At Protheragen, we offer specialized biomarker analysis services tailored exclusively for Lymphoma drug discovery and preclinical development. Our comprehensive biomarker panel is designed to facilitate a deep understanding of Lymphoma pathophysiology, enabling the identification and characterization of molecular targets relevant to therapeutic development. Please note that our services are strictly focused on research and preclinical stages and do not encompass clinical diagnostic applications.
The foundation of effective Lymphoma therapeutic intervention lies in the robust discovery and identification of relevant biomarkers. Protheragen’s biomarker discovery services are integral to the drug development pipeline, providing insights into disease mechanisms and facilitating the selection of promising molecular targets. Our workflow encompasses high-throughput screening of candidate biomarkers, followed by rigorous validation processes to ensure specificity, reproducibility, and relevance to Lymphoma biology. This systematic approach accelerates the transition from basic research findings to actionable targets for preclinical drug development.
Multi Omics: Leveraging cutting-edge -omics technologies, Protheragen employs genomics, transcriptomics, proteomics, and related approaches to enable a comprehensive study of biological systems in Lymphoma. Our multi-omics strategy allows for the identification and characterization of DNA, RNA, protein, and metabolite biomarkers, providing a holistic view of molecular alterations and regulatory networks. These technologies facilitate the elucidation of key disease pathways, such as those involving apoptosis regulation, immune evasion, and cell proliferation, all of which are critical in Lymphoma pathogenesis.
Candidate Validation: Our candidate validation and prioritization process utilizes a combination of experimental and computational strategies to confirm biomarker relevance to Lymphoma pathophysiology. Initial screening involves assessing the association of candidates with disease mechanisms, followed by secondary validation in relevant preclinical models. Criteria for prioritization include biological plausibility, reproducibility across sample sets, and potential translational value for therapeutic targeting. This ensures that only the most promising biomarker candidates advance in the drug discovery pipeline.
Diverse Technological Platforms: Protheragen offers custom assay development capabilities across a diverse range of technological platforms. Our laboratory infrastructure is adaptable to project-specific requirements, enabling the development and optimization of assays for novel and established biomarkers alike. Platforms include immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology and imaging modalities, ensuring flexibility and precision for diverse biomarker analysis needs.
Immunoassays: We utilize enzyme-linked immunosorbent assays (ELISA), chemiluminescent immunoassays, and multiplex immunoassays for quantitative and qualitative protein detection, supporting high sensitivity and specificity.
Mass Spectrometry: Liquid chromatography-tandem mass spectrometry (LC-MS/MS) is employed for precise quantification and characterization of peptides, proteins, and metabolites, enabling comprehensive proteomic and metabolomic profiling.
Flow Cytometry: Flow cytometry platforms are used for high-throughput, multiparametric analysis of cell surface and intracellular markers, facilitating cellular phenotyping and biomarker quantification in heterogeneous populations.
Molecular Diagnostics: We offer nucleic acid-based assays, including PCR, qPCR, and next-generation sequencing, for the detection and quantification of genetic and transcriptomic biomarkers relevant to Lymphoma.
Histopathology And Imaging: Advanced histopathology and imaging techniques, such as immunohistochemistry and digital pathology, are utilized for spatial localization and visualization of biomarker expression in tissue sections.
Rigorous Method Validation: All assay development and validation processes at Protheragen adhere to established scientific guidelines and best practices. We assess key performance characteristics including specificity, sensitivity, linearity, accuracy, precision, and reproducibility. Rigorous quality control measures are implemented at each step to ensure the reliability and robustness of analytical results, supporting high-confidence data generation for preclinical research.
Our facilities are equipped for quantitative biomarker analysis, enabling precise measurement of molecular concentrations in various biological matrices. We employ validated protocols and calibrated instruments to ensure data accuracy, with analytical outputs supporting robust statistical interpretation and data-driven decision-making in Lymphoma research.
Sample Analysis: Protheragen handles a wide range of sample types, including cell lines, primary cells, animal tissues, and biofluids (e.g., serum, plasma). Each sample undergoes standardized processing and analysis protocols tailored to the biomarker and platform in use. Stringent quality assurance procedures, including sample tracking, contamination control, and data verification, are maintained to guarantee sample integrity and analytical consistency.
High Throughput Capabilities: Our high-throughput analytical platforms support multiplexed analysis, allowing simultaneous evaluation of multiple biomarkers within the same sample. This approach enhances efficiency, conserves valuable samples, and accelerates data acquisition timelines. Multiplexing strategies are integrated across immunoassays, flow cytometry, and molecular methods to maximize throughput without compromising data quality.
| Gene Target | Biological Function | Application as a Biomarker |
|---|---|---|
| ALK receptor tyrosine kinase (ALK) | Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase belonging to the insulin receptor superfamily. ALK is primarily expressed in the developing nervous system and plays a role in neuronal differentiation, proliferation, and survival. The receptor is activated by ligand binding, leading to autophosphorylation and subsequent activation of downstream signaling pathways, including the RAS/MAPK, PI3K/AKT, and JAK/STAT cascades. In normal physiology, ALK expression is largely restricted to embryonic tissues and certain regions of the adult brain. | ALK gene rearrangements, amplifications, and mutations have been identified in several malignancies, most notably in anaplastic large cell lymphoma (ALCL), non-small cell lung cancer (NSCLC), and neuroblastoma. Detection of ALK alterations is utilized to guide targeted therapy decisions, such as the use of ALK inhibitors in NSCLC patients with ALK gene fusions. Immunohistochemistry, fluorescence in situ hybridization (FISH), and next-generation sequencing are commonly used to assess ALK status in clinical samples. |
| BCL2 apoptosis regulator (BCL2) | BCL2 (B-cell lymphoma 2) is a key regulator of apoptosis, primarily functioning as an anti-apoptotic protein. It is localized mainly to the outer mitochondrial membrane, where it inhibits the intrinsic (mitochondrial) pathway of apoptosis by binding and sequestering pro-apoptotic proteins such as BAX and BAK. This prevents mitochondrial outer membrane permeabilization and the subsequent release of cytochrome c, thereby blocking caspase activation and cell death. BCL2 plays a critical role in cellular homeostasis, immune system regulation, and tissue development by controlling cell survival. | BCL2 expression is frequently assessed in clinical and research settings as a biomarker in hematological malignancies, particularly in follicular lymphoma and other B-cell lymphomas, where its overexpression is commonly observed due to chromosomal translocations such as t(14;18). Immunohistochemical detection of BCL2 is used to aid in the diagnosis, classification, and prognostic evaluation of lymphoid neoplasms. Additionally, BCL2 expression levels have been studied in various solid tumors and are sometimes correlated with disease progression and response to therapy. |
| BCL6 transcription repressor (BCL6) | BCL6 (B-cell lymphoma 6) is a zinc finger transcriptional repressor that plays a critical role in the regulation of gene expression during B-cell development, particularly in the formation and function of germinal centers. BCL6 represses the transcription of target genes involved in cell cycle arrest, DNA damage response, and differentiation, thereby promoting the proliferation and survival of germinal center B cells. It exerts its function by binding to specific DNA sequences and recruiting corepressor complexes, such as histone deacetylases, to modify chromatin structure and inhibit gene transcription. BCL6 is also involved in the regulation of T follicular helper cell differentiation and function. | BCL6 is widely used as an immunohistochemical marker in the diagnosis and classification of lymphoid malignancies, particularly in distinguishing germinal center-derived B-cell lymphomas such as diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma. Its expression pattern can aid in subclassifying DLBCL into germinal center B-cell-like and non-germinal center B-cell-like subtypes, which may have prognostic and therapeutic implications. Additionally, BCL6 expression is assessed in the evaluation of other hematologic and non-hematologic tumors to help determine cell lineage and origin. |
| CD274 molecule (CD274) | CD274, also known as programmed death-ligand 1 (PD-L1), is a transmembrane protein that plays a critical 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 a variety of non-hematopoietic cells, including some tumor cells. CD274 binds to its receptor, programmed death-1 (PD-1), which is expressed on activated T cells and B cells. The interaction between CD274 and PD-1 transmits an inhibitory signal that reduces the proliferation of T cells, decreases cytokine production, and promotes T cell exhaustion. This mechanism contributes to the maintenance of immune homeostasis and the prevention of autoimmunity by limiting excessive immune activation. In the context of cancer, tumor cells can upregulate CD274 expression to evade immune surveillance by inhibiting antitumor T cell activity. | CD274 expression is utilized as a biomarker in several clinical contexts, particularly in oncology. Immunohistochemical assessment of CD274 (PD-L1) expression in tumor tissue is commonly used to identify patients who may benefit from immune checkpoint inhibitor therapies targeting the PD-1/PD-L1 pathway. The level of CD274 expression can inform therapeutic decisions and may be associated with response rates to these immunotherapies in certain cancers, such as non-small cell lung cancer, urothelial carcinoma, and others. Additionally, CD274 expression is studied as a prognostic and predictive biomarker in various malignancies, aiding in patient stratification and clinical trial design. |
| MYC proto-oncogene, bHLH transcription factor (MYC) | The MYC proto-oncogene, bHLH transcription factor (MYC), encodes a nuclear phosphoprotein that functions as a transcription factor. MYC regulates the expression of numerous target genes involved in cell cycle progression, apoptosis, metabolism, protein synthesis, and cellular differentiation. It forms heterodimers with MAX, another bHLH protein, to bind specific DNA sequences known as E-boxes, thereby modulating gene transcription. MYC activity is tightly controlled under normal physiological conditions, and its dysregulation can lead to increased cell proliferation and impaired differentiation. | MYC is frequently overexpressed or genomically amplified in a variety of human cancers, including lymphomas, breast cancer, and colorectal cancer. Its altered expression is associated with tumor aggressiveness, high proliferative index, and poor prognosis in several malignancies. In clinical and research contexts, MYC status—such as gene amplification, mRNA overexpression, or protein levels—has been used to characterize tumor subtypes and to stratify patients according to risk. Additionally, MYC is utilized in studies evaluating response to targeted therapies and in monitoring disease progression. |
| enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) | Enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) is a histone methyltransferase that serves as the catalytic component of the Polycomb Repressive Complex 2 (PRC2). EZH2 mediates the trimethylation of histone H3 at lysine 27 (H3K27me3), a key epigenetic modification associated with transcriptional repression. Through this activity, EZH2 plays a critical role in regulating gene expression, maintaining stem cell pluripotency, and controlling cellular differentiation. Dysregulation of EZH2 can lead to altered gene silencing and has been implicated in various developmental processes and diseases. | EZH2 expression levels and mutation status have been studied as biomarkers in several cancer types, including prostate cancer, breast cancer, and lymphomas. Elevated EZH2 expression has been correlated with tumor aggressiveness, poor prognosis, and advanced disease stage in certain malignancies. Additionally, EZH2 mutations are associated with specific subtypes of hematological cancers, such as follicular lymphoma and diffuse large B-cell lymphoma. Assessment of EZH2 can aid in disease classification, prognostication, and may inform therapeutic strategies targeting epigenetic regulators. |
| interleukin 2 receptor subunit alpha (IL2RA) | Interleukin 2 receptor subunit alpha (IL2RA), also known as CD25, is a component of the high-affinity interleukin-2 (IL-2) receptor complex. IL2RA is expressed on the surface of activated T cells, regulatory T cells (Tregs), activated B cells, and certain other immune cell types. Its primary biological function is to bind IL-2 in conjunction with the beta (CD122) and gamma (CD132) subunits, thereby facilitating IL-2-mediated signaling. This signaling promotes T cell proliferation, differentiation, and survival, and is critical for the development and maintenance of immune tolerance, particularly through the function of Tregs. | IL2RA is used as a biomarker in a variety of clinical and research contexts. Elevated levels of soluble IL2RA (sIL2RA) in serum or plasma have been associated with immune activation and are observed in several conditions, including autoimmune diseases (such as multiple sclerosis and type 1 diabetes), hematological malignancies (such as adult T-cell leukemia/lymphoma), and following organ transplantation. Measurement of IL2RA expression on cell surfaces is also employed to identify and quantify regulatory T cells in immunophenotyping studies. |
| interleukin 6 (IL6) | Interleukin 6 (IL6) is a multifunctional cytokine produced by a variety of cell types, including monocytes, fibroblasts, endothelial cells, and T cells. It plays a central role in the regulation of immune responses, inflammation, hematopoiesis, and the acute-phase reaction. IL6 stimulates the differentiation of B cells into antibody-producing plasma cells, promotes the proliferation and differentiation of T cells, and induces the production of acute-phase proteins in the liver. It is also involved in the regulation of metabolic, regenerative, and neural processes. IL6 signaling occurs through binding to its receptor complex, which activates intracellular pathways such as JAK/STAT, MAPK, and PI3K/AKT. | IL6 is widely measured as a biomarker of inflammation and immune activation. Circulating IL6 levels are frequently assessed in the context of infectious diseases, autoimmune disorders, and inflammatory conditions. Elevated IL6 concentrations have been observed in patients with sepsis, rheumatoid arthritis, COVID-19, and certain cancers. Its measurement in blood or other biological fluids can provide information about the presence and intensity of inflammatory responses and may be used to monitor disease progression or response to therapy in various clinical settings. |
| tumor necrosis factor (TNF) | Tumor necrosis factor (TNF) 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 mediating inflammation, immune system regulation, and apoptosis. It exerts its effects by binding to two distinct receptors, TNFR1 and TNFR2, which activate multiple intracellular signaling pathways, including the NF-κB and MAPK pathways. These pathways regulate the expression of genes involved in cell survival, differentiation, and inflammatory responses. TNF is involved in host defense mechanisms against infections and is also implicated in the pathogenesis of various inflammatory and autoimmune disorders. | TNF levels are measured in biological fluids such as serum, plasma, or synovial fluid to assess the presence and severity of inflammatory processes. Elevated TNF concentrations have been reported in conditions such as rheumatoid arthritis, inflammatory bowel disease, sepsis, and certain cancers. Measurement of TNF can assist in monitoring disease activity, evaluating response to anti-TNF therapies, and providing information about the inflammatory status in various clinical settings. |
| tumor protein p53 (TP53) | Tumor protein p53 (TP53) encodes a transcription factor that plays a central role in regulating the cell cycle, maintaining genomic stability, and mediating cellular responses to DNA damage and other stress signals. Upon activation by cellular stress, p53 can induce cell cycle arrest, facilitate DNA repair, initiate senescence, or trigger apoptosis, depending on the context and severity of damage. p53 achieves these effects primarily by regulating the expression of target genes involved in cell cycle control (such as CDKN1A/p21), DNA repair (such as GADD45), and apoptosis (such as BAX and PUMA). p53 activity is tightly regulated under normal conditions, mainly through interaction with its negative regulator MDM2, which targets p53 for proteasomal degradation. Loss or mutation of TP53 disrupts these protective mechanisms, often contributing to tumorigenesis. | TP53 is widely used as a biomarker in oncology. Mutations in TP53 are among the most frequent genetic alterations observed in human cancers, and their presence can aid in the diagnosis, classification, and prognostic assessment of various tumor types. Immunohistochemical detection of p53 protein accumulation is employed as a surrogate marker for TP53 mutations in tissue samples. Additionally, TP53 mutation status is used to inform risk stratification in certain malignancies and may be considered in therapeutic decision-making, as some studies have shown associations between TP53 alterations and response to specific treatments. |
Explore Research Opportunities with Protheragen. Our biomarker research services offer comprehensive analytical capabilities for the exploration and characterization of molecular targets relevant to Lymphoma drug discovery and preclinical development. Please note that all biomarkers discussed are research targets only and are not claimed as validated or mandatory for any application. Our focus is exclusively on the preclinical research stages, and we maintain scientific objectivity in all our exploratory investigations.
We invite you to engage with Protheragen for collaborative discussions on biomarker research in Lymphoma. Our services are dedicated to the exploratory investigation of molecular markers, facilitating scientific collaboration and knowledge exchange in the preclinical research space.
Make Order
Experimental Scheme
Implementation
Conclusion