What is the process of new drug discovery and development?

21 March 2025
Introduction to Drug Discovery and Development
New drug discovery and development is a highly complex, multidisciplinary process that combines basic biological research, chemistry, pharmacology, toxicology, and clinical sciences to bring a novel therapeutic from concept into widespread clinical use. This process takes many years, requires huge financial investments, and involves extensive collaboration between academic researchers, industry scientists, clinicians, regulatory agencies, and many other stakeholders. Overall, the journey from laboratory discovery to an approved and marketable drug is divided into two broad phases: “drug discovery” and “drug development.” These phases must be integrated thoughtfully in order to increase the chance of success, reduce attrition rates, and ultimately bring safe, efficacious drugs to patients.

Overview of Drug Discovery
Drug discovery is the initial stage of the process and involves the identification of promising biological targets, the detection of compounds that interact with these targets, and the subsequent validation of these compounds as viable starting points for therapeutic development. Modern drug discovery relies on robust experimental strategies such as high-throughput screening, combinatorial chemistry, and data‐driven computational approaches that enhance the identification and validation of novel drug candidates. This phase is inherently exploratory and focuses on generating chemical diversity and optimizing interactions with the biological target so that further improvement is possible. It also incorporates advances in genomics, bioinformatics, and structural biology that provide critical insights into disease mechanisms and allow more targeted approaches to be developed.

Overview of Drug Development
Drug development follows discovery and comprises everything needed to ensure the safety, efficacy, quality, and manufacturing controls of a candidate drug before it reaches the market. This phase can be further subdivided into preclinical testing, clinical trials in humans, and finally, the regulatory approval process along with post-marketing surveillance. The drug development stage is not only about confirming that the candidate has the desired therapeutic effect in humans but also about establishing an acceptable safety profile and optimized pharmacokinetics and pharmacodynamics. The entire process is tightly regulated by national and international authorities to ensure that only medicines with a favorable benefit-risk profile eventually reach patients.

Stages of Drug Discovery
The drug discovery stage is characterized by a series of iterative steps that steadily narrow down the vast chemical space to a few promising candidates. These include the identification of targets, the screening for hit compounds, and iterative optimization through lead identification and lead optimization.

Target Identification and Validation
The process begins with target identification, which involves selecting a biological process or molecular component (such as a receptor, enzyme, or gene) that is critically involved in disease pathology. Advanced techniques including data mining, genomics, proteomics, and high-resolution structural determination (using X-ray crystallography, NMR, or cryo-EM) are available to pinpoint viable targets. Once the target is identified, target validation follows to confirm that modulation of this target by a therapeutic compound will yield a beneficial clinical outcome. Target validation may include genetic manipulations (e.g., knockout/knockdown studies), pharmacological validation using tool compounds, and biochemical assays to define the role of the target in disease. This stage requires rigorous careful experimental design and is critical, because if the target is not “druggable” or its modulation does not translate into a therapeutic benefit, subsequent drug development efforts could fail despite successful hit identification.

Lead Compound Identification
After validation, the next stage is hit identification, wherein large chemical libraries are screened for compounds that interact with the target with acceptable potency. The hits are then refined to yield “lead compounds” which show a combination of desirable properties—including favorable binding affinity, selectivity, metabolic stability, and lower toxicity. Both experimental methods (such as high-throughput screening, fragment-based screening, and phenotypic assays) and computational approaches such as virtual screening and computer-aided drug design (CADD) are employed to identify initial hits. These hits are characterized extensively through structure-activity relationship (SAR) studies, and the most promising ones become the lead compounds that further undergo chemical modifications. A critical aspect of hit identification is the use of orthogonal methods; for example, hits from a high-throughput screening may be cross-validated using crystallographic fragment screening to confirm binding and offer structural information for further optimization.

Lead Optimization
Once lead compounds are identified, lead optimization is undertaken to improve their properties and to ensure the candidate has the highest chance for translation into a successful drug. Lead optimization includes systematic chemical modifications directed at increasing potency, selectivity, oral bioavailability, metabolic stability, and minimizing off-target toxicities. In this stage, computational modeling—including quantitative structure–activity relationship (QSAR) analyses and molecular dynamics simulations—play an increasing role as they help predict how chemical changes will alter pharmacological and pharmacokinetic profiles. The optimization process can use iterative cycles of synthesis and testing coupled with advanced analytical instrumentation to rapidly profile changes. Ultimately, the goal is to produce a compound that not only binds effectively to the target but also demonstrates favorable absorption, distribution, metabolism, excretion (ADME) properties, and low toxicity in preliminary in vitro and in vivo assays.

Stages of Drug Development
After a candidate compound has been optimized, it enters the drug development phase—a series of structured, regulated steps that further test and verify the candidate’s suitability for clinical use in humans.

Preclinical Testing
Preclinical testing is the stage where the candidate drug is rigorously evaluated in vitro (cell-based assays) and in vivo (animal models) for its safety and biological activity. These studies are designed to establish proof-of-concept and evaluate toxicological limits, pharmacokinetics (PK), pharmacodynamics (PD), and bioavailability. Preclinical studies involve dose ranging, toxicology studies (acute, subchronic, and chronic toxicity evaluations), and safety pharmacology studies that assess any unwanted effects on vital systems (e.g., cardiovascular, respiratory, and CNS effects). Often, the number of molecules entering preclinical trials is much larger than those that eventually proceed to human testing, as extensive preclinical evaluation is critical to ensure that only safe candidates advance. These studies must conform to Good Laboratory Practice (GLP) standards and produce a complete data package required for submission as part of an Investigational New Drug (IND) application.

Clinical Trials (Phase I, II, III)
Once preclinical testing establishes the candidate drug’s safety and potential efficacy, clinical trials in humans commence. Clinical research is typically divided into three main phases:

Phase I:
Phase I trials are the first studies conducted in humans and are primarily focused on establishing safety, tolerability, pharmacokinetics, and pharmacodynamics of the candidate drug in a small number (usually 10–50) of healthy volunteers or patients in special cases. The primary objective in Phase I is to determine the maximum tolerated dose as well as to profile side effects. The data obtained during this phase is crucial for setting the dosing regimen for later phases. These early trials also help in understanding how the drug is absorbed, distributed, metabolized, and excreted in the human body.

Phase II:
Phase II trials are designed to obtain preliminary data on the efficacy of the drug as well as to further evaluate its safety in a larger group of patients affected by the target disease (typically several dozen to a few hundred). This phase often includes dose-ranging studies to determine the optimal therapeutic dose. Phase II trials may be randomized, controlled (with placebo or active comparator), and include more intensive monitoring of adverse events. These studies continue to elucidate the drug’s pharmacologic profile and help optimize its use while beginning to assess clinical endpoints related to efficacy.

Phase III:
Phase III trials are pivotal studies conducted in larger patient populations (ranging from hundreds to thousands) to confirm the drug’s efficacy and safety on a statistically robust scale. These trials are often randomized, double-blind, and controlled studies that compare the new drug to standard therapies or placebo. The key outcome metrics may include clinical endpoints such as overall survival, disease progression, and quality-of-life improvements, which are critically evaluated to determine if the benefit-risk ratio of the drug supports its approval for marketing. This phase also provides safety data to detect less common adverse events and forms the basis for the final marketing authorization submission to regulatory bodies.

Regulatory Approval and Post-Marketing Surveillance
Following the successful completion of Phase III, a comprehensive data package is assembled for submission to regulatory agencies (e.g., the FDA in the United States or the EMA in Europe) for a New Drug Application (NDA) or Biologics License Application (BLA). Regulatory authorities review data on safety, efficacy, manufacturing quality, and overall benefit-risk balance before granting marketing approval. The review process involves a detailed evaluation by multidisciplinary teams, and depending on the jurisdiction, the approval process can take months to a year. Upon approval, the drug is launched into the market.

Once a drug is approved and marketed, post-marketing surveillance (Phase IV) is initiated to continuously monitor the drug’s performance in the real world. Pharmacovigilance systems collect adverse event reports from healthcare professionals and patients, while additional studies may be conducted to monitor long-term safety and efficacy. Post-marketing studies are crucial for detecting rare or long-term side effects that may not have been evident in the more controlled environment of clinical trials. Data from spontaneous reporting systems, healthcare databases, active surveillance programmes, and systematic reviews contribute to ensuring that the drug continues to have an acceptable benefit-risk profile in the broader population.

Challenges and Innovations
The entire new drug discovery and development process faces various challenges that result in high attrition rates, significant time delays, and immense financial burdens. However, at the same time, technological innovations are driving transformative changes that help address these challenges.

Current Challenges in Drug Discovery and Development
One of the major challenges in drug discovery is the high failure rate of candidates as they advance through preclinical and clinical stages, with estimates indicating that only one in several thousand compounds eventually reaches clinical use.
The discovery process itself is hampered by immense chemical space and variability in biological systems. Identifying viable targets and finding compounds with a favorable safety and efficacy profile requires extensive screening and validation studies, and many potential targets ultimately prove non-druggable or fail to yield a meaningful clinical benefit.
Drug development is challenged by regulatory hurdles, the need to comply with rigorous standards such as Good Laboratory Practice (GLP) and Good Manufacturing Practice (GMP), and the inherent limitations of animal models that may not fully predict human responses.
In clinical trials, designing studies that are both statistically robust and ethically sound is difficult. Challenges include patient heterogeneity, issues of safety dosing in Phase I, determining optimal endpoints in Phase II, and ensuring that Phase III trials are definitive yet cost-effective.
Furthermore, the entire process is time-consuming and expensive; the total development time for a new drug is often over a decade, with costs sometimes exceeding billions of dollars. Delays can occur at each step—from target validation to clinical trials to regulatory review—and safety concerns discovered post-marketing may lead to drug withdrawal.

Technological Innovations and Their Impact
Technological advances have begun to transform both drug discovery and drug development. Innovations in genomics, proteomics, and bioinformatics enable researchers to analyze vast datasets and identify novel targets based on genetic and molecular profiles. These developments help in stratifying patient populations for more personalized therapies and in selecting drug candidates that are more likely to succeed based on molecular similarity and biological relevance.
Computational methods, including computer-aided drug design (CADD), molecular docking, quantitative structure–activity relationships (QSAR), and machine learning algorithms, have significantly improved the speed and accuracy of screening and lead optimization. For example, deep-learning techniques that predict binding affinities and generate novel chemical structures have accelerated the identification of promising leads, thereby reducing the burden on high-throughput screening laboratories.
In the domain of drug development, advances in high-throughput screening technologies, the use of high-resolution analytical instruments, and the application of three-dimensional bioprinting in the production of more human-like tissue models for safety and efficacy testing are reshaping preclinical studies. These innovations not only improve the reliability of preclinical data but also help in better predicting clinical outcomes.
Moreover, adaptive clinical trial designs that use real-time data analyses are being adopted to expedite decision-making and lower costs. Novel regulatory pathways such as accelerated approval, conditional marketing authorization, and priority review processes have been introduced by agencies like the FDA and EMA to bring lifesaving drugs to patients sooner while maintaining rigorous safety oversight.
Post-marketing surveillance is also evolving through the integration of electronic healthcare databases and new signal detection algorithms, including the use of social media listening for pharmacovigilance. These approaches have the potential to detect adverse events earlier and with greater precision than traditional spontaneous reporting systems, thereby enhancing drug safety after market launch.

Future Directions
Looking forward, the field of drug discovery and development is expected to continue evolving with an emphasis on translational medicine, precision therapeutics, and next-generation data analytics. The future directions in the process are driven by both the need to overcome current challenges and the promise offered by emerging technologies.

Emerging Trends
There is an increasing trend toward personalized medicine, whereby therapies are tailored to individual genetic profiles and disease characteristics. Advances in next-generation sequencing and big data analytics allow the identification of molecular biomarkers that can be used to predict patient response, leading to more effective clinical trials and a reduction in late-stage failures.
The integration of artificial intelligence and machine learning into every aspect of drug discovery—from target identification to lead optimization and clinical trial design—is rapidly gaining momentum. These tools are being used to analyze complex multidimensional datasets, identify patterns in patient responses, and optimize drug candidates based on predicted ADME and toxicity profiles.
Adaptive trial designs and real-world evidence are also emerging as powerful tools to accelerate the clinical development process. Novel trial designs that incorporate Bayesian statistics and interim analyses allow for modifications during the study, thereby increasing efficiency and reducing patient exposure to ineffective treatments.
Additionally, there is growing enthusiasm for using patient-derived organoids and three-dimensional bioprinted tissues as in vitro models that more closely mimic human physiology. Such models have the potential to bridge the gap between animal studies and human clinical trials, enhancing the predictive power of preclinical models and reducing development costs.

Future Research Directions
Future research in drug discovery and development will likely focus on further integrating multi-omics data (genomics, proteomics, metabolomics) with advanced computational techniques to identify novel therapeutic targets with greater precision than ever before. Research is also needed to develop robust methods for high-throughput de novo drug design that incorporate adaptive machine learning algorithms for rapid identification of lead compounds.
There will also be a strong emphasis on improving the translatability of preclinical findings into clinical success. Advances in tissue engineering, such as the development of organ-on-chip technologies and bioprinted models, are expected to play a crucial role in this regard by offering more physiologically relevant platforms for early drug testing.
In clinical development, future research should further explore innovative trial designs that combine adaptive algorithms with real-world data collection, making it possible to monitor drug safety and efficacy continuously even after market approval. Regulatory science itself is an area of active research as agencies around the world initiate pilot projects and harmonization efforts to expedite reviews without compromising safety.
Finally, continued interdisciplinary collaboration between academic, industrial, and regulatory stakeholders will be essential to drive innovation. Research into the development of new biomarkers for efficacy and safety, more precise adverse event detection methodologies using large-scale electronic health records, and improved standards for data quality and integration across different systems will be critical to further reduce the risk of post-marketing failures and improve the overall success of drug development programs.

Conclusion
In summary, the process of new drug discovery and development is a long, iterative, and complex journey that begins with the discovery of a novel biological target and moves through several stages of compound screening, validation, and optimization to eventual human testing in a highly regulated clinical setting. The discovery process relies on modern methods including high-throughput screening, advanced computational modeling, and multi-dimensional target validation strategies to identify promising hits followed by rigorous lead optimization. Once a candidate has been optimized, it enters the development phase where extensive preclinical testing in cells and animal models is performed to determine safety and pharmacokinetics, ultimately paving the way for clinical trials in humans. These clinical phases (Phase I, II, III) are designed to assess safety, dosage, efficacy, and confirm the benefit-risk profile before comprehensive regulatory review and marketing approval. After approval, continuous post-marketing surveillance ensures that any rare or long-term adverse events are detected and addressed promptly.

The challenges in this process are substantial: high costs, long timelines, high attrition and safety issues that necessitate immense scrutiny at every step. Yet, technological innovations—from genomics and bioinformatics to adaptive clinical trial designs and high-resolution tissue models—are continuously transforming the landscape, making the process more precise, efficient, and predictive. Looking ahead, emerging trends such as personalized medicine, integration of artificial intelligence, and novel preclinical models promise to further streamline drug discovery and improve clinical outcomes, while regulatory bodies are evolving to support faster approvals under defined conditions, thereby addressing urgent unmet medical needs.

Overall, the new drug discovery and development continuum is a dynamic interplay between scientific exploration, technological innovation, clinical validation, and regulatory oversight. It requires a robust infrastructure, cross-disciplinary collaboration, and a commitment to ensuring that safety, efficacy, and quality remain at the forefront. With continued advances in technology and data analytics and the incorporation of real-world evidence and adaptive designs, the future of drug development appears promising. The ultimate goal remains the same: to deliver safe, effective, and innovative treatments to patients as efficiently as possible while minimizing risks and costs. This integrated, comprehensive approach is vital for sustained progress in pharmaceutical sciences and for improving patient outcomes worldwide.

In conclusion, the process—from target validation through lead optimization, rigorous preclinical studies, and exhaustive clinical testing to final regulatory review and post-marketing surveillance—represents a journey of scientific innovation, rigorous testing, and continuous learning. By integrating cutting-edge technologies and embracing adaptive and collaborative approaches, the industry is working to overcome existing challenges and shape a future where drug discovery and development are faster, safer, and more cost-effective. This not only enhances therapeutic efficacy and safety for patients but also drives further innovation in the ever-evolving field of pharmaceutical sciences.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成