Introduction to Biomarkers
Biomarkers are measurable indicators of biological processes in health, disease and drug treatment. Biomedical research and clinical practice rely on objective measurements to assess health status, disease progression and treatment outcomes. The results of biomarker detection tests are used to guide patient stratification, therapy selection and treatment design, empowering molecular diagnostics, personalized medicine and precision medicine. Biomarkers are equally indispensable for companion diagnostics by helping clinicians tailor therapies to specific patient populations, particularly in oncology and rare diseases.
Depending on their function and role, biomarkers can be divided into several classes, summarized below.
The importance of biomarkers has encouraged pharmaceutical companies to construct biomarker development pipelines. These pipelines start with translational research and cell biology to identify a potential indicator as a candidate biomarker, followed by validation in preclinical and clinical studies before getting qualified for routine use.
Rationale for Biomarker Discovery and Development
Role in Modern Healthcare and Research
Biomarkers can be considered the bridge between disease research and drug discovery and development. By revealing the hallmarks of disease mechanisms, they help researchers identify therapeutic targets and guide drug design, dose selection and safety assessments. Biomarker discovery informs the development of targeted therapies in oncology, immunology and rare diseases, among many other fields.¹
Alongside therapy design, biomarkers serve as functional diagnostics and precision medicine tools by translating disease-associated cellular processes into measurable clinical tools. Clinical implementation of validated biomarkers is essential for early and accurate diagnosis, as well as personalized treatment and patient care.²
Regulatory agencies, such as the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), oversee the validation of biomarkers and provide guidelines for their qualification and use in clinical trials. These frameworks ensure scientific rigor, reproducibility and clinical utility in newly developed biomarkers.
Phases in Biomarker Discovery and Development
Stepwise Process
A typical biomarker development pipeline consists of the following steps:
-
Hypothesis generation and candidate biomarker identification: Mechanistic and data-driven approaches, including genomics, proteomics, metabolomics or imaging, are used to identify potential biomarkers. Candidate biomarkers are correlated to disease pathways, therapeutic targets or measurable clinical outcomes.
-
Assay development and analytical validation: Assays are developed to accurately detect and quantify the biomarker. Analytical validation evaluates assay parameters such as sensitivity, specificity, reproducibility and robustness.
-
Biomarker validation: The scientific relevance of the candidate biomarker is evaluated in three steps.
- Content validity reveals how well the biomarker measures the biological process it is meant to measure
- Construct validity evaluates its association with the underlying disease mechanism.
- Criterion validity assesses its correlation with established clinical outcomes.
-
Evaluation of clinical utility: Clinical trials help determine whether the biomarker improves diagnosis, prognosis or therapeutic decision-making compared to existing standards.
-
Biomarker qualification and regulatory readiness: The innovator prepares an application that includes evidence of analytical validity and clinical utility in documentation formats set out by regulatory agencies. Adhering to regulatory requirements ensures a prompt approval and delivery to the market.
Regulatory considerations
Regulatory agencies in the US (FDA) and Europe (EMA) determine the criteria biomarkers need to fulfil for approval.
FDA requires detailed documentation, including assay validation data, clinical trial evidence and proof of clinical significance. It also provides guidance for the FDA Biomarker Qualification Program (BQP) and Evidentiary Framework. Similarly, EMA requires submitting a Qualification Dossier containing analytical and clinical validity and utility evidence. It follows the Qualification of Novel Methodologies (QoNM) process to run assessments. Both agencies may issue either scientific advice or a qualification opinion following evaluation.
Increasing emphasis is placed on regulatory harmonization between the FDA and EMA to facilitate global biomarker adoption. Parallel consultation with both agencies ensures the worldwide accessibility of biomarkers and biomarker-driven therapies.
Technologies and Methodologies in Biomarker Discovery
A plethora of technologies, platforms and assays facilitates biomarker discovery.
Omics Technologies
Omics technologies provide comprehensive, system-level insights into biomolecular processes while uncovering mutations or variations that underlie disease-associated cellular behavior and phenotype. Furthermore, integrative approaches combine multiple omics datasets to improve biomarker accuracy and reproducibility, improving biomarkers' predictive or prognostic value and clinical relevance.³
Proteins constitute the majority of biomarkers as integral elements of essential biological pathways. A typical proteomics-based biomarker discovery workflow involves sample collection, protein extraction, digestion, peptide separation and analysis by mass spectrometry or affinity-based techniques. Candidate protein biomarkers are identified through differential expression analysis, where fold changes reveal overexpression or underexpression patterns.¹
Analytical and Discovery Platforms
Biomarker discovery platforms enable rapid identification of several markers with great precision. While next-generation sequencing (NGS) and mass spectrometry (MS) are helpful in omics profiling, high-throughput screening (HTS) is essential in the rapid validation of the function and clinical relevance of candidate biomarkers.⁴ Liquid biopsy is another powerful method for biomarker discovery. It involves a minimally invasive approach to analyzing the macromolecule content in blood and biofluid samples.⁵
Assay Design and Optimization
Reliable biomarker discovery requires standardized sample preparation protocols to minimize pre-analytical variability. Therefore, researchers must choose appropriate extraction, storage, fractionation and enrichment methods. Additionally, assay optimization is required to improve sensitivity and specificity, ensuring the detection of low-abundance biomarkers and the separation of true signals from background noise.
Best Practices and Validation Strategies
Methodologies for Validation
Preclinical and clinical biomarker validation is necessary to ensure reliability and clinical relevance. Validation is undertaken in three areas:⁶
- Content validity ensures the biomarker measures the intended biological or clinical concept.
- Construct validity confirms that the biomarker accurately reflects underlying disease mechanisms or therapeutic effects.
- Criterion validity evaluates correlation with established clinical endpoints or gold-standard measures.
Clinical trials must consider additional study design factors for a seamless validation. Randomization reduces systematic differences between experimental groups. Blinding minimizes observer bias during sample analysis and outcome assessment. Furthermore, clinical researchers must implement controls for confounders and pre-analytical variability to prevent false biomarker-to-outcome correlations. Thus, they ensure that observed biomarker signals truly reflect biological phenomena rather than methodological artifacts.⁷
Performance Evaluation
Biomarker performance is quantitatively assessed using statistical metrics.
- Sensitivity measures the ability to identify true positives.
- Specificity measures the ability to identify true negatives.
- Area Under the Receiver Operating Characteristic Curve (AUC-ROC) depicts the power of a biomarker detection method to distinguish between true and false positives. Other metrics, such as positive predictive value (PPV), negative predictive value (NPV) and likelihood ratios, further contextualize clinical utility.⁸
Data Analysis and Computational Approaches
Role of Data Science
Biomarker research generates a high volume of complex data that can only be interpreted through bioinformatics and biostatistics. Bioinformatics pipelines involve preprocessing, normalizing and integrating multi-omics datasets, while biostatistical methods feature hypothesis testing and correlation analysis to measure false discovery rates. Together, these approaches ensure rigor, reproducibility and biological interpretability in biomarker research.⁹
AI and Machine Learning
Machine learning (ML) algorithms are widely used for feature selection, classification and prediction of disease states. In addition, deep learning and neural networks can aid the analysis of images, sequences and multimodal datasets. Together, these technologies help researchers identify novel biomarkers, predict therapeutic response and elicit novel disease subtypes from heterogeneous datasets.¹⁰'¹¹
Tools and Infrastructure
Bioinformatics tools facilitate data preprocessing, visualization, functional annotation, network analysis and multi-omics integration.12 Nonetheless, effective data management is critical to handle high-volume, high-velocity datasets. This can be achieved by employing cloud computing platforms, database management systems and high-performance computing (HPC) infrastructure, which enable secure storage, break complex computational analyses into tasks and batches and empower data sharing across research groups.¹³'¹⁴
Applications and Clinical Implementation
Drug and Diagnostic Development
Biomarker discovery is pivotal in drug development, guiding the identification of therapeutic targets, patient stratification and treatment optimization. Predictive biomarkers help identify patients most likely to benefit from a therapy, while pharmacodynamic biomarkers can be used to monitor treatment response.¹⁵'¹⁶
In clinical trials, biomarkers assist with selecting patient populations, optimizing trial designs and using surrogate endpoints. Companion diagnostics are developed alongside targeted drugs to ensure that only patients with the appropriate biomarker profile receive treatment.¹⁷
Real-World Implementation
Biomarker discovery transforms clinical practice by improving precision in disease diagnosis, prognosis and therapeutic decision-making. Biomarkers uncover details about pathogenic variants, disease subtypes and druggable targets, guiding personalized treatment strategies. Clinicians can also utilize biomarker tests to monitor the response profile and predict adverse events.¹⁸
Disease-Specific Biomarker Discovery Approaches
Biomarker discovery is frequently utilized in several disease research areas, from oncology and neurology to metabolic and rare genetic disorders.
Multi-omics analysis is used in cancer research to identify mutations, differential gene expression, tumor-associated proteins, post-translational modifications and metabolic abnormalities.¹⁹ Liquid biopsy technologies are also being adopted to analyze the content of circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs).⁵
Neurology is another example of disease-specific application, as it focuses on digital biomarker discovery. Digital biomarkers leverage wearable sensors, mobile devices and other digital tools to capture real-world, continuous measurements of neurological function.² Thus, they help monitor motor movements, speech, sleep and cognitive activity in neurodegenerative diseases, such as Parkinson’s, Alzheimer’s and multiple sclerosis. They also complement traditional molecular biomarkers when tracking therapeutic responses.²⁰
For metabolic disorders, biomarkers help identify individuals at risk, assess disease progression and personalize interventions for conditions such as diabetes, obesity and cardiovascular disorders.²¹
Challenges and Opportunities
Biomarker discovery and development pipelines face a number of challenges that hinder the translation of candidate biomarkers to clinical applications.
Reproducibility of biomarker analysis and validation is a key issue, often caused by disease heterogeneity, inter-individual variability, sample quality differences and operational variations across laboratories and clinical trial sites. Furthermore, high-throughput technologies generate massive, complex datasets, requiring robust data management, integration and computational infrastructure. Simple missteps in sample collection, storage and processing may introduce variabilities that jeopardize the credibility of novel biomarkers.⁶
Following regulatory guidelines and adopting innovative approaches can overcome these challenges. Programs such as the FDA Biomarker Qualification Program and EMA’s Qualification of Novel Methodologies provide a benchmark for structured validation and clinical adoption pathways. In addition, integrating digital and real-world data through mobile devices and wearables provides evidence complementary to traditional clinical data.² These data sources improve patient monitoring by capturing longitudinal real-time trends.
Multimodal biomarker discovery, supported by AI/ML technologies, generates more comprehensive and systemic insights into disease and response profiles. In doing so, they accelerate predictive modelling, candidate biomarker selection and validation, paving the way for precision medicine initiatives.¹⁰
See how Danaher Life Sciences can help
FAQs
What are the main types of biomarkers used in drug development?
Key biomarkers include diagnostic biomarkers (identify disease presence), prognostic biomarkers (predict disease progression), predictive biomarkers (forecast treatment response), pharmacodynamic biomarkers (monitor drug effect) and safety biomarkers (indicate toxicity or adverse events).
How are candidate biomarkers identified and validated?
Candidates are first discovered through hypothesis-driven or high-throughput screening methods. Validation involves assessing content, construct and criterion validity, confirming reproducibility and demonstrating clinical utility across independent cohorts.
What technologies are commonly used in biomarker discovery?
Omics platforms (genomics, transcriptomics, proteomics, metabolomics), next-generation sequencing (NGS), mass spectrometry, high-throughput screening and liquid biopsies are widely used to uncover and quantify potential biomarkers.
How do diagnostic, prognostic and predictive biomarkers differ?
Diagnostic biomarkers detect disease, prognostic biomarkers forecast disease outcome and predictive biomarkers anticipate treatment response.
How does artificial intelligence enhance biomarker discovery and analysis?
AI and machine learning integrate multi-dimensional datasets, identify complex patterns, prioritize candidates and improve predictive accuracy, accelerating translational research.
References
- Kwon YW, Jo H-S, Bae S, Seo Y, Song P, Song M, et al. Application of proteomics in cancer: recent trends and approaches for biomarkers discovery. Front Med 2021;8:747333.
- Haghayegh F, Norouziazad A, Haghani E, Feygin AA, Rahimi RH, Ghavamabadi HA, et al. Revolutionary point‐of‐care wearable diagnostics for early disease detection and biomarker discovery through intelligent technologies. Adv Sci 2024;11(36):2400595.
- Dar MA, Arafah A, Bhat KA, Khan A, Khan MS, Ali A, et al. Multiomics technologies: role in disease biomarker discoveries and therapeutics. Brief Funct Genomics 2023;22(2):76-96.
- Reilly L, Seddighi S, Singleton AB, Cookson MR, Ward ME, Qi YA. Variant biomarker discovery using mass spectrometry-based proteogenomics. Front Aging 2023;4:1191993.
- Seyhan AA. Circulating liquid biopsy biomarkers in glioblastoma: advances and challenges. Int J Mol Sci 2024;25(14):7974.
- Purkayastha K, Dhar R, Pethusamy K, Srivastava T, Shankar A, Rath GK, et al. The issues and challenges with cancer biomarkers. J Cancer Res Ther 2023;19(Suppl 1):S20-S35.
- Ou F-S, Michiels S, Shyr Y, Adjei AA, Oberg AL. Biomarker discovery and validation: statistical considerations. J Thorac Oncol 2021;16(4):537-545.
- Attwood K. ROC methods in biomarker development. Modern Inference Based on Health-Related Markers: Elsevier; 2024:283-313.
- Cephe A, Koçhan N, Aksel EG, İpekten F, Yerlitaş Sİ, Zararsız GE, et al. Bioinformatics and biostatistics in precision medicine. Oncology: genomics, precision medicine and therapeutic targets: Springer; 2023:189-235.
- Prelaj A, Miskovic V, Zanitti M, Trovo F, Genova C, Viscardi G, et al. Artificial intelligence for predictive biomarker discovery in immuno-oncology: a systematic review. Ann Oncol 2024;35(1):29-65.
- Mann M, Kumar C, Zeng W-F, Strauss MT. Artificial intelligence for proteomics and biomarker discovery. Cell Syst 2021;12(8):759-770.
- Franz M, Lopes CT, Fong D, Kucera M, Cheung M, Siper MC, et al. Cytoscape. js 2023 update: a graph theory library for visualization and analysis. Bioinformatics 2023;39(1):btad031.
- Flöther FF, Blankenberg D, Demidik M, Jansen K, Krishnakumar R, Krishnakumar R, et al. How quantum computing can enhance biomarker discovery. Patterns 2025;6(6).
- Hemme CL, Beaudry L, Yosufzai Z, Kim A, Pan D, Campbell R, et al. A cloud-based learning module for biomarker discovery. Brief Bioinform 2024;25(Supplement_1):bbae126.
- Salawu A, Hernando-Calvo A, Chen RY, Araujo DV, Oliva M, Liu ZA, et al. Impact of pharmacodynamic biomarkers in immuno-oncology phase 1 clinical trials. Eur J Cancer 2022;173:167-177.
- Tarighati E, Keivan H, Mahani H. A review of prognostic and predictive biomarkers in breast cancer. Clin Exp Med 2023;23(1):1-16.
- Orellana García LP, Ehmann F, Hines PA, Ritzhaupt A, Brand A. Biomarker and companion diagnostics—a review of medicinal products approved by the European Medicines Agency. Front Med 2021;8:753187.
- Hansson O, Blennow K, Zetterberg H, Dage J. Blood biomarkers for Alzheimer’s disease in clinical practice and trials. Nat Aging 2023;3(5):506-519.
- Aung TN, Bates KM, Rimm DL. High-plex assessment of biomarkers in tumors. Mod Pathol 2024;37(3):100425.
- Chudzik A, Śledzianowski A, Przybyszewski AW. Machine learning and digital biomarkers can detect early stages of neurodegenerative diseases. Sensors 2024;24(5):1572.
- Metwaly A, Reitmeier S, Haller D. Microbiome risk profiles as biomarkers for inflammatory and metabolic disorders. Nat Rev Gastroenterol Hepatol 2022;19(6):383-397.