Protheragen offers specialized biomarker analysis services tailored for Neuroblastoma research and therapeutic development. Our comprehensive biomarker panel is designed to advance the understanding of disease pathophysiology and support the discovery and development of novel drug candidates. All services are exclusively focused on drug discovery through preclinical development stages and do not include clinical diagnostic services. By leveraging state-of-the-art technologies and a deep understanding of Neuroblastoma biology, we enable researchers to identify, validate, and characterize biomarkers that inform target selection and therapeutic strategies.
Effective therapeutic intervention in Neuroblastoma begins with the discovery and identification of robust biomarkers. Protheragen’s biomarker discovery services form the foundation for successful drug development by systematically identifying molecular signatures associated with disease mechanisms. Our approach involves high-throughput screening of candidate genes, proteins, and pathways using advanced omics platforms, followed by rigorous validation to ensure relevance to Neuroblastoma pathogenesis. We integrate literature mining, in silico analyses, and experimental screening to generate and prioritize biomarker candidates, supporting the entire continuum from initial discovery to preclinical validation.
Multi Omics: Our multi-omics approach harnesses cutting-edge genomics, transcriptomics, proteomics, and metabolomics technologies to provide a holistic view of the biological systems underlying Neuroblastoma. By integrating data across DNA, RNA, protein, and metabolite levels, we comprehensively profile tumor biology and identify biomarkers reflective of disease state, progression, and therapeutic response. This systems biology perspective enables the elucidation of critical pathways—such as cell cycle regulation, apoptosis inhibition, and neurodevelopmental signaling—that are central to Neuroblastoma. Our multi-omics analyses uncover actionable targets and molecular phenotypes that inform preclinical drug discovery.
Candidate Validation: Protheragen employs robust validation strategies to confirm the association of candidate biomarkers with Neuroblastoma pathophysiology. Our process includes preliminary screening using in vitro and in vivo models, orthogonal validation with independent assay platforms, and bioinformatic analyses to assess specificity and functional relevance. Criteria for prioritizing promising candidates include biological plausibility, reproducibility across datasets, differential expression in disease versus normal tissues, and involvement in key oncogenic pathways. This rigorous workflow ensures that only the most relevant and actionable biomarkers advance to further preclinical development.
Diverse Technological Platforms: We offer custom assay development capabilities across a variety of technological platforms, adapting methodologies to meet specific research requirements for Neuroblastoma. Our portfolio includes immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology and imaging systems. Each platform is optimized for sensitivity, specificity, and throughput, enabling precise quantification and characterization of diverse biomarker types. Our team collaborates closely with partners to design and implement assays tailored to unique project needs.
Immunoassays: We develop and deploy ELISA, chemiluminescent, and multiplex immunoassays for quantitative detection of protein biomarkers in cell and tissue samples.
Mass Spectrometry: Our LC-MS/MS capabilities enable sensitive, high-resolution analysis of proteins, peptides, and metabolites relevant to Neuroblastoma biology.
Flow Cytometry: We utilize flow cytometry for multiparametric analysis of cell populations, including surface and intracellular biomarker expression.
Molecular Diagnostics: We offer nucleic acid-based assays, including qPCR, digital PCR, and FISH, for the detection of gene amplifications, mutations, and translocations.
Histopathology And Imaging: Our services include immunohistochemistry, immunofluorescence, and digital pathology for spatial localization and quantification of biomarkers in tissue sections.
Rigorous Method Validation: All biomarker assay methods undergo rigorous validation in accordance with established research guidelines. We assess key performance characteristics such as sensitivity, specificity, linearity, reproducibility, and robustness. Comprehensive quality control measures are implemented throughout the process, including the use of appropriate controls, calibration standards, and regular performance monitoring. This ensures the reliability and reproducibility of analytical results in preclinical research settings.
Protheragen provides advanced quantitative analysis capabilities, enabling precise measurement of biomarker abundance and dynamics in diverse biological samples. Our analytical workflows are designed to deliver high sensitivity and accuracy, supporting robust statistical interpretation and data-driven decision-making in Neuroblastoma research.
Sample Analysis: We handle a wide range of sample types, including cell lines, animal tissues, and biofluids relevant to Neuroblastoma models. Our analysis protocols encompass sample preparation, extraction, and processing steps optimized for each biomarker class and platform. Stringent quality control procedures are applied at every stage to ensure sample integrity and validity of results.
High Throughput Capabilities: Our high-throughput analytical platforms facilitate the simultaneous analysis of multiple biomarkers across large sample cohorts. Multiplexed assays and automated workflows enhance efficiency, reduce turnaround times, and conserve valuable samples. These capabilities are particularly advantageous for exploratory studies and large-scale screening projects in preclinical Neuroblastoma research.
| 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 and development. Upon ligand binding, ALK undergoes autophosphorylation, activating downstream signaling pathways such as the MAPK/ERK, PI3K/AKT, and JAK/STAT pathways, which regulate cell proliferation, survival, and differentiation. In normal adult tissues, ALK expression is limited, but aberrant activation can occur due to genetic alterations. | ALK serves as a biomarker in oncology, particularly in the context of certain cancers where ALK gene rearrangements, amplifications, or mutations are present. Notably, ALK gene fusions (such as EML4-ALK) are detected in a subset of non-small cell lung carcinomas (NSCLC), anaplastic large cell lymphomas (ALCL), and some pediatric cancers like neuroblastoma. Detection of ALK alterations can inform diagnosis, prognosis, and therapeutic decision-making, including the use of ALK-targeted tyrosine kinase inhibitors. |
| MDM2 proto-oncogene (MDM2) | The MDM2 proto-oncogene encodes an E3 ubiquitin-protein ligase that plays a critical role in the regulation of the p53 tumor suppressor pathway. MDM2 binds directly to the transactivation domain of p53, inhibiting its transcriptional activity and promoting its ubiquitination and subsequent proteasomal degradation. Through this mechanism, MDM2 controls p53 protein levels and activity, thereby influencing cell cycle progression, apoptosis, and DNA repair. MDM2 expression and activity are themselves regulated by p53 in a negative feedback loop, maintaining cellular homeostasis under normal physiological conditions. | MDM2 is used as a biomarker in oncology, particularly in the context of certain sarcomas and other malignancies. Amplification or overexpression of MDM2 can be detected in various tumor types, most notably in well-differentiated and dedifferentiated liposarcomas, where MDM2 status is used to aid in diagnosis and differential diagnosis from histologically similar lesions. MDM2 assessment is also applied in research and clinical settings to study tumor biology, prognostic implications, and potential therapeutic targeting. |
| 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 genes involved in cell proliferation, growth, apoptosis, metabolism, and differentiation. It forms heterodimers with MAX, another bHLH protein, to bind E-box sequences in DNA and activate or repress target gene transcription. MYC activity is tightly regulated in normal cells; dysregulation can lead to uncontrolled cell growth and oncogenesis. MYC is implicated in cell cycle progression, ribosome biogenesis, and modulation of cellular metabolism. | MYC is frequently used as a biomarker in oncology. Its overexpression, amplification, or translocation is observed in a variety of human cancers, including Burkitt lymphoma, breast cancer, lung cancer, and colorectal cancer. MYC status can be assessed by immunohistochemistry, fluorescence in situ hybridization (FISH), or molecular techniques, and may provide information regarding tumor subtype, prognosis, and disease progression. |
| MYCN proto-oncogene, bHLH transcription factor (MYCN) | MYCN proto-oncogene, bHLH transcription factor (MYCN), encodes a member of the MYC family of transcription factors. MYCN is characterized by a basic helix-loop-helix (bHLH) domain that enables DNA binding and dimerization with other transcription factors. It plays a critical role in the regulation of gene expression during embryonic development, particularly in the nervous system. MYCN is involved in the control of cell proliferation, differentiation, and apoptosis by regulating the transcription of target genes associated with these processes. Its expression is normally tightly regulated and predominantly observed in developing tissues. | MYCN gene amplification and/or overexpression is used as a biomarker in several cancers, most notably neuroblastoma. In neuroblastoma, MYCN amplification is associated with high-risk disease and is correlated with aggressive tumor behavior and poor prognosis. Assessment of MYCN status is incorporated into clinical risk stratification protocols for neuroblastoma and can inform treatment decisions. MYCN expression and amplification have also been studied in other tumor types, such as medulloblastoma and retinoblastoma, where they may provide prognostic information. |
| baculoviral IAP repeat containing 5 (BIRC5) | Baculoviral IAP repeat containing 5 (BIRC5), also known as survivin, is a member of the inhibitor of apoptosis (IAP) protein family. BIRC5 functions primarily to inhibit caspase activation, thereby preventing apoptotic cell death. It plays a critical role in cell division by participating in the regulation of mitosis, particularly in chromosome segregation and cytokinesis. BIRC5 is typically expressed during embryonic development and is largely absent in most differentiated adult tissues, but is often re-expressed in various malignancies. | BIRC5 is frequently overexpressed in a wide range of human cancers, including lung, breast, colorectal, and liver cancers. Its elevated expression has been associated with tumor progression, poor prognosis, and resistance to therapy. BIRC5 has been investigated as a biomarker for cancer diagnosis, prognosis, and as a potential indicator of therapeutic response. Its differential expression between tumor and most normal tissues underlies its application in cancer biomarker studies. |
| cyclin dependent kinase 4 (CDK4) | Cyclin dependent kinase 4 (CDK4) is a serine/threonine protein kinase that plays a central role in regulating the cell cycle. CDK4 forms an active complex with D-type cyclins (primarily cyclin D1, D2, or D3), which phosphorylates the retinoblastoma protein (pRB). This phosphorylation leads to the release of E2F transcription factors, enabling the transcription of genes necessary for progression from the G1 to the S phase of the cell cycle. CDK4 activity is tightly controlled by cyclin-dependent kinase inhibitors such as p16INK4a. Dysregulation of CDK4 activity can result in uncontrolled cell proliferation. | CDK4 has been studied as a biomarker in various malignancies, including melanoma, sarcomas, and certain carcinomas. Overexpression, amplification, or altered activity of CDK4 has been observed in several tumor types and is associated with cell cycle dysregulation. Immunohistochemical detection of CDK4 protein expression, as well as analysis of gene amplification or mutation, has been utilized in research and clinical settings to aid in tumor classification, prognosis, and to identify patients who may benefit from targeted therapies involving CDK4/6 inhibitors. |
| neurotrophic receptor tyrosine kinase 1 (NTRK1) | Neurotrophic receptor tyrosine kinase 1 (NTRK1), also known as TrkA, is a member of the neurotrophic tyrosine kinase receptor family. It encodes a transmembrane receptor that binds nerve growth factor (NGF) with high affinity. Upon ligand binding, NTRK1 undergoes autophosphorylation and activates several downstream signaling pathways, including the MAPK/ERK, PI3K/AKT, and PLCγ pathways. These cascades regulate neuronal differentiation, survival, and proliferation. NTRK1 is essential for the development and function of the nervous system, particularly in the differentiation and maintenance of sympathetic and sensory neurons. | NTRK1 gene fusions and rearrangements have been identified in a variety of tumor types, including certain sarcomas, thyroid carcinomas, and secretory carcinomas of the salivary gland. The presence of NTRK1 gene fusions can be used as a diagnostic biomarker to identify tumors that may respond to targeted therapies such as TRK inhibitors. Additionally, detection of NTRK1 alterations may provide prognostic information in specific cancer contexts and aid in guiding therapeutic decision-making. |
| paired like homeobox 2B (PHOX2B) | Paired like homeobox 2B (PHOX2B) is a transcription factor that plays a critical role in the development of the autonomic nervous system. It is involved in the differentiation and maintenance of noradrenergic neurons and is essential for the proper formation of neural crest-derived structures. PHOX2B regulates the expression of genes necessary for neuronal specification and survival, and its activity is crucial during embryogenesis for the development of structures such as the sympathetic ganglia, enteric nervous system, and certain cranial nerves. | PHOX2B is utilized as a biomarker in the diagnosis and characterization of neuroblastoma and other tumors of neural crest origin, such as ganglioneuroblastoma and ganglioneuroma. Its expression is also assessed in the context of congenital central hypoventilation syndrome (CCHS), where mutations in the PHOX2B gene are detected for diagnostic purposes. Immunohistochemical detection of PHOX2B protein is commonly used to confirm neural crest lineage in tumor samples. |
| solute carrier family 6 member 2 (SLC6A2) | SLC6A2 encodes the norepinephrine transporter (NET), a membrane protein primarily responsible for the reuptake of the neurotransmitter norepinephrine from the synaptic cleft into presynaptic neurons. This transporter plays a key role in regulating noradrenergic signaling in the central and peripheral nervous systems by terminating the action of released norepinephrine and recycling it for subsequent neurotransmission. SLC6A2 is expressed predominantly in noradrenergic neurons and is essential for maintaining the homeostasis of norepinephrine, thereby influencing processes such as attention, arousal, mood, and cardiovascular function. | SLC6A2 expression levels and genetic variants have been investigated as biomarkers in neuropsychiatric and cardiovascular disorders. Alterations in SLC6A2 have been associated with conditions such as depression, attention-deficit/hyperactivity disorder (ADHD), and orthostatic intolerance. Measurement of SLC6A2 gene expression, protein levels, or functional activity has been used in research settings to explore disease mechanisms, stratify patient populations, and monitor responses to therapeutic interventions targeting noradrenergic signaling. |
| somatostatin receptor 2 (SSTR2) | Somatostatin receptor 2 (SSTR2) is a G protein-coupled receptor that binds the peptide hormone somatostatin. Upon ligand binding, SSTR2 mediates inhibition of adenylyl cyclase activity, leading to decreased intracellular cAMP levels. This signaling cascade results in the inhibition of hormone secretion, cell proliferation, and neurotransmitter release in various tissues. SSTR2 is expressed in multiple organs, including the brain, pancreas, and gastrointestinal tract, and plays a key role in regulating endocrine and neuroendocrine functions. | SSTR2 is used as a biomarker for the detection and characterization of neuroendocrine tumors (NETs), as these tumors frequently overexpress this receptor. Immunohistochemical staining for SSTR2 can aid in tumor classification. Additionally, SSTR2 expression is exploited in molecular imaging techniques, such as somatostatin receptor scintigraphy and positron emission tomography (PET) using radiolabeled somatostatin analogs, to localize and assess NETs. SSTR2 status is also considered in determining eligibility for somatostatin analog-based therapies. |
Explore Research Opportunities with Protheragen. Our biomarker research services for Neuroblastoma leverage advanced analytical platforms and a multi-omics approach to support preclinical drug discovery and development. All biomarkers discussed are research targets only; we do not claim any biomarker as validated or mandatory for any application. Our work is exclusively focused on exploratory, preclinical research stages, maintaining scientific objectivity and rigor. We invite partners to collaborate in advancing the understanding of Neuroblastoma biology and therapeutic innovation.
We welcome discussions about exploratory biomarker research for Neuroblastoma. If you are interested in scientific collaboration, knowledge exchange, or learning more about our preclinical biomarker analysis capabilities, please contact us. Let’s advance Neuroblastoma research together through objective and innovative approaches.
Make Order
Experimental Scheme
Implementation
Conclusion