence of MET dysregulation is associated with tumor aggressiveness, resistance to certain therapies, and may inform eligibility for clinical trials involving MET-targeted agents.
epidermal growth factor receptor (EGFR) Epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein and member of the ErbB family of receptor tyrosine kinases. Upon binding to its ligands, such as epidermal growth factor (EGF) or transforming growth factor-alpha (TGF-α), EGFR undergoes dimerization and autophosphorylation of its intracellular tyrosine kinase domain. This initiates a cascade of downstream signaling pathways, including the RAS-RAF-MEK-ERK and PI3K-AKT pathways, which regulate cellular processes such as proliferation, differentiation, survival, and migration. EGFR is widely expressed in epithelial tissues and plays a critical role in normal development and tissue homeostasis. EGFR is utilized as a biomarker in several clinical and research contexts, particularly in oncology. In non-small cell lung cancer (NSCLC), specific activating mutations in the EGFR gene are associated with sensitivity to EGFR tyrosine kinase inhibitors. EGFR protein overexpression or gene amplification has also been observed in other malignancies, such as colorectal cancer and glioblastoma. Assessment of EGFR status can inform therapeutic decision-making, including the selection of targeted therapies, and may provide prognostic information in certain cancer types.
erb-b2 receptor tyrosine kinase 2 (ERBB2) ERBB2 (erb-b2 receptor tyrosine kinase 2), also known as HER2 or neu, encodes a member of the epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases. ERBB2 does not have a known ligand but forms heterodimers with other ligand-bound EGFR family members, leading to activation of intracellular signaling pathways such as the PI3K/AKT and MAPK pathways. These signaling cascades regulate cellular processes including proliferation, differentiation, and survival. Overexpression or amplification of ERBB2 can result in constitutive activation of these pathways, contributing to oncogenic transformation and tumor progression. ERBB2 is used as a biomarker in oncology, particularly in breast cancer, where its gene amplification or protein overexpression is assessed for diagnostic, prognostic, and therapeutic purposes. Detection of ERBB2 status helps stratify patients who may benefit from targeted therapies such as trastuzumab and other HER2-directed agents. ERBB2 testing is also applied in other cancers, including gastric and gastroesophageal junction adenocarcinomas, to guide therapeutic decision-making.
fibroblast growth factor receptor 2 (FGFR2) Fibroblast growth factor receptor 2 (FGFR2) is a transmembrane receptor tyrosine kinase that belongs to the FGFR family. It binds to fibroblast growth factors (FGFs), leading to receptor dimerization and activation of its intrinsic kinase activity. This activation initiates a cascade of downstream signaling pathways, including the RAS-MAPK, PI3K-AKT, and PLCγ pathways, which regulate cellular processes such as proliferation, differentiation, migration, and survival. FGFR2 plays a critical role in embryonic development, tissue repair, angiogenesis, and the maintenance of epithelial and mesenchymal tissues. Mutations or aberrant regulation of FGFR2 have been implicated in developmental disorders and various cancers. FGFR2 has been investigated as a biomarker in several contexts. In oncology, FGFR2 gene amplifications, mutations, and fusions have been identified in tumors such as gastric, breast, endometrial, and cholangiocarcinoma. These genomic alterations are associated with tumorigenesis and may predict sensitivity to targeted FGFR inhibitors. Additionally, FGFR2 expression levels and genetic variants have been explored in relation to cancer prognosis and risk stratification. In the context of pharmacogenomics, FGFR2 status is used to inform patient selection for clinical trials involving FGFR-targeted therapies.
microRNA 21 (MIR21) microRNA 21 (MIR21) encodes a small non-coding RNA molecule that functions primarily as a post-transcriptional regulator of gene expression. MIR21 binds to complementary sequences in the 3' untranslated regions (3' UTRs) of target messenger RNAs (mRNAs), leading to mRNA degradation or inhibition of translation. It is widely expressed in various tissues and has been shown to regulate multiple biological processes, including cell proliferation, apoptosis, differentiation, and immune responses. MIR21 is known to target tumor suppressor genes such as PTEN and PDCD4, thereby influencing pathways involved in cell survival and growth. MIR21 has been extensively studied as a biomarker in oncology and other diseases. Its expression levels are frequently elevated in a variety of solid tumors, including breast, lung, colorectal, and gastric cancers. Increased MIR21 expression in tissue, blood, or other body fluids has been associated with tumor presence, progression, and prognosis. In addition to cancer, altered MIR21 levels have been reported in cardiovascular and inflammatory diseases. Quantification of MIR21 can be utilized for disease detection, monitoring, and risk stratification in research and clinical settings.
mucin 1, cell surface associated (MUC1) Mucin 1 (MUC1) is a transmembrane glycoprotein expressed on the apical surface of most glandular epithelial cells. It plays a role in forming protective mucous barriers on epithelial surfaces and is involved in cell signaling, adhesion, and immune modulation. The extracellular domain of MUC1 is heavily glycosylated, contributing to its function in protecting epithelial cells from pathogens and mechanical damage. MUC1 also participates in intracellular signaling pathways that regulate cell proliferation, differentiation, and survival. MUC1 is utilized as a biomarker in several clinical contexts, particularly in oncology. Its overexpression and altered glycosylation patterns are frequently observed in various carcinomas, including breast, ovarian, pancreatic, and lung cancers. MUC1 can be detected in tissue samples and body fluids, and its measurement is used in disease diagnosis, prognosis, and monitoring of therapeutic response. For example, the cancer antigen 15-3 (CA15-3) assay detects circulating MUC1 fragments and is commonly used in the management of breast cancer.
phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) encodes the p110α catalytic subunit of class IA phosphatidylinositol 3-kinase (PI3K). PI3K is a lipid kinase that phosphorylates phosphatidylinositol-4,5-bisphosphate (PIP2) to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3), a second messenger involved in intracellular signaling. Activation of PI3K leads to the recruitment and activation of downstream signaling proteins, including AKT, which regulate diverse cellular processes such as cell growth, proliferation, survival, metabolism, and motility. PIK3CA is ubiquitously expressed and plays a central role in mediating signaling from receptor tyrosine kinases and G protein-coupled receptors. PIK3CA is frequently mutated in a variety of human cancers, including breast, colorectal, endometrial, and other solid tumors. The presence of activating mutations in PIK3CA can be detected in tumor tissue or, in some cases, in circulating tumor DNA. These mutations are used to stratify patients for targeted therapies, particularly PI3K inhibitors, and may provide prognostic or predictive information regarding therapeutic response. PIK3CA mutation status is incorporated into molecular profiling panels to inform clinical decision-making in oncology.
tumor protein p53 (TP53) Tumor protein p53 (TP53) encodes a transcription factor that plays a central role in regulating the cell cycle, DNA repair, apoptosis, and genomic stability. Upon cellular stress or DNA damage, p53 is stabilized and activates the transcription of target genes involved in cell cycle arrest, allowing for DNA repair or, if the damage is irreparable, inducing programmed cell death (apoptosis). p53 also contributes to senescence and inhibits angiogenesis. Its function as a tumor suppressor is critical in 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. Assessment of TP53 mutation status or protein expression can provide information relevant to tumor classification, prognosis, and, in some contexts, therapeutic response. Detection of TP53 mutations in tumor tissue or circulating DNA is utilized in research and clinical studies to stratify patients and to monitor disease progression.

Partner with Alfa Cytology to Advance Innovation

Explore Research Opportunities with Alfa Cytology. Our biomarker research services for stomach cancer leverage advanced technologies and rigorous scientific methodologies to support drug discovery and preclinical development. We offer an extensive array of analytical platforms and exploratory research capabilities, focusing on the identification, validation, and characterization of research biomarkers. Please note: The biomarkers described are research targets only and are not claimed as validated or mandatory markers. Our services are strictly limited to preclinical research stages, and we do not provide clinical diagnostic or validation claims. Alfa Cytology maintains a commitment to scientific objectivity and collaborative innovation.

We invite you to connect with Alfa Cytology to discuss exploratory biomarker research opportunities in stomach cancer. Our focus is on scientific collaboration, knowledge exchange, and advancing preclinical understanding of disease biology. Let's work together to drive innovation in biomarker discovery—contact us to explore new research possibilities.

HOW WE WORK

Make Order

Make Order

Experimental Scheme

Experimental Scheme

Implementation

Implementation

Conclusion

Conclusion
Make an Inquiry