
Why Drug Development Can Take Decades
Developing a new medicine is an extraordinarily long, expensive and risky process. On average, bringing a new drug from initial discovery to market approval takes well over a decade. For example, industry surveys and analyses estimate 12–13 years for a new drug to reach patients efpia.eu biostock.se. In emerging fields such as gene therapy or novel biologics, this span can extend 20–30+ years statnews.com biostock.se. The pipeline begins with screening thousands of compounds and ends with one approved product. Only ~1–2 of every 10,000 compounds entering laboratory studies will ultimately become a marketed drug efpia.eu biostock.se. This attrition, combined with rigorous safety and efficacy requirements, drives the long timelines.
! https://biostock.se/en/2019/10/biostocks-article-series-on-drug-development-the-process/
Figure: Typical drug development timeline, showing major stages from discovery to market approval. Numbers indicate approximate durations and attrition (e.g. thousands of candidates in discovery/preclinical, dwindling to one approved drug). Sources: FDA/IFPMA data biostock.se biostock.se.
Discovery and Preclinical Research
The first phase is discovery and preclinical research, which itself can span several years. During discovery researchers identify biological targets (genes, proteins, pathways) implicated in a disease and then screen thousands of compounds (small molecules or biologics) for activity. Modern in silico and high-throughput methods expedite this, but only a tiny fraction show promise. Estimates suggest that initial screening may start with 5,000–10,000 candidate molecules【54†, of which roughly 100–200 become “lead” candidates for further study【54†. These leads then undergo intensive medicinal chemistry or engineering optimization to improve potency, selectivity and drug-like properties.
Selected lead compounds proceed to preclinical development, involving in vitro (cell-based) and in vivo (animal) studies to assess pharmacology and toxicity biostock.se fda.gov. Preclinical work typically takes on the order of 3–6 years biostock.se. Investigators perform repeated-dose toxicity testing in at least two animal species, genotoxicity, safety pharmacology (cardiac, respiratory, CNS), and early pharmacokinetic studies. The goal is to demonstrate an acceptable safety margin before human testing. Preclinical studies must comply with Good Laboratory Practice (GLP) standards and are summarized in a regulatory application. In the U.S., sponsors submit an Investigational New Drug (IND) application to the FDA (or a Clinical Trial Application (CTA) in the EU) after completing preclinical work fda.gov biostock.se. The IND includes animal safety data, manufacture/CMC information and clinical protocols fda.gov. Regulators have 30 days (in the U.S.) to review the IND; if no clinical hold is issued, the program may proceed to human trials.
Because the discovery phase involves fundamental research and compound optimization, it is inherently time-consuming. The International Federation of Pharmaceutical Manufacturers (IFPMA) notes that discovery + preclinical development can take about 4–7 years before an IND/CTA filing biostock.se. During this time most candidates fail due to toxicity or lack of efficacy. By one estimate only 1–2 of every 10,000 tested molecules emerge as viable drugs efpia.eu. These success odds reflect the many hurdles (unmet target validation, off-target effects, poor ADME properties) in early development.
Clinical Development (Phase I–III)
Once an IND/CTA is cleared, the drug enters clinical trials, which are conventionally divided into Phases I–III. Each phase is progressively larger and longer, with strict regulatory oversight. The overall clinical development timeline (Phase I through NDA/BLA submission) can be 8–10 years or more, and trials account for the bulk of the cost and time. Median clinical development time for recent new drugs has been estimated at ~8–9 years pmc.ncbi.nlm.nih.gov, with more complex or rare-disease programs often taking longer. Importantly, the probability of a candidate succeeding through all phases is very low: industry analyses cite roughly 10–15% cumulative clinical success pmc.ncbi.nlm.nih.gov pmc.ncbi.nlm.nih.gov (i.e. roughly 9–10 out of 10 candidates that enter trials ultimately fail).
Phase I – First-in-Human Safety
Phase I trials are the first tests in humans, primarily assessing safety, tolerability and pharmacokinetics. These are small studies (typically 20–80 healthy volunteers, or sometimes patients for oncology drugs) fda.gov. Phase I generally lasts several months to a year fda.gov. Researchers gradually escalate doses to determine a safe range and study ADME (Absorption, Distribution, Metabolism, Excretion). By definition, Phase I verifies whether human PK/PD matches preclinical models and uncovers any unexpected toxicity.
Success rates out of Phase I are moderate: about 60–70% of compounds move on to Phase II fda.gov. Reasons for failure here include toxicity or intolerable side effects in humans. Typical Phase I timelines range up to 12 months (though often shorter), partly due to stringent safety monitoring and regulatory approvals. Enrollment challenges can also slow phase I (needing healthy volunteers with no comorbidities).
Phase II – Proof-of-Concept in Patients
Phase II trials are the first studies to test efficacy (in addition to safety) in patients who have the target disease. These trials generally involve a few dozen to several hundred patients (commonly 100–300 patients) and can last 1–2 years fda.gov biostock.se. Phase II is often subdivided into Phase IIa (dose-finding) and IIb (efficacy testing). Investigators use randomized, often placebo- or standard-of-care–controlled designs to determine if the drug has sufficient clinical effect.
Attrition spikes in Phase II: historically only ~30–35% of compounds entering Phase II progress to Phase III fda.gov. Inadequate efficacy is the leading cause of Phase II failures (roughly 40–50% of clinical failures are due to lack of efficacy pmc.ncbi.nlm.nih.gov), as many promising preclinical candidates do not translate into patient benefit. Other Phase II failures arise from unexpected toxicities or pharmacokinetic issues that appear under therapeutic dosing. Designing Phase II studies can be challenging: choosing endpoints, biomarkers, optimal dose and patient population requires careful planning. If Phase II is slow to recruit or shows equivocal data, sponsors may run out of resources or terminate the program.
Recruitment and retention are critical bottlenecks. Complex protocols and stringent inclusion criteria often make patient accrual slow. Industry analyses (Tufts CSDD, 2012) found that 53% of trials needed to extend enrollment timelines beyond plan appliedclinicaltrialsonline.com, and up to 90% of trials face delays that miss original timelines appliedclinicaltrialsonline.com. Even after enrollment, patient drop-out rates are significant (for example, ~17% of patients may drop out before trial completion appliedclinicaltrialsonline.com). These factors lengthen Phase II (and III) durations.
Phase III – Pivotal Confirmation
Phase III trials are large, definitive studies intended to conclusively demonstrate safety and efficacy for regulatory approval. They typically enroll hundreds to thousands of patients (often 300–3,000 or more) in one or more long-term studies fda.gov. Phase III can take 2–4 years on average fda.gov biostock.se. By this stage the therapeutic dose and population are defined, and trials are powered to detect statistically meaningful clinical outcomes (e.g. survival, symptom improvement).
Given their size and complexity, Phase III trials are especially costly and risky. Only about 25–30% of drugs entering Phase III succeed (i.e. proceed to NDA/BLA) fda.gov. Major reasons for Phase III failure include insufficient efficacy (the drug fails to outperform control) or uncovering safety issues that were rare or unobserved in smaller trials. Because participants are exposed to the drug for longer periods, Phase III can reveal long-term side effects. A Phase III program may require multiple trials in different regions or subpopulations, each requiring regulatory approval and potentially adding years. For example, a single global oncology or cardiology Phase III program can easily take 3–5 years and involve coordination of hundreds of sites worldwide.
Like Phase II, Phase III is prone to trial delays. As one review notes, only 47% of studies complete enrollment on schedule, and over half require significant timeline extensions appliedclinicaltrialsonline.com. Complex endpoints (e.g. clinical outcomes in chronic diseases) and competition with other trials further challenge recruitment. Because Phase III results form the core of the approval submission, sponsors often invest heavily in data quality. All adverse events must be collected and analyzed meticulously for regulatory filings.
Clinical Trial Challenges
Across all trial phases, sponsors face several common obstacles. Designing optimal protocols is difficult: overly complex designs (multiple arms, biomarker stratification, frequent visits) increase cost and slow enrollment. Conversely, too-simple protocols risk missing critical efficacy or safety signals. Patient heterogeneity (differences in disease severity, comorbidities) can obscure treatment effects. In rare diseases or genetic conditions, small patient populations make enrollment very slow or sometimes impossible. The requirement for placebo control or multiple comparison groups (standard-of-care vs new drug) can deter patient participation. Trial amendments (mid-study protocol changes) are frequent and can add months to timelines. Overall, nearly 90% of trials experience delays or fail to meet initial timelines appliedclinicaltrialsonline.com, indicating how challenging clinical development can be.
Regulatory Review and Approval
Successful completion of Phase III enables submission of a marketing application. In the U.S., sponsors file a New Drug Application (NDA) for small-molecule drugs or a Biologics License Application (BLA) for biologics. In the EU, a centralized Marketing Authorization Application (MAA) is submitted to the European Medicines Agency (EMA) (or a national procedure for some products).
FDA Review (U.S.)
Under the Prescription Drug User Fee Act (PDUFA), the FDA has performance goals for review. Once an NDA/BLA is filed, the FDA must act by a set “PDUFA date,” typically 10 months after filing for standard review, or 6 months for priority review (granted to drugs addressing serious conditions) pharmacytimes.com. Priority or expedited pathways (Fast Track, Breakthrough Therapy, Accelerated Approval) exist to shorten timelines: for example, Accelerated Approval allows based on surrogate endpoints with post-marketing confirmatory trials. In practice, many new drugs (especially orphan or breakthrough therapies) benefit from these programs. As of 2024, ~66% of approved drugs used one or more expedited designation pharmacytimes.com.
During FDA review, multidisciplinary teams (clinical, statistical, pharmacology, CMC, etc.) evaluate the full NDA/BLA dossier pharmacytimes.com. If issues are found, the FDA issues a Complete Response Letter (CRL), requiring additional data or clarification, which can add months or years. Otherwise, the agency may approve the application. Overall, FDA review adds roughly 0.5–1 year to the timeline. Once approved, the FDA inspects manufacturing facilities to ensure cGMP compliance before or after approval. The FDA also regulates labeling and post-approval requirements (e.g. REMS safety programs).
EMA Review (EU)
In the EU, the centralized procedure through EMA leads to one marketing authorization valid in all member states. The Committee for Medicinal Products for Human Use (CHMP) conducts the scientific assessment. A standard EMA review takes up to 210 active days (excluding clock-stops for sponsor responses) to reach a CHMP opinion eurogct.org. A sponsor can request an Accelerated Assessment to shorten this to 150 days for medicines of major public health interest eurogct.org. After CHMP issues a positive opinion, the European Commission gives a final marketing authorization (usually within 1–2 months).
EMA also has special pathways: PRIME (PRIority MEdicines) for early support and Accelerated Assessment for faster review. For advanced therapies (gene/cell therapy products, an ATMP category), the Committee for Advanced Therapies (CAT) vets the quality/safety data before CHMP review, reflecting their complexity eurogct.org. Despite accelerated tracks, real-world MAAs often take 8–12 months from submission to Commission decision (210 days active + clock stops). Like the FDA, EMA may require additional post-approval studies and strictly enforces pharmacovigilance.
Manufacturing and Scale-Up
Before or during Phase III, the sponsor must scale up manufacturing from laboratory batches to commercial supply. Chemistry, Manufacturing and Controls (CMC) is a critical component of any regulatory submission. Manufacturers progressively develop and validate their production process through laboratory, pilot, and commercial scales seed.nih.gov. This process “scale-up” involves optimizing yields, purity, and reproducibility while complying with cGMP standards.
Small-molecule drugs (chemically synthesized) generally have fewer critical process steps, and processes are relatively well-defined. By contrast, biologics (proteins, antibodies, cell therapies) are produced in living systems (microbial or mammalian cell cultures) seed.nih.gov seed.nih.gov. Biologic manufacturing is inherently complex and variable, requiring large bioreactors, precise culture conditions, and extensive purification. For example, an antibody drug may involve weeks-long cell fermentations followed by multiple chromatography steps. Any change in the production process can alter the product (the “process is the product” for biologics) seed.nih.gov. Thus scale-up for biologics demands rigorous process characterization and validation (e.g. to control glycosylation patterns, viral clearance, aggregate formation).
Typically, sponsors establish a CMC plan outlining how they will scale from bench to commercial lot seed.nih.gov. Early-phase clinical supplies may come from pilot-scale facilities, but before approval a final commercial-scale process must be locked down. For small molecules, this might mean going from gram to multi-kilogram batches using industrial reactors. For biologics, it means scaling from lab flask to 1,000+ liter bioreactors. All batches must be shown to meet specifications for identity, strength, purity, and stability. The FDA and EMA will inspect manufacturing sites (often multiple global sites) for cGMP compliance. Completing scale-up and obtaining regulatory approval of the CMC package can add many months to the development timeline. In practice, manufacturing readiness is often on the “critical path”: any delays in process validation or regulatory compliance directly delay product launch.
Post-Approval Surveillance and Market Access
Even after approval, a drug’s development journey continues. Phase IV (post-marketing) studies and pharmacovigilance are mandated to monitor long-term safety and, in some cases, additional efficacy. Regulators recognize that pre-approval trials cannot uncover all adverse events. The FDA notes that the true safety profile of a drug “evolves over the months and even years” on the market fda.gov. Systems like FDA’s MedWatch/FAERS (or EMA’s EudraVigilance) collect spontaneous adverse event reports. Initiatives like FDA’s Sentinel use electronic health data to actively detect safety signals fda.gov. If serious risks emerge, regulators may require label changes, restricted distribution programs, or even withdrawal. Many drugs also undergo additional studies post-approval (e.g. Phase IV trials for special populations or long-term follow-up) as a condition of approval.
Meanwhile, commercial and market factors come into play. Patent protection and exclusivity clock starts at submission; in the U.S. innovators typically get 5 years of NCE exclusivity (plus any remaining patent life) and 7 years orphan exclusivity for rare diseases, while in the EU the data exclusivity is 8+2 years (with an optional year extension). Because patents are usually 20 years from filing, a long development timeline means less effective market exclusivity remaining. Companies must consider market size, competition (e.g. other drugs or generics), and pricing/reimbursement when investing in development. In fact, about 10% of development failures are attributed not to science but to commercial or strategic reasons (e.g. market shifts, funding shortfalls) pmc.ncbi.nlm.nih.gov. Health economics and payor negotiations increasingly influence late-stage trials (drug pricing expectations can shape trial designs and endpoints).
In summary, the path from lab discovery to patient use is long due to a sequence of scientific, regulatory, manufacturing and commercial steps biostock.se pmc.ncbi.nlm.nih.gov. Each stage – discovery, preclinical research, three clinical phases, regulatory review, and manufacturing – has its own timelines and failure modes. Even after approval, monitoring and market dynamics require time to ensure the drug’s safety and viability. Taken together, these factors explain why end-to-end drug development can take up to 20 years or more.
Special Considerations and Drug Class Differences
Development timelines and challenges vary by drug modality. Small-molecule drugs (conventional chemical compounds) have the most established pathways: their synthetic manufacture and oral dosing make scale-up relatively straightforward. Biologics (proteins, monoclonal antibodies, therapeutic enzymes) are more complex: they often require living cell culture production and sterile formulation, and they may provoke immune responses. As one review notes, biologics can be “vastly more complex” than small molecules, sometimes by three orders of magnitude in molecular size seed.nih.gov. This complexity means biologics often need longer process development and more stringent characterization. Nevertheless, modern biomanufacturing has accelerated; FDA approved ~56 biologics in 2022 (roughly 45% of approvals) pharmacytimes.com. Biologics also benefit from a similar regulatory process (BLA vs NDA) but typically incur longer CMC review due to their intricacy.
Gene therapies and advanced therapies (ATMPs) represent a new frontier. These treatments (viral gene vectors, CAR-T cells, gene-edited cells, etc.) often target rare, high-unmet-need diseases with one-time curative intent. The science is newer, so basic research gaps and clinical unpredictability add time. Regulatory agencies have established special pathways (FDA’s RMAT designation, EMA’s CAT and PRIME) to expedite these. For example, the FDA listed dozens of licensed cell and gene therapies as of 2024 fda.gov fda.gov, reflecting a growing pipeline. However, manufacturing is even more complex (viral vector supply, cell logistics) and long-term follow-up is needed to assess durability and safety (e.g. insertional oncogenesis). Consequently, clinical programs for gene therapies often involve small patient numbers (pediatric or rare-disease trials) and adaptive trial designs. Some gene therapies have been approved relatively quickly under accelerated programs (e.g. Luxturna for retinal disease had ~7 years from IND to approval), but generally these products still take many years to develop due to the novelty of the technology.
Generic drugs and biosimilars also impact the landscape. After patent expiry, generics (small molecules) or biosimilars (biologics) enter, greatly changing market dynamics. The prospect of generic competition can influence investment decisions, as companies aim to recoup R&D costs within the exclusivity window. Both FDA and EMA have detailed guidances on generics and biosimilars, but these are separate pathways outside the innovator’s development process.
Key Challenges and Risk Factors
Several recurring challenges add time and risk at each phase:
-
High Attrition: As noted, ~90% of drugs entering clinical trials fail pmc.ncbi.nlm.nih.gov. This attrition means many projects are dropped, wasting prior years of work. Discovery failures (no efficacy or untenable toxicity) often occur late, sending projects back to the lab or out of development entirely.
-
Trial Design and Recruitment: Crafting effective protocols is non-trivial. Overly restrictive criteria or complex endpoints slow recruitment, while too-broad trials risk ambiguous results. Patient recruitment is a chronic bottleneck: up to 20–30% of sites may enroll zero patients appliedclinicaltrialsonline.com, forcing sponsors to add sites or extend timelines. In global trials, cultural and regulatory differences across regions can further complicate enrollment.
-
Clinical Endpoints and Biomarkers: Especially in areas like oncology or neurology, choosing the right endpoints (survival vs surrogate markers) can lengthen trials. Recent efforts (e.g. FDA’s Project Optimus for oncology dosing) aim to optimize phase I/II design, but these are iterative advances.
-
Safety/Toxicity Surprises: Unanticipated toxicities can derail late-stage trials. For example, cardiac or liver toxicity not seen in animals may emerge in Phase II/III, forcing dose reductions or trial halts. Such findings require additional studies or reformulation, adding years.
-
Regulatory Hurdles and Variability: Different regions have different requirements (e.g. EMA’s inclusion of pediatric data, FDA’s REMS or black-box labeling). Harmonizing global trials can be slow. Regulatory reviews themselves sometimes take longer than goal dates, especially if agencies request major revisions.
-
Commercial Viability: Market factors can force program changes. For instance, if a competitor launches a similar drug while yours is in Phase III, the company might pivot or accelerate trials to maintain competitiveness. Reimbursement pressures (payers demanding evidence of cost-effectiveness) may also require added health-economic studies.
These challenges collectively ensure that drug development is not a linear “light-speed” process. At each stage, months can pass refining studies, waiting for patient accrual, and negotiating with regulators. Since even small delays compound, a 10‑15 year timeline is not unusual. Longer cases (up to 20+ years) often involve novel science or repeated setbacks.
Conclusion
In summary, the decade(s)-long timeline for drug development reflects the scientific depth and regulatory rigor of the process. From basic research (years spent understanding biology and identifying targets) through preclinical safety testing and three phases of clinical trials, to regulatory review and manufacturing scale-up, each step is time‑intensive. Regulatory agencies (FDA, EMA) add structured review intervals (often 6–12 months each) pharmacytimes.com eurogct.org, and sponsors must compile vast data packages to demonstrate quality, safety and efficacy. Common hurdles – trial recruitment delays, high attrition, and complex production – further lengthen the schedule. All told, most new drugs require 10–15 years (or more) from discovery to approval efpia.eu biostock.se. Only by successfully navigating every phase (with its inherent risks of toxicity, lack of efficacy, or commercial failure) can a drug finally reach the market and benefit patients.
Sources: Drug development timelines and statistics are drawn from industry and regulatory sources efpia.eu biostock.se pmc.ncbi.nlm.nih.gov appliedclinicaltrialsonline.com. Regulatory processes are summarized from FDA and EMA guidance pharmacytimes.com eurogct.org. Manufacturing considerations are based on FDA CMC guidelines seed.nih.gov seed.nih.gov. Clinical trial challenges and attrition rates are supported by literature pmc.ncbi.nlm.nih.gov appliedclinicaltrialsonline.com. All data are current as of 2024–2025.
DISCLAIMER
The information contained in this document is provided for educational and informational purposes only. We make no representations or warranties of any kind, express or implied, about the completeness, accuracy, reliability, suitability, or availability of the information contained herein. Any reliance you place on such information is strictly at your own risk. In no event will IntuitionLabs.ai or its representatives be liable for any loss or damage including without limitation, indirect or consequential loss or damage, or any loss or damage whatsoever arising from the use of information presented in this document. This document may contain content generated with the assistance of artificial intelligence technologies. AI-generated content may contain errors, omissions, or inaccuracies. Readers are advised to independently verify any critical information before acting upon it. All product names, logos, brands, trademarks, and registered trademarks mentioned in this document are the property of their respective owners. All company, product, and service names used in this document are for identification purposes only. Use of these names, logos, trademarks, and brands does not imply endorsement by the respective trademark holders. IntuitionLabs.ai is an AI software development company specializing in helping life-science companies implement and leverage artificial intelligence solutions. Founded in 2023 by Adrien Laurent and based in San Jose, California. This document does not constitute professional or legal advice. For specific guidance related to your business needs, please consult with appropriate qualified professionals.