At Alfa Cytology, we provide specialized biomarker analysis services exclusively focused on supporting drug discovery and preclinical development for Liver Cancer research. Our comprehensive biomarker panel is designed to advance the understanding of disease pathophysiology, enabling the identification and characterization of molecular targets relevant to therapeutic development. Please note that all our services are research-oriented and are not intended for clinical diagnostics or patient management.
The foundation of effective therapeutic intervention lies in the discovery and identification of robust biomarkers that elucidate disease mechanisms and therapeutic response. Alfa Cytology offers a streamlined biomarker discovery service that integrates high-throughput screening with sophisticated validation protocols. Our process encompasses the systematic identification of candidate biomarkers, followed by rigorous screening and orthogonal validation to ensure relevance to Liver Cancer biology. This approach accelerates the translation of molecular insights into actionable research targets for drug development.
Multi Omics: Leveraging state-of-the-art -omics technologies, Alfa Cytology conducts integrated genomics, transcriptomics, proteomics, and metabolomics analyses to provide a comprehensive view of biological systems implicated in Liver Cancer. Our multi-omics approach enables the identification and quantification of DNA, RNA, protein, and metabolite biomarkers, facilitating the exploration of key pathways such as Wnt/beta-catenin signaling, immune modulation, and metabolic regulation. Through this holistic methodology, we uncover molecular signatures and networks critical to Liver Cancer pathogenesis and therapy response.
Candidate Validation: Our candidate validation strategy combines advanced molecular profiling with functional assays to establish associations between candidate biomarkers and Liver Cancer pathophysiology. Preliminary screening involves quantitative and qualitative assessment of biomarker expression, mutation status, and pathway activation in relevant preclinical models. Promising candidates are prioritized based on criteria including specificity, biological relevance, detection feasibility, and potential druggability, ensuring that only the most informative biomarkers progress to further development.
Diverse Technological Platforms: Alfa Cytology offers custom assay development tailored to the specific requirements of Liver Cancer biomarker research. Our technological platforms are adaptable, supporting a wide range of analytical modalities including immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology. This versatility allows us to design and optimize assays for diverse biomarker types and research objectives.
Immunoassays: We employ ELISA, chemiluminescent immunoassays, and multiplexed bead-based platforms for the sensitive and quantitative detection of protein biomarkers.
Mass Spectrometry: Our LC-MS/MS workflows enable precise quantification and characterization of proteins, peptides, and metabolites relevant to Liver Cancer.
Flow Cytometry: Flow cytometry assays are utilized for the analysis of cell-surface and intracellular biomarkers, as well as for immunophenotyping in preclinical samples.
Molecular Diagnostics: We implement PCR-based and next-generation sequencing methods for the detection of gene mutations, copy number variations, and expression changes.
Histopathology Imaging: Advanced histopathological techniques and digital imaging are used to localize and quantify biomarkers within tissue architecture, supporting spatial analysis and co-localization studies.
Rigorous Method Validation: All analytical methods undergo a rigorous validation process in accordance with established research guidelines. We assess key performance characteristics including sensitivity, specificity, linearity, accuracy, precision, and reproducibility. Comprehensive quality control measures are implemented throughout assay development and execution to ensure data integrity and reliability for preclinical research.
Our platform supports robust quantitative analysis of biomarker levels across multiple sample types. We employ validated calibration standards, internal controls, and digital quantification tools to ensure accurate measurement and comparability of results. Data analysis pipelines are tailored to support high-dimensional datasets, facilitating in-depth interpretation of biomarker dynamics in Liver Cancer models.
Sample Analysis: We handle a wide variety of preclinical sample types, including cell lines, tissue lysates, plasma, serum, and other biological matrices relevant to Liver Cancer research. Our analysis protocols incorporate standardized sample processing, stringent quality assessment, and contamination control to maximize data quality and reproducibility.
High Throughput Capabilities: Alfa Cytology utilizes multiplexed analytical platforms and automation to enable high-throughput biomarker analysis. This approach increases efficiency, reduces turnaround times, and conserves valuable samples by allowing simultaneous measurement of multiple biomarkers from minimal input material. Our scalable workflows are designed to support both exploratory studies and larger preclinical screening campaigns.
| 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. CD274 is expressed on various cell types, including antigen-presenting cells, some epithelial cells, and many tumor cells. Its primary biological function involves binding to the programmed cell death protein 1 (PD-1) receptor on T cells and other immune cells. This interaction leads to the inhibition of T cell activation and proliferation, promoting immune tolerance and preventing autoimmunity. In the context of cancer, tumor cell expression of CD274 can contribute to immune evasion by suppressing anti-tumor immune responses. | CD274 (PD-L1) expression is used as a biomarker in oncology, particularly in the context of immunotherapy. Its expression levels on tumor cells or within the tumor microenvironment are assessed to help identify patients who may benefit from immune checkpoint inhibitor therapies targeting the PD-1/PD-L1 pathway. Immunohistochemistry is commonly employed to detect CD274 expression in tumor tissue samples. CD274 expression has been studied as a predictive biomarker for response to therapies such as anti-PD-1 and anti-PD-L1 monoclonal antibodies in various cancers, including non-small cell lung cancer, melanoma, and urothelial carcinoma. |
| alpha fetoprotein (AFP) | Alpha fetoprotein (AFP) is a glycoprotein primarily produced by the fetal liver, yolk sac, and, to a lesser extent, the gastrointestinal tract during embryonic development. In the fetus, AFP functions as a plasma transport protein, binding and transporting various ligands such as bilirubin, fatty acids, and steroids. AFP is structurally related to serum albumin and is present at high concentrations in fetal serum, with levels declining rapidly after birth as albumin synthesis increases. In healthy adults, AFP is typically present at very low concentrations. | AFP is widely used as a biomarker in clinical practice, particularly for the detection and monitoring of hepatocellular carcinoma (HCC) and certain germ cell tumors, such as non-seminomatous testicular cancer. Elevated serum AFP levels can also be observed in chronic liver diseases, including hepatitis and cirrhosis, as well as in some benign and malignant conditions. In prenatal screening, maternal serum AFP measurement is utilized as part of assessments for fetal neural tube defects and certain chromosomal abnormalities. Interpretation of AFP levels requires consideration of clinical context and other diagnostic findings. |
| catenin beta 1 (CTNNB1) | Catenin beta 1 (CTNNB1) encodes beta-catenin, a multifunctional protein that plays a central role in cell-cell adhesion and the regulation of gene transcription. Beta-catenin is a key component of the cadherin protein complex at the plasma membrane, where it links cadherins to the actin cytoskeleton, thereby maintaining adherens junctions and tissue architecture. In addition, beta-catenin functions as a principal effector in the canonical Wnt signaling pathway. In the absence of Wnt signals, beta-catenin is targeted for degradation by a destruction complex. Upon Wnt activation, beta-catenin accumulates in the cytoplasm and translocates to the nucleus, where it interacts with TCF/LEF transcription factors to regulate genes involved in cell proliferation, differentiation, and survival. | Alterations in CTNNB1, including mutations and abnormal protein accumulation, have been observed in various types of cancer, such as hepatocellular carcinoma, colorectal cancer, and endometrial carcinoma. CTNNB1 mutations, particularly those affecting exon 3, can lead to constitutive activation of the Wnt/beta-catenin pathway. Immunohistochemical detection of nuclear or cytoplasmic beta-catenin has been used to support the diagnosis and classification of certain tumors. Additionally, CTNNB1 mutation status may provide information relevant to tumor characterization and prognosis in specific cancer types. |
| fibronectin 1 (FN1) | Fibronectin 1 (FN1) encodes a high-molecular weight glycoprotein that is a major component of the extracellular matrix. FN1 is involved in cell adhesion, migration, growth, and differentiation. It interacts with integrins and other extracellular matrix components such as collagen, fibrin, and heparan sulfate proteoglycans. FN1 plays a critical role in wound healing, embryonic development, and maintenance of tissue architecture. Its functions are mediated through both soluble plasma fibronectin and insoluble cellular fibronectin, which participate in diverse physiological and structural processes. | FN1 has been studied as a biomarker in various pathological conditions, including cancer, fibrosis, and cardiovascular diseases. Altered expression or deposition of FN1 has been observed in tumor tissues and in the circulation of patients with certain malignancies, where it is associated with tumor progression and metastasis. In addition, elevated FN1 levels have been reported in fibrotic diseases and in some inflammatory conditions. Quantification of FN1 in biological samples has been used in research settings to assess disease presence, progression, or response to therapy. |
| interleukin 6 (IL6) | Interleukin 6 (IL6) is a multifunctional cytokine produced by various cell types, including T cells, B cells, macrophages, fibroblasts, endothelial cells, and others. IL6 plays a central role in the regulation of immune responses, acute phase reactions, hematopoiesis, and inflammation. It is involved in the stimulation of B cell differentiation, promotion of T cell proliferation and differentiation, and induction of hepatic acute-phase protein synthesis. IL6 can also modulate metabolic, regenerative, and neural processes. Its signaling occurs through binding to the IL6 receptor (IL6R) and subsequent activation of the JAK/STAT pathway, among others. | IL6 is widely used as a biomarker for inflammation and immune activation. Elevated levels of IL6 in blood or other biological fluids have been associated with a variety of clinical conditions, including infection, sepsis, autoimmune disorders, and certain cancers. IL6 measurements are utilized to assess the severity and progression of inflammatory diseases, as well as to monitor response to therapy in specific clinical contexts. |
| mechanistic target of rapamycin kinase (MTOR) | The mechanistic target of rapamycin kinase (MTOR) is a serine/threonine protein kinase that functions as a central regulator of cell growth, proliferation, metabolism, and survival. MTOR integrates signals from nutrients, growth factors, energy status, and cellular stress to modulate key cellular processes such as protein synthesis, autophagy, lipid biosynthesis, and cytoskeletal organization. MTOR exists in two distinct multiprotein complexes: mTORC1 and mTORC2, each with unique components and substrate specificities. mTORC1 primarily regulates protein synthesis and autophagy in response to nutrient and energy cues, while mTORC2 is involved in the regulation of the actin cytoskeleton and cell survival pathways. | MTOR has been investigated as a biomarker in various contexts, including oncology and metabolic diseases. Alterations in MTOR expression levels, activity, or pathway activation status have been associated with tumorigenesis, cancer progression, and therapeutic response in several cancer types. Additionally, MTOR pathway dysregulation has been studied in relation to metabolic disorders such as type 2 diabetes and obesity. Assessment of MTOR or its downstream effectors in tissue samples, blood, or other biological fluids has been used in research settings to evaluate disease state, prognosis, and potential response to targeted therapies. |
| microRNA 122 (MIR122) | microRNA 122 (MIR122) is a liver-enriched, small non-coding RNA that plays a critical role in the regulation of gene expression at the post-transcriptional level. It is highly abundant in hepatocytes and is involved in various physiological processes, including lipid metabolism, cholesterol homeostasis, and the modulation of hepatic cell differentiation. MIR122 regulates the stability and translation of target messenger RNAs by binding to complementary sequences, thereby influencing pathways such as fatty acid metabolism and the cell cycle. It is also implicated in the replication of certain viruses, such as hepatitis C virus, by interacting with viral RNA and host factors. | MIR122 has been extensively studied as a biomarker for liver injury and disease. Its high specificity and abundance in the liver result in elevated levels of MIR122 in the bloodstream following hepatocellular damage, such as that caused by drug-induced liver injury, viral hepatitis, or non-alcoholic fatty liver disease. Measurement of circulating MIR122 in plasma or serum has been used in research to detect and monitor liver injury, assess disease severity, and evaluate therapeutic responses. Its performance as a biomarker has been compared to traditional liver enzymes, with studies indicating that MIR122 may reflect hepatocellular damage with high sensitivity and specificity. |
| microRNA 21 (MIR21) | MicroRNA 21 (MIR21) is a small, non-coding RNA molecule that functions primarily as a post-transcriptional regulator of gene expression. It binds to complementary sequences on target messenger RNAs (mRNAs), leading to mRNA degradation or translational repression. MIR21 is widely expressed in various tissues and is known to regulate multiple cellular processes, including proliferation, apoptosis, differentiation, and migration. Its activity has been associated with the modulation of signaling pathways such as the PI3K/AKT and TGF-β pathways, often through the targeting of tumor suppressor genes like PTEN and PDCD4. | MIR21 has been investigated as a biomarker in a variety of pathological conditions, most notably in oncology. Its elevated expression has been frequently reported in tumor tissues, blood, and other body fluids from patients with several types of cancer, including breast, lung, colorectal, and gastric cancers. MIR21 levels have been studied for their potential utility in cancer diagnosis, prognosis, and monitoring of disease progression or therapeutic response. Additionally, MIR21 has been explored as a biomarker in non-malignant diseases, such as cardiovascular and inflammatory disorders. |
| mucin 1, cell surface associated (MUC1) | Mucin 1 (MUC1) is a transmembrane glycoprotein that is predominantly expressed on the apical surface of epithelial cells in various tissues, including the respiratory tract, gastrointestinal tract, and reproductive organs. MUC1 plays a key role in forming a protective mucous barrier, contributing to lubrication and defense against pathogens. Its extracellular domain is heavily glycosylated, which aids in shielding epithelial cells from physical and chemical injury. MUC1 is also involved in intracellular signaling pathways that regulate cell growth, differentiation, and immune responses. Alterations in its expression and glycosylation patterns are frequently observed in pathological conditions, such as inflammation and malignancy. | MUC1 has been utilized as a biomarker in several clinical contexts, most notably in oncology. Elevated expression and aberrant glycosylation of MUC1 are characteristic of various carcinomas, including breast, ovarian, pancreatic, and lung cancers. MUC1 is detected in tissue samples and body fluids, and its presence or increased levels have been associated with tumor diagnosis, prognosis, and disease monitoring. For example, the CA 15-3 and CA 27.29 assays, which measure circulating MUC1 fragments, are used in the management of breast cancer. Additionally, MUC1 is being explored as a target for therapeutic interventions and as a marker for monitoring treatment response. |
| tumor necrosis factor (TNF) | Tumor necrosis factor (TNF), also known as TNF-alpha, is a pro-inflammatory cytokine primarily produced by activated macrophages, as well as other immune cells such as T lymphocytes and natural killer cells. TNF plays a central role in the regulation of immune responses, inflammation, and apoptosis. It exerts its effects by binding to TNF receptors (TNFR1 and TNFR2) on the surface of various cell types, leading to the activation of signaling pathways such as NF-κB and MAPK. These pathways mediate the expression of genes involved in inflammation, cell survival, and cell death. TNF is a key mediator in the acute phase response to infection and injury, promoting the recruitment of immune cells to sites of inflammation and inducing the expression of other cytokines and adhesion molecules. Dysregulation of TNF production or signaling has been implicated in the pathogenesis of several inflammatory and autoimmune diseases. | TNF levels in blood, tissue, or other biological samples are commonly measured as an indicator of inflammatory activity. Elevated TNF concentrations have been observed in a range of conditions, including rheumatoid arthritis, inflammatory bowel disease, sepsis, and certain cancers. Measurement of TNF can aid in the assessment of disease activity, monitoring of therapeutic response, and evaluation of inflammatory status in clinical and research settings. TNF is also used in studies investigating the mechanisms of inflammation and the effects of anti-TNF therapies. |
Explore Research Opportunities with Alfa Cytology. Our biomarker research services for Liver Cancer offer comprehensive capabilities in discovery, validation, and assay development, supporting exploratory and preclinical research. All biomarkers discussed are research targets only, provided for scientific investigation without claims of validation or clinical necessity. Alfa Cytology does not endorse any biomarker as mandatory or validated for Liver Cancer. Our services are strictly limited to preclinical research stages, ensuring scientific objectivity and flexibility for your therapeutic development programs.
We invite you to engage with Alfa Cytology to discuss collaborative biomarker research opportunities in Liver Cancer. Our focus is on scientific exploration and knowledge exchange, supporting the advancement of preclinical research through objective and rigorous analysis. Connect with us to explore how our expertise can enhance your research initiatives.
Make Order
Experimental Scheme
Implementation
Conclusion