Data Handling and Insight Generation in Drug Discovery and Development

CRISPR Cas9

Drug discovery and development is a complex, multi-stage journey, from initial target identification and lead optimization to preclinical evaluation, clinical trials and eventual market approval. Each stage generates large volumes of heterogeneous information, making robust data handling essential to ensure data integrity, reproducibility and timely decision-making.

Despite providing comprehensive molecular insights into disease mechanisms and drug responses, recent advances in multi-omics technologies add to the data complexity.1

Simultaneously, the rise of digital health platforms has been reshaping pharmaceutical R&D through real-time monitoring, remote data capture and patient-centered approaches.2

Overall, robust data handling technologies must be developed to accommodate the increasing diversity of data structures, ultimately creating more efficient and data-driven drug development pipelines.

Integration of Complex Data in the Drug Discovery and Development Pipeline

Modern drug discovery relies on integrating diverse data types to inform decisions across every stage of development. These data types include omics datasets, clinical trial data and real-world evidence, each playing distinct roles at various stages.

Integrating highly dimensional data signifies a shift from traditional drug development models to data-driven, digital health-supported pipelines that emphasize computational tools, centralized data management and real-time data capture.

Data Curation, Integration and Sharing

Strategies and Importance

Effective data curation, integration, storage and sharing are essential for extracting actionable insights from complex datasets. Biopharmaceutical organizations face several challenges in this space, including data heterogeneity, scale, inconsistent formats and a lack of validation. Addressing these challenges is necessary to achieve data integrity, reproducibility, collaboration and regulatory compliance milestones.

Tools

To manage these challenges, pharmaceutical companies employ a variety of technological solutions:

Understanding Diverse Data Types in Drug Discovery

Major Data Sources

Drug discovery relies on a wide range of data types to decipher disease mechanisms and develop effective therapeutics. These datasets include: 8

Data Complexity and Challenges

Harmonizing the myriad of datasets can be difficult. More specifically, structured tabular records from laboratory experiments must be integrated with unstructured data, including clinical notes, medical images and free-text reports.8

Researchers must follow a three-fold approach to address this issue:

Data Management and Quality across the Discovery Pipeline

Foundations of Effective Data Management

High-quality data handling forms the backbone of modern drug discovery. Core data handling principles, such as data integrity, validation, standardization and quality, ensure that results are accurate, reproducible and meaningful. Pharmaceutical companies must adhere to these principles for regulatory compliance and audit readiness, as data accuracy and traceability directly affect approvals and patient safety.3

Infrastructure and Systems

Robust infrastructure is required to manage the scale and diversity of research data. Cloud-based data management solutions are adopted to provide scalability, secure storage and global accessibility. These platforms feature centralized database architectures to unify access to information, reducing data redundancy and improving consistency across teams. Cloud systems also impose strict security protocols to store sensitive research and clinical data and maintain confidentiality. 9,10

Workflow and Automation

Automation plays a pivotal role in accelerating data management and minimizing human error. Workflow automation streamlines repetitive tasks such as data capture, annotation and transfer, as well as more complex tasks involving real-time data management. 11

Best practices and Compliance

Regardless of cutting-edge data management tools that reduce the need for manual data handling, researchers must adhere to practices for effective data management.

Data collection in preclinical research must follow the FAIR (Findable, Accessible, Interoperable, Reusable) principles to improve transparency and reproducibility.12

Similarly, in clinical trials, researchers must stick to regulatory-compliant data management frameworks to safeguard patient well-being while meeting submission requirements.3

Data Analysis and Computational Methods for Insight Generation

Core Analytical Methods

Data analytics is a fundamental tool in drug discovery, enabling actionable insights from complex datasets. It helps researchers identify target-phenotype patterns, rank drug candidates and anticipate clinical outcomes. These approaches collectively support hypothesis generation and validation. Moreover, data visualization converts complex omics and clinical trial data into interpretable and presentable formats that guide decision-making across different legs of the drug development lifecycle.8

Advanced Computational Tools

Computational science lies at the center of data analytics.

Together, these tools help uncover novel associations between targets, pathways and therapeutic outcomes.

AI-Driven Insight Generation

Artificial intelligence has been reshaping data insight generation in several stages of drug discovery. Machine learning and deep learning algorithms can predict ADME properties, guide patient stratification and identify drug-protein interactions with greater accuracy than traditional workflows.15-17

While AI-supported tools are already valuable as standalone applications, their embedding into data management workflows systemizes data handling across entire drug discovery pipelines. Thus, converging AI and data infrastructure supports more efficient, predictive and scalable drug development.

Translational Research, Biomarker Discovery and Multi-Omics Integration

Bridging Research to Clinical Outcomes

Translational research serves as the critical link between laboratory findings and clinical applications, transforming data insights into meaningful action plans for patient care. Omics data, ranging from genomics to metabolomics, combined with digital health tools, help translate preclinical discoveries into clinically relevant endpoints. A key objective in translational research is the identification of biomarkers that guide patient stratification, predict therapeutic response and improve trial design.18

Integrated datasets of molecular profiles, clinical trial records and real-world evidence have provided a myriad of diagnostic models in cancer, infectious diseases and metabolic disorders.19-21

Centralized cloud platforms can strengthen data management and analysis in translational research by fostering data standardization, ultimately benefiting collaboration efforts across research groups and institutions. 22

Ensuring regulatory compliance and reproducibility

Standards and Submission-Readiness

Regulatory compliance validates the credibility of any pharmaceutical research and must be approached with utmost care. Data management standardization is instrumental in preparing companies for submission. Standardized documentation ensures data integrity, consistency, transparency and traceability across the pipeline. Well-structured datasets and carefully crafted applications simplify regulatory review and reduce the likelihood of delays or resubmissions.3

Pharmaceutical companies must use audit-ready, secure and validated data systems compliant with Good Clinical Practice (GCP), Good Laboratory Practice (GLP) and 21 CFR Part 11 requirements. They must also follow high-quality data handling practices, such as rigorous documentation, version control and encryption, to cultivate transparent and objective research ethics.23

Real-World Data, Digital Health and Post-Market Surveillance

Applications in Pharmacovigilance

Given the importance of monitoring a drug's long-term effects and market performance, data management must transcend drug discovery pipelines and become a routine practice in post-market surveillance.

To that end, real-world evidence (RWE) drawn from electronic health records, patient registries, mobile health applications and digital monitoring platforms provides continuous data insights beyond the scope of clinical trials. By capturing data from diverse populations and longer-term use, RWE strengthens pharmacovigilance and supports adaptive regulatory decision-making.4

Digital health platforms are at the core of pharmacovigilance. They can monitor adverse events, treatment adherence and patient-reported outcomes. These insights allow for proactive risk detection and improved patient safety when combined with advanced analytics.2

Insights from digital health platforms must be leveraged through collaborative data sharing between stakeholders, regulators, healthcare providers and pharmaceutical companies. Such collaboration fosters a comprehensive view of drug performance, ensuring that both safety concerns and therapeutic benefits are tracked effectively across populations.2

Innovation in Data Infrastructure

The pharmaceutical R&D landscape continues to evolve with advancements in data handling.

One major trend is the rise of cloud-based data management platforms, which provide scalable storage while mediating secure access and collaboration across global research teams. These platforms reduce reliance on local infrastructure, improve data traceability and support integration of increasingly complex datasets.24

Simultaneously, AI-driven automation and cheminformatics improve many aspects of discovery workflows. Machine learning-aided bioinformatic and cheminformatics can standardize compound screening and structure–activity relationship analyses, allowing researchers to process massive chemical and biological datasets efficiently.13

Together, these innovations point toward a future in which data handling is more dynamic and informative, providing real-time information along the journey from molecule to medicine.

See how Danaher Life Sciences can help

Talk to an expert

FAQs

Why is data handling critical in the drug discovery and development process?

Effective data handling ensures accuracy, reproducibility and regulatory compliance across the pipeline, reducing errors and accelerating decision-making.

What are the best practices for data management in drug discovery?

Key practices include data standardization, validation, adherence to FAIR principles and maintaining audit-ready, secure systems.

How do multi-omics approaches contribute to drug discovery?

Genomics, proteomics, metabolomics and transcriptomics provide holistic insights into disease biology, supporting target identification and biomarker discovery.

How is artificial intelligence used for insight generation in drug development?

AI supports predictive modeling, biomarker identification, trial optimization and automated analysis of high-dimensional datasets.

What role does real-world data play in drug development?

Real-world evidence informs post-market surveillance, safety monitoring and evaluation of drug effectiveness across diverse populations.

What are centralized data management platforms?

These platforms unify storage, access and integration of diverse datasets, enhancing collaboration and interoperability in pharma R&D.

References

  1. Mukherjee A, Abraham S, Singh A, Balaji S, Mukunthan K. From data to cure: A comprehensive exploration of multi-omics data analysis for targeted therapies. Mol Biotechnol 2025;67(4):1269-1289.
  2. Zarour M, Alenezi M, Ansari MTJ, Pandey AK, Ahmad M, Agrawal A, et al. Ensuring data integrity of healthcare information in the era of digital health. Healthc Technol Lett 2021;8(3):66-77.
  3. Madabushi R, Seo P, Zhao L, Tegenge M, Zhu H. Role of model-informed drug development approaches in the lifecycle of drug development and regulatory decision-making. Pharm Res 2022;39(8):1669.
  4. Lavertu A, Vora B, Giacomini KM, Altman R, Rensi S. A new era in pharmacovigilance: toward real‐world data and digital monitoring. Clin Pharmacol Ther 2021;109(5):1197-1202.
  5. Famili P, Cleary S. Laboratory Information Management System (LIMS) and Electronic Data. Analytical Testing for the Pharmaceutical GMP Laboratory 2022:345-373.
  6. Liu W, Li Y, Li X, Wang F, Qi R, Zhu T, et al. Pooled Analysis of the Effect of Pre-Existing Ad5 Neutralizing Antibodies on the Immunogenicity of Adenovirus Type 5 Vector-Based COVID-19 Vaccine from Eight Clinical Trials. Vaccines 2025;13(3):333.
  7. Fu J, Zhang Y, Wang Y, Zhang H, Liu J, Tang J, et al. Optimization of metabolomic data processing using NOREVA. Nat Protoc 2022;17(1):129-151.
  8. Coltman NJ, Roberts RA, Sidaway JE. Data science in drug discovery safety: Challenges and opportunities. Exp Biol Med 2023;248(21):1993-2000.
  9. Gomase VS, Ghatule AP, Sharma R, Sardana S, Dhamane SP. Cloud Computing Facilitating Data Storage, Collaboration, and Analysis in Global Healthcare Clinical Trials. Rev Recent Clin Trials 2025.
  10. Ramapraba PS, Babu BR, Paul NRR, Sharmila V, Babu VR, Ramya R, et al. Implementing cloud computing in drug discovery and telemedicine for quantitative structure-activity relationship analysis. IJECE 2025;15(1):1132-1141.
  11. Singh S. Automation of Drug Design and Development. Generative Artificial Intelligence for Biomedical and Smart Health Informatics 2025:73-87.
  12. Gadiya Y, Ioannidis V, Henderson D, Gribbon P, Rocca-Serra P, Satagopam V, et al. FAIR data management: what does it mean for drug discovery? Front Drug Discov (Lausanne) 2023;3:1226727.
  13. Parikh PK, Savjani JK, Gajjar AK, Chhabria MT. Bioinformatics and cheminformatics tools in early drug discovery. Bioinformatics tools for pharmaceutical drug product development 2023:147-181.
  14. Liu C, Zhang H. Data processing for high-throughput mass spectrometry in drug discovery. Expert Opin Drug Discov 2024;19(7):815-825.
  15. Rehman AU, Li M, Wu B, Ali Y, Rasheed S, Shaheen S, et al. Role of artificial intelligence in revolutionizing drug discovery. Fundam Res 2025;5(3):1273-1287.
  16. Yin J, Qi Y, Zhu F, Zeng S. The Application of Artificial Intelligence in Drug ADME Research. BSP; 2025.
  17. Glaab E, Rauschenberger A, Banzi R, Gerardi C, Garcia P, Demotes J. Biomarker discovery studies for patient stratification using machine learning analysis of omics data: a scoping review. BMJ open 2021;11(12):e053674.
  18. Wichman C, Smith LM, Yu F. A framework for clinical and translational research in the era of rigor and reproducibility. J Clin Transl Sci 2021;5(1):e31.
  19. Xu R, Wang J, Zhu Q, Zou C, Wei Z, Wang H, et al. Integrated models of blood protein and metabolite enhance the diagnostic accuracy for Non-Small Cell Lung Cancer. Biomark Res 2023;11(1):71.
  20. Bourgonje AR, van Goor H, Faber KN, Dijkstra G. Clinical value of multiomics-based biomarker signatures in inflammatory bowel diseases: Challenges and opportunities. Clin Transl Gastroenterol 2023;14(7):e00579.
  21. Veyel D, Wenger K, Broermann A, Bretschneider T, Luippold AH, Krawczyk B, et al. Biomarker discovery for chronic liver diseases by multi-omics–a preclinical case study. Sci Rep 2020;10(1):1314.
  22. 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.
  23. Ladner T, Weh C, Dhillon A, Giffard M, Iacovelli D. Data computation platform (DCP): empowering pharma 4.0 innovation through GxP-compliant and scalable software platform enabling advanced data analytics and real-time process monitoring in regulated environments. J Intell Manuf 2025:1-19.
  24. Rehan H. Advancing cancer treatment with ai-driven personalized medicine and cloud-based data integration. J Mach Learn Res 2024;4(2):1-40.