Clinical Trial Phases: A Guide to Phase I, II, III & IV

The 4 Phases of Clinical Trials: A Deep Dive into Phase I–IV
Executive Summary
Clinical trials are the scientific cornerstone of modern medicine, providing the evidence base for the safety and efficacy of new drugs and therapies. They are traditionally organized into four successive phases – I, II, III, and IV – each with distinct objectives, designs, and regulatory roles. Phase I trials represent the first human tests of a new investigational drug, focusing primarily on safety, tolerability, and pharmacokinetics in a small group (typically 20–100) of healthy volunteers or sometimes patients ([1]). Phase II trials expand to several hundred patients with the target disease, aiming to obtain preliminary efficacy and further safety data under controlled conditions ([2]). Phase III trials are large-scale (hundreds to thousands of subjects) randomized controlled trials that definitively establish clinical efficacy and monitor infrequent adverse events; these studies form the core of data for regulatory approval ([3]). Following market authorization, Phase IV (post-marketing) studies collect real-world data on long-term safety, effectiveness, and off-label use in broader populations ([4]) ([5]).
Despite rigorous testing, drug development has a high attrition rate. Only a small fraction of compounds that enter human trials ultimately gain approval. Recent analyses indicate overall success rates (Phase I through approval) on the order of 5–10% for industry-sponsored programs ([6]). In practical terms, roughly one in ten investigational drugs that begin Phase I will eventually receive FDA approval. Much of the attrition occurs in Phase II: many candidate drugs that show safety in early trials fail to demonstrate adequate efficacy or acceptable safety in patients. These statistics underscore the challenge of translating preclinical promise into clinical benefit.
The four-phase model has evolved over decades in response to scientific, ethical, and regulatory developments. Its history spans from early antiseptic trials and the first randomized controlled trials of the mid-20th century, through the post–Thalidomide regulatory overhaul of the 1960s, to the modern era of globalized, highly regulated drug development. Regulatory agencies such as the U.S. FDA and EMA provide formal definitions and guidance for each phase, shaping trial design and execution. For example, the FDA defines Phase I studies as “the initial introduction of an investigational new drug into humans…designed to determine” metabolic action, dose, and side effects ([1]), while Phase IV studies are explicitly tied to post-approval surveillance and market experience ([4]).
This report provides an in-depth exploration of each clinical trial phase from multiple perspectives. It examines the scientific aims, methodological features, and typical scale of Phase I–III trials, and the role of Phase IV research in the life cycle of a medicine. It analyzes current data on trial success rates, sample sizes, and timelines. Ethical considerations and regulatory frameworks for each phase are discussed, as are emerging trends such as adaptive and seamless trial designs that blur traditional phase boundaries. Detailed case studies illustrate key points: for instance, first-in-human trials in oncology often enroll advanced cancer patients rather than healthy volunteers, and a notorious Phase I trial (TGN1412) dramatically highlighted the need for caution in dosing new biologics. Another case highlights how overlapping trials expedited COVID-19 vaccine development under Emergency Use protocols. The report also examines specialized topics like conditional approvals, orphan drugs, and global harmonization of trial standards. Data tables summarize stage-specific attributes (objectives, population, size, endpoints) and major regulatory milestones.
By integrating historical context, quantitative evidence, and expert insights, this comprehensive review elucidates how the four phases of clinical trials function individually and together. It highlights the critical importance of each phase in ensuring that new therapies are both effective and safe. Finally, it considers future directions: innovations such as digital health data, patient-reported outcomes, and platform trials promise to make clinical research faster and more inclusive, but also require careful methodological adaptation. The conclusion synthesizes the implications for drug developers, regulators, healthcare providers, and patients. All claims and data herein are supported by authoritative sources from regulators, peer-reviewed journals, and industry analyses ([1]) ([4]) ([6]) ([7]).
Introduction and Background
Randomized controlled clinical trials (RCTs) have roots in antiquity (e.g. James Lind’s scurvy trial in 1747) and were formalized in modern medicine during the 20th century (for example, the 1948 streptomycin trial for tuberculosis) ([8]). However, the structured system of four sequential phases for drug trials emerged as regulatory scrutiny increased. In the U.S., key legislation shaped this system. The 1938 Federal Food, Drug, and Cosmetic Act first required proof of safety before marketing; the 1962 Kefauver–Harris Amendments then mandated substantial evidence of efficacy through “adequate and well-controlled investigations” – essentially embedding Phase III-scale RCTs into law ([9]). Worldwide, harmonization efforts (e.g. the International Council for Harmonisation, ICH) have since standardized requirements. For example, ICH E6 (Good Clinical Practice) guides the design/conduct of trials across all phases, ensuring ethical principles and quality standards in human research ([10]).
Under the predominant model, clinical development of a new drug proceeds through four phases:
-
Phase I: First-in-human trials (FIH) for safety and dose-finding. Small cohorts (often 20–100 subjects) are given single or escalating doses. Traditionally performed in healthy volunteers (unless the drug is too toxic, e.g. chemotherapy in cancer patients). The focus is on adverse effects, pharmacokinetics (absorption, metabolism, etc.), and identifying a safe dose range ([1]).
-
Phase II: Exploratory efficacy trials in patients. Several hundred subjects with the target disease receive the investigational drug (and often comparator arms or placebo). Primary goals include preliminary assessment of clinical efficacy signals and continued safety/tolerability evaluation ([2]), often across multiple dose levels or doses. Phase II may be split into IIa (pilot efficacy) and IIb (dose-ranging) in some terminologies.
-
Phase III: Confirmatory trials to establish definitive efficacy and safety for regulatory approval.Large sample sizes (hundreds or thousands) allow statistical power to detect treatment effects and rarer side effects ([3]). These are typically randomized, controlled, multicenter trials. Success in Phase III is needed to file a New Drug Application (NDA) or Biologics License Application (BLA) with the FDA (or equivalent submissions abroad).
-
Phase IV: Post-approval (post-marketing) studies. These monitor the drug’s performance in real-world clinical practice. This phase can include well-organized observational studies, registries, long-term safety monitoring, and trials in new populations or indications. Importantly, Phase IV captures rare adverse events and long-term outcomes that may not be evident in pre-approval trials ([4]) ([5]). For example, a safety signal detected in Phase IV might trigger a “Dear Doctor” letter, a label change, or even withdrawal (as with rofecoxib/Vioxx in 2004).
The four-phase structure is not an immutable law; overlap and exceptions occur. For instance, adaptive or “seamless” trial designs combine phases (e.g. an open-label dose-escalation Phase I/IIa in oncology). Accelerated pathways (Fast Track, Breakthrough, etc.) can compress transitions between phases for serious diseases ([7]). Nevertheless, the Phase I–IV language remains the lingua franca of clinical research, reflecting increasing scale and maturity of evidence.
Table 1 (below) summarizes core features of each phase. Key points include the shift from safety-focused, healthy-subject trials in Phase I to efficacy-focused, patient-centered trials in Phases II/III, then to broad surveillance in Phase IV ([1]) ([4]).
| Phase | Typical Participants | Primary Objectives | Typical Size (Subjects) | Key Endpoints / Outcomes |
|---|---|---|---|---|
| I | Healthy volunteers (or very ill patients in oncology/toxo cases) ([1]) | Assess safety, tolerability, pharmacokinetics/dynamics, maximum tolerated dose ([1]) | ~20–100 (dose-escalation cohorts) ([1]) | Safety/adverse events; dose levels; PK/PD parameters |
| II | Patients with target disease conditions | Evaluate preliminary efficacy; further evaluate safety and dosing ([2]) | 100s of patients (multiple groups/doses) ([2]) | Efficacy signals (biomarkers/clinical outcomes); adverse effects |
| III | Patients with target disease (confirmed by diagnosis) | Confirm efficacy and safety on a large scale; support labeling ([3]) | Hundreds to several thousands ([3]) | Clinical efficacy (primary endpoints), safety in diverse patients |
| IV | Broad real-world patient population | Monitor long-term safety; assess effectiveness, compliance, new uses ([4]) ([5]) | Variable (post-marketing studies or registries) | Rare adverse events, long-term outcomes, off-label insights |
Table 1. Summary comparison of Clinical Trial Phases I–IV. (Regulatory guidance: FDA/CDER Phase definitions ([1]); Phase IV as real-world surveillance ([4]).)
Historical Context
The rationale for sequential clinical phases arises from both science and tragedy. In the early 20th century, clinical research was unstructured: therapies often entered practice based on anecdote. The streptomycin trial (1948) and polio vaccine trials (1950s) demonstrated the power of randomized evidence. However, adverse events such as the thalidomide disaster (early 1960s) – where a sedative given to pregnant women caused thousands of birth defects – spotlighted the need for systematic safety and efficacy testing. In response, the 1962 Kefauver–Harris amendments in the U.S. required that manufacturers prove a drug’s effectiveness before marketing through “adequate and well-controlled” trials ([9]). Over ensuing decades, this global shift formalized regulators’ expectation that testing proceeds from small safety studies to large efficacy trials (essentially Phases I–III) before approval is granted. Phase IV arose more informally: once a product is on the market, regulators and sponsors continue to collect data on long-term or rare risks that were not fully captured pre-approval ([5]).
The phases also reflect ethical progression. Injecting new drugs first into healthy volunteers (Phase I) ties to minimal risk principles, whereas later trials in patients (Phase II/III) justify higher risk by potential benefit. Oversight by Institutional Review Boards (IRBs) and regulatory bodies (FDA, EMA, etc.) ensures each trial phase balances risks and benefits. modern guidelines like ICH-E6 (Good Clinical Practice) codify these protections across all phases ([10]).
The Broad Role of Phases in Drug Development
Each phase serves a gatekeeping function, pressing candidate therapies for increasingly stringent evidence. After preclinical (animal/lab) work establishes basic safety and activity, Phase I is the first human test. If Phase I is tolerable and identifies useful dosing, Phase II tests whether the drug likely works in patients. Failure at Phase II often halts a program (lack of efficacy or unacceptable toxicity is common). Succeeding Phase II, a robust Phase III trial is run to “confirm” that efficacy and safety are sufficient for labeling. Only after positive Phase III (and sometimes additional efficacy studies) will a regulatory submission proceed. Even after approval, Phase IV and pharmacovigilance continue to refine the understanding of the drug. Thus, the four-phase model is the framework through which nearly all new drugs (and many biologics) must pass on their path to patients.
Figure 1 (see below) diagrams the typical flow of drug candidates through the phases, including attrition and timelines. The figure highlights that the probability of success declines at each transition. For example, industry studies estimate only ~47–60% of drugs in Phase II proceed to Phase III, and only ~50–70% of Phase III programs lead to approval ([11]) ([6]). The overall chance that a drug entering Phase I reaches the market is often cited around 10% ([6]) ([12]). These attrition figures underscore why the drug development process is long, costly, and risky.
Phase I: First-in-Human Studies
Purpose and Design. Phase I trials are the first exposure of humans to an investigational drug, typically following extensive animal safety testing. The primary goal is to characterize safety and tolerability, usually by gradually increasing the dose until side effects limit further escalation. Regulators expect sponsors to justify the initial human dose (often using the “No Observed Adverse Effect Level” in animals) and to set clear stopping rules ([13]). Phase I often includes pharmacokinetic (PK) and pharmacodynamic (PD) measurements: how the body handles the drug and how the drug affects biomarkers or physiology. In some cases (e.g. oncology, gene therapies), Phase I may also seek any indication of biological activity (efficacy).
Traditionally, most Phase I trials are conducted in healthy volunteers. The logic is to isolate the drug’s basic behavior without confounding disease. Typical Phase I protocols use a “single ascending dose (SAD)” design (one group gets a low dose; if safe, the next group gets a higher dose) followed by a “multiple ascending dose (MAD)” phase where repeated dosing is tested. The total number of subjects in a Phase I study is generally small – often between 20 and 80 participants ([1]). Table 2 (later) provides typical parameters for reference. The cohort sizes are usually around 4–10 volunteers per dose level. Endpoints are mainly PK parameters (C_max, AUC), vital sign changes, lab values, and adverse events. The dose at which consistent toxicity or intolerability is seen defines the maximum tolerated dose (MTD), guiding recommended Phase II dosing.
In some therapeutic areas, Phase I trials differ in design. Oncology drugs (especially cytotoxic or targeted anticancer agents) may be given first to cancer patients rather than healthy people, because of the high risk and potential direct benefit. These “first-in-human” oncology trials often enroll patients with advanced, refractory cancers. Such Phase I oncology protocols may combine safety assessment with initial efficacy signals (e.g. tumor response), blurring into Phase II objectives ([14]) ([15]). For example, UGT1a-targeted drug trials might concurrently increase dose and monitor tumor shrinkage.
Ethical Considerations. Phase I studies raise particular ethical issues. Participants (especially healthy volunteers) derive no therapeutic benefit but face unknown risks ([16]). They are often motivated by payment, raising concerns about undue inducement. Research shows Phase I cohorts frequently include disproportionately high numbers of economically disadvantaged minorities ([16]), underscoring vulnerabilities. Ethical oversight (IRB review, informed consent, risk minimization) is critical. Contemporary discussions emphasize fair compensation, clear communication of novel risks, and rigorous monitoring to protect these participants ([16]).
Paradoxically, even though Phase I trials enroll few subjects, unforeseen outcomes can be dramatic. The 2006 TGN1412 case is instructive: an investigational monoclonal antibody (anti-CD28) was given to six healthy volunteers. All six experienced a fulminant “cytokine storm” within hours, becoming critically ill ([17]). This led to life-threatening complications (two required prolonged intensive care) and illustrated the limits of animal testing’s predictive power. Such events have prompted guidelines (e.g. the EMA’s revised “First-in-Human” guidance ([13])) that tighten risk assessment and sentinel dosing strategies.
Implementation and Current Trends. Phase I trials are typically single-center or small multi-center studies. The investigator and sponsor must prepare an Investigational New Drug (IND) or Clinical Trial Application (CTA) submission. Incremental continuation of dosing (e.g. from cohort to cohort) depends on review of accumulating data. Increasingly, designs incorporate modern tools: modeling and simulation (PK/PD models) to optimize dose escalations, biomarkers to de-risk failure, and in some cases accelerated titration (giving more rapid dose increments). There is also a movement toward “Phase 0” or exploratory IND trials with microdosing (truly tiny doses to gather PK data without expecting effect); these are outside the traditional I–IV, but have been explored in oncology ([18]).
Case Study – Oncology Phase I. Oncology Phase I trials differ significantly from typical Phase I. An example is a first-in-human study of an oncolytic virus given to patients with advanced solid tumors ([17]) – illustrating that oncology Phase I can involve severely ill patients. Oncologists often use such trials to assess tolerability and any anti-tumor activity. The MDICT guidelines (2022) for Phase I oncology reflect this, recommending integrated designs (SAD + MAD) and careful safety monitoring ([19]). In practice, oncology Phase I protocols may enroll 20–50 patients, escalating doses cautiously. If partial responses are seen at a tolerated dose, that dose may be taken forward into Phase II.
Despite the small scale, Phase I provides critical go/no-go decisions. For example, if a novel drug shows severe toxicity at low doses (as in the TGN1412 trial ([17])), further development may be halted or reformulated. Conversely, a clean Phase I safety profile with promising biomarker effects can support expensive Phase II work.
Phase II: Dose-Finding and Proof-of-Concept Studies
Goals and Types. Phase II trials are often called “proof-of-concept” studies. Their aim is to evaluate whether the drug appears to work in patients and to further characterize safety in the target population. These trials provide the first controlled evidence of efficacy, albeit in a limited number of patients. They help answer: “Does this drug have enough biological effect to justify a large Phase III trial?” and “What dose/regimen is most promising?”
Phase II is sometimes subdivided: Phase IIa (pilot studies, maybe uncontrolled or open-label) focused on biological activity, and Phase IIb (placebo- or active-controlled dose-ranging studies) focused on determining optimal dosing. For example, several parallel arms might receive different dose levels to select the best candidate dose for Phase III. In any case, Phase II involves patients (unlike healthy volunteers) and typically uses intermediate or surrogate endpoints (like tumor shrinkage, viral load reduction, symptom scores) instead of hard clinical outcomes.
Design Considerations. By this point, the design often becomes randomized and sometimes blinded, to reduce bias. Classic Phase II designs include single-arm trials (especially in oncology where tumor reduction can be measured against historical control) or small randomized trials (e.g. drug vs. placebo or dose A vs. dose B). The sample size is usually in the low hundreds, often determined by feasibility rather than full statistical power. However, biostatisticians increasingly use more formal designs: randomized Phase II with power estimates, two-stage designs (stopping early for futility), or adaptive randomization. In oncology, for instance, the “Simon two-stage” design is popular for early signals (stop if insufficient responses).
Key endpoints are chosen to gauge whether larger trials are warranted. Often these are intermediate efficacy measures or composite safety-efficacy scores. Secondary data may include biomarker changes, pharmacodynamics, and detailed adverse event collection. Phase II also refines inclusion/exclusion criteria, methodologies, and feasibility assessments for Phase III plans.
Because Phase II bridges early safety to late confirmatory trials, its failure rate is high. Many drug candidates “fail” at Phase II due to lack of clear efficacy or unacceptable side effects in patients that were not seen in Phase I. One industry report noted “Phase II remains one of the most challenging steps in clinical drug development” ([20]), in part because effects there must be large enough to pursue.
Case Study – Single-Arm vs. Randomized Phase II. Historically, many Phase II trials, especially in oncology and rare diseases, have been single-arm (all patients get the drug) and use historical controls for comparison. However, this approach risks false positives due to selection biases. Recent guidance in oncology encourages at least some controls even in Phase II ([21]). For example, a Phase II cancer drug trial might randomize patients 2:1 to drug vs. standard-of-care, allowing an internal control and more reliable efficacy estimate before proceeding to Phase III. In non-oncology fields, randomized Phase II is also common. For a new migraine medication, for instance, a Phase II trial might randomize 100 patients to low dose, high dose, or placebo to verify that the doses improve monthly migraine days over placebo.
Phase II in the Regulatory Context. While Phase II trials alone are not sufficient for approval, positive Phase II results can sometimes be used for accelerated pathways. For example, in serious or unmet-need conditions, a strong Phase II outcome might justify a conditional approval with the commitment of further Phase III or Phase IV data. Additionally, “Phase II” trials may include exploratory objectives like identifying biomarkers of response for later trials.
Phase III: Pivotal Confirmatory Trials
Purpose and Scale. Phase III trials are the linchpin of drug development. Their goal is to confirm that a drug is effective and safe for its intended indication in a large, representative patient population. Due to their pivotal nature, Phase III studies are typically randomized controlled trials (RCTs) with pre-specified primary clinical endpoints (e.g. survival, disease recurrence, symptom improvement rates) that directly support labeling. They usually involve hundreds to thousands of patients across multiple sites (often multinational) to ensure geographic and demographic diversity.
Given their size and cost, Phase III trials are usually funded and organized by pharmaceutical companies and require extensive planning: protocol development, site selection, randomization systems, data monitoring committees for safety, and so on. They must adhere to strict statistical requirements for demonstrating efficacy. For instance, if the primary endpoint is the reduction of HbA1c in diabetes, the trial must show a statistically significant and clinically meaningful improvement over control (usually placebo or existing therapy) at a predetermined alpha (commonly 0.05) and power (80–90%).
Typical Phase III trials compare the new drug to the best available standard of care, or placebo if no standard exists. Many modern Phase III studies are event-driven: they continue until a target number of outcome events (e.g. heart attacks, cancers) occur. Sample size is thus a function of expected effect size and outcome incidence. As a rough guide, cardiovascular outcomes trials enrollment often exceeds 5,000 patients, whereas many Phase III oncology trials enroll 300–1,000. The heterogeneity of modern drug classes means there is wide variability: a rare disease Phase III may only have 100 patients worldwide, whereas a blockbuster indication trial could exceed 10,000.
Success and Failure Modes. Succeeding in Phase III means demonstrating a favorable benefit-risk profile. Regulatory agencies expect that Phase III primary endpoints are met in a robust manner. Failure modes include: lack of efficacy (most common), unexpected toxicity, or manufacturing/quality issues. Sometimes a drug that looked promising in Phase II fails in Phase III. For instance, a therapy for Alzheimer’s might show cognitive score improvement in small trials but then fail to affect clear endpoints in a large, longer trial. These failures incur huge costs – a single Phase III program can cost hundreds of millions.
Conversely, successful Phase III results allow a company to file for approval. If the data convincingly show safety and efficacy, regulators (like the FDA) may approve the drug for marketing. The companies then present the entire package (Phase I–III data) in an NDA or BLA submission.
Global and Regulatory Variations. Phase III designs often account for global differences. Many Phase III trials are now multinational, enrolling in the US, Europe, Asia, etc., to meet multiple regulatory authorities’ requirements in parallel. Regulatory guidelines for Phase III differ slightly: for example, the FDA often expects two adequate and well-controlled Phase III trials (or one very large trial), while the EMA may accept evidence from a single large trial plus a supporting study. Special designations can modify how Phase III is done. Fast Track or Breakthrough designation can allow rolling submissions; Priority Review or Accelerated Approval may accept surrogate endpoints for serious conditions (e.g. tumor shrinkage as a surrogate for survival in cancer).
Phase III also now often involves advanced elements: adaptive designs, where interim analyses may allow changes (e.g. sample size re-estimation); platform trials testing multiple therapies under one protocol (e.g. I-SPY in cancer); noninferiority trials (showing new drug is not worse than standard by a margin, often used when placebo is unethical); and pragmatic trials that mimic clinical practice settings. For example, in pain management, a pragmatic Phase III trial might use broad inclusion and real-world comparators. All such designs must balance flexibility with pre-specification to avoid bias.
Case Study – COVID-19 Vaccine Trials. A recent high-profile example of Phase III innovation was the development of several COVID-19 vaccines in 2020–21. Under tremendous public health urgency, developers overlapped and accelerated phases. Phase I/II combined safety/immunogenicity in small cohorts was quickly followed by large Phase III RCTs (some enrolling 30,000+ participants) ([7]). Regulatory agencies (like FDA) granted Emergency Use authorizations based on Phase III results much faster than normal. Notably, manufacturers like Pfizer-BioNTech and Moderna produced vaccine doses “at risk” before their Phase III trials were complete ([7]). These trials required tens of thousands of subjects and used primary endpoints of confirmed COVID-19 cases. The success of these trials (showing ~95% efficacy) within 12 months was unprecedented, demonstrating how crisis conditions can compress and accelerate the traditional Phase I–III timeline while still adhering to core principles. ([7])
Phase IV: Post-Marketing Surveillance and Beyond
Purpose of Phase IV. After regulatory approval, a new drug enters broader use in the general population. Phase IV (post-marketing) activities are crucial to monitor the drug’s performance outside the controlled confines of a trial. As one authority notes, “ [n]ot all Phase IV studies are PMS but every PMS [post-marketing surveillance] study is a Phase IV study.” In practice, Phase IV encompasses any research conducted after approval, including both observational and interventional studies ([4]).
The main objectives are:
-
Long-term Safety Monitoring: Clinical trials usually involve a few thousand patients at most and relatively short follow-up (often <2 years). Many adverse effects (especially rare events or those arising from chronic use) will only appear after millions have taken the drug. Phase IV pharmacovigilance (spontaneous adverse event reporting, registries) is essential to capture these. For example, serious liver toxicity or cardiac risk might only become evident post-approval. The famous example of rofecoxib (Vioxx) illustrates this: after FDA approval based on Phase III data, post-marketing reports and a pooled meta-analysis eventually showed increased heart attack risk, leading to withdrawal ([22]) ([23]). Not all drugs fare so poorly, but continued surveillance is mandatory.
-
Effectiveness in Real World: There can be a gap between efficacy (performance in trials) and effectiveness (performance in routine practice). Phase IV studies – often non-randomized cohorts or pragmatic trials – assess how a drug works in more diverse, comorbid, or adherent populations. This reflects the commentary that Phase IV is “the real test… the drug is tested in the real world” ([24]). For instance, a blood pressure drug approved in trials might be studied in Phase IV for long-term outcomes in elderly or pediatric patients.
-
New Indications and Comparative Use: Phase IV can open new uses. Observational data may suggest benefit in non-approved conditions, leading to formal trials (and possibly supplemental approvals). Sometimes head-to-head trials with competitors are done post-approval. Registries of special populations (e.g. pregnant women, patients with organ failure) are common Phase IV endeavors.
-
Regulatory Commitments: Often, regulators will require Phase IV studies as a condition of approval – a risk management plan (RMP) in Europe or a REMS in the U.S. These studies might focus narrowly, such as a long-term safety study mandated for a potential carcinogen.
Methods of Phase IV. Unlike pre-approval phases, Phase IV typically involves large observational cohorts, disease registries, and sometimes additional controlled trials. For example, a new epilepsy drug might have a Phase IV registry tracking thousands of patients for seizure control and side effects over years. Passive surveillance (e.g. FDA’s FAERS or WHO’s VigiBase) collects spontaneous reports of adverse events worldwide. Database studies (insurance or electronic health records) may be used to study drug utilization and outcomes. If still needed, post-approval trials (so-called Phase IV trials) can also be randomized, but often they are less controlled. For instance, many vaccines have Phase IV trials to monitor long-term immunity or rare adverse events in thousands post-licensure.
Crucially, Phase IV “never ends” as long as the product is marketed ([25]). Safety monitoring continues through advertising, scientific publications, and even patient feedback. Electronic systems (like FDA’s Sentinel) now proactively mine health records for safety signals.
Implications of Phase IV. Because Phase IV data often prompt label changes, dose adjustments, or usage restrictions, they can be contentious. Pressure to reveal honest post-market findings can clash with commercial interests ([23]). There have been cases where adverse outcomes in Phase IV prompted lawsuits or black-box warnings. Effective Phase IV relies on transparency (e.g. doctors and patients report adverse events), robust data infrastructure, and independent analysis. Recent innovation includes using “real-world evidence” (RWE) analysts to supplement or sometimes substitute for RCTs, but rigorous methodology remains vital.
Clinical Trial Pathways and Regulatory Context
While the phase framework is global, key differences exist in how it operates under different regulatory regimes. Here we highlight some overarching regulatory considerations:
-
U.S. FDA Regulations: In the U.S., the FDA’s Center for Drug Evaluation and Research (CDER) and Center for Biologics (CBER) oversee drug trials. Any trial in the U.S. generally requires an IND (Investigational New Drug application). The FDA defines Phase I–III trials (as per [21]) and has guidance documents for each stage. For instance, FDA guidance for “Phase I cGMP” and “Expanded Access” outline how to proceed. The FDA can allow “accelerated” approvals (using surrogate endpoints) in serious diseases, meaning that confirmatory Phase IV studies are needed as a condition ([7]).
-
European Medicines Agency (EMA): The EMA coordinates drug approval in the EU. Clinical trials must follow the EU Clinical Trials Regulation and Good Clinical Practice standards. The EMA publishes scientific guidelines including specific advice (e.g. on FIH trial design ([13])). Conditional marketing authorization in the EU also relies on post-authorization studies. EMA’s new EU Clinical Trial Regulation (effective 2022) streamlines multinational trials, impacting how Phases II-III are often run.
-
Other Jurisdictions: Japan (PMDA), Canada (Health Canada), China (NMPA), and others have analogous structures. For example, China has been reforming its approval process, including creating an expedited review pathway; recent analyses show Chinese Phase I trials have historically lower progression rates (as low as 20% in 2016–2020) ([26]), perhaps reflecting different risk approaches or emerging infrastructure.
-
International Harmonization: ICH guidelines (e.g. E8, E6, E9) provide a common framework. ICH E8 (R1) emphasizes “quality by design” for trials (including all phases) and risk-based approaches. A new revision E6(R3) is being implemented globally to modernize GCP.
-
Ethics and IRBs: Regardless of phase, all trials must obtain ethical review and informed consent. Early phases often have added protections (e.g. sentinel dosing or staggered enrolment). The Common Rule (US), Declaration of Helsinki, and local laws apply throughout.
Current Trends and Innovations
Traditional Phase I–III models are evolving. Key current trends include:
-
Adaptive and Seamless Designs: Trials that adapt in response to interim data are increasingly common. Examples include umbrella and basket trials in oncology, which test multiple drugs or indications under one protocol (e.g. NCI MATCH, Lung-MAP). Some trials combine Phase II and III (a single protocol with an interim look that can lead to expansion to pivotal Phase III enrollment). The COVID-19 vaccine efforts mentioned earlier exemplify this: Phase I/II → interim analysis → immediate transition to an expanded Phase III ([7]), all within one rolling development.
-
Decentralized and Digital Trials: Techniques like telemedicine, remote monitoring devices, and electronic patient-reported outcomes are being integrated at all phases. For example, a recent trend is “microtrials” or “phase 0” which use very low doses and modern bioanalytics to gather PD data in <10 subjects (still human trials, but pre-Phase I in the classic sense). Wearable biosensors can enable safety monitoring outside the clinic.
-
Personalized Medicine and Biomarkers: Precision medicine (targeted oncology, genetic disorders) means that even large Phase III trials may rely on biomarker-selected populations. Regulatory guidance now often links trial phase definitions to companion diagnostics (e.g. testing only patients with a mutation). This can shrink Phase II/III sizes but increase complexity.
-
Artificial Intelligence & Trial Optimization: AI and ML are being applied to simulate virtual trials, optimize trial design (predict dropout, find eligible patients), and analyze imaging or genomic data. While still maturing, these tools promise to speed dose-finding in Phase I or patient selection in Phase III.
-
Real-World Evidence (RWE): Due to Phase IV data growth, regulatory agencies are exploring use of RWE from electronic health records or claims to support label expansions or confirm trial findings. The 21st Century Cures Act in the U.S. legally encourages RWE use. This could blur the line between Phase III and Phase IV evidence.
-
Global and Ethical Shifts: There is growing emphasis on diversity (ensuring trials represent women, minorities, elderly). Regulators now often require African American or Asian representation commensurate with disease prevalence. Patient groups have more influence on trial design (patient-focused endpoints).
Career Case Studies and Real-World Examples
To illustrate these concepts, below are representative case studies from each phase:
-
Phase I – The TGN1412 Catastrophe: As mentioned, six healthy volunteers given a superagonist anti-CD28 antibody (TGN1412) in a Phase I trial all developed life-threatening systemic inflammatory syndrome ([17]). This event in 2006 impacted guidelines worldwide. Ministries of health mandated sentinel dosing (only one participant dosed first) and stricter monitoring in FIH trials. It exemplifies how Phase I vigilance is paramount.
-
Phase II – Alzheimer’s Drug Failures: A series of Phase II trials of amyloid-targeting drugs (e.g. solanezumab, aducanumab initial studies) showed disappointing efficacy. These Phase II results (lack of clear cognitive improvement) led companies either to halt programs or require even larger Phase III (often with biomarker endpoints). Some drugs, like aducanumab, eventually sought accelerated approval under FDA’s “surrogate endpoint” rules, based largely on Phase II amyloid reduction. These controversial cases highlight the high stakes and complex decision-making in Phase II for chronic diseases.
-
Phase III – KEYTRUDA in Melanoma: The PD-1 inhibitor pembrolizumab (Keytruda) was tested in pivotal Phase III trials enrolling ~800 patients with advanced melanoma. The RCT showed unprecedented survival benefits over chemotherapy. This trial’s success (and similarly nivolumab’s) ushered in a new era of immunotherapy. These Phase III programs (randomized, multicenter, placebo-controlled) set a gold standard for demonstrating a transformative clinical benefit, leading to approval and changed standard of care.
-
Phase IV – Rofecoxib (Vioxx) Withdrawal: Merck’s anti-inflammatory drug rofecoxib received FDA approval in 1999 after Phase III trials. However, early post-marketing surveillance (Phase IV) noted excess heart attacks. Meta-analyses published in 2003–2004 confirmed increased cardiovascular risk ([22]). By September 2004 the drug was pulled. This case underscores that Phase IV safety data can dramatically reverse earlier conclusions. It also ignited debate on trial transparency (a BMJ editorial argued companies should disclose adverse events before approval ([23])).
-
Seamless Phase I/II – Oncology Basket Trials: NCI’s MATCH trial assigns patients to treatments based on tumor genomics. It accommodates dose-escalation (Phase I) and signal-finding (Phase II) arms under one umbrella protocol. Similarly, the PANDA trial in autoimmune diseases uses adaptive features across phases. Such designs reduce the need to restart from scratch between phases, exemplifying modern flexibility.
-
Emergency Use Authorization – COVID-19 Vaccines: As earlier noted, in 2020 Pfizer-BioNTech, Moderna, and others shifted rapidly through Phases I–III. Preliminary data from Phase I/II mRNA vaccine trials (safety and neutralizing antibody titers) led directly to Phase III trials with 30,000+ participants ([7]). The vaccines showed ~95% efficacy within a few months. This unprecedented acceleration was possible because of prior knowledge of the SARS-CoV spike protein, massive funding (Operation Warp Speed), and rolling regulatory review. It illustrates how external pressures (pandemic) can overlay on the four-phase model.
These examples demonstrate the unity and diversity of how phases function. They also show that real-world events (disease outbreaks, scientific breakthroughs, or tragedies) can prompt adjustments in how phases are implemented.
Data and Analysis
Attrition and Success Rates
Quantifying success rates at each phase has been a major focus for industry analysts. Published reports (e.g. by Biotechnology Innovation Organization, Biomed trackers, etc.) have long documented attrition: historically only ~11% of drugs entering Phase I reached approval. Recent dynamic analyses show changing trends: an analysis of 9,682 molecules (2001–2023) found that overall success (approval) remained low (around 5–12%, depending on sponsor and time window) ([27]) ([6]). Breaking this down by stage, the drawn conclusions include:
-
Phase I to II (P1SR): Current estimates put this around 50–60% (i.e. roughly half of phase I programs progress) ([11]). This rate has declined over time (it used to be ~70% in the early 2000s, now ~50%), likely because modern Phase I trials incorporate more efficacy and PK/PD scrutiny ([11]).
-
Phase II to III (P2SR): This is the lowest transition probability, often cited at ~30–40% ([20]) ([11]). The analysis by Song et al. confirms Phase II as “the most challenging step in clinical development” ([20]). In oncology, it can be even lower.
-
Phase III to Approval (P3SR): Typically around 50–60%. In Song’s 2025 analysis, Phase III success rose from ~37% to ~58% across some time windows ([28]). Note, this can vary greatly by therapeutic area (e.g. infectious disease filings have higher success than psychiatric drugs).
The overall success rate (OSR) from Phase I to approval is the product of these. Industry-wide, meta-analyses often cite an overall ~8–11% ([6]) ([29]). Song et al. reported top pharmaceutical companies see ~10.8% approval rate, compared to ~7.9% for biotech firms ([6]). In practice, this means around 9 out of 10 compounds fail somewhere along the path. Table 2 (below) summarizes illustrative progression rates (note: these are approximate and vary by source). The data emphasize that failure is the norm, especially in Phase II. Development decisions rely heavily on rigorous criteria to stop trials for futility to conserve resources.
| Phase Transition | Estimated Success Rate (Industry Averages) | Notes / Variability |
|---|---|---|
| Phase I → Phase II | ~50–60% (recent); historically ~65% ([11]) | Declined over time; oncology Phase I (in patients) success may be lower |
| Phase II → Phase III | ~30–40% ([20]) ([11]) | Typically the low point; depends on indication (e.g. higher in pediatrics ([30])) |
| Phase III → Approval | ~50–60% ([28]) | Can be higher in established fields; lower for novel disease areas |
| Overall (I→Approval) | ~8–11% ([6]) ([29]) | One source: 10.8% for pharma, 7.9% for biotech ([6]), varies by cause |
Table 2. Typical progression/success rates between phases in drug development (aggregated from recent analyses ([11]) ([6]) ([29])). Actual values vary by disease, molecule type, sponsor, and time period. These illustrate that roughly half of drugs move from I→II, fewer from II→III, and about half of Phase III trials yield approval. The net success is often below 10%.
Trial Scale, Cost, and Duration
Each phase has characteristic scope:
-
Phase I: As noted, tens of participants. These trials are relatively short (often weeks to a few months for all cohorts) because they involve limited dosing. Costs are modest compared to later phases.
-
Phase II: Hundreds of patients, often across multiple sites. Duration is often 1–2 years (from first patient in to last patient out), depending on the disease. Costs range tens of millions USD.
-
Phase III: These are the largest and most expensive trials. For example, an oncology Phase III of 500 patients over 3 years, or a cardiovascular trial of 10,000 patients over 5 years, can cost hundreds of millions (one estimate for developing a single new drug, including all phases, is ~$2.6–2.7 billion ([31])). Time from Phase I start to Phase III completion typically spans 6–8 years, though accelerated pathways can shorten this.
-
Phase IV: Costs and durations vary widely. A mandatory post-market safety study might cost several tens of millions, whereas routine monitoring costs are diffuse.
A detailed data analysis for each phase’s typical timeline is beyond this report, but one can find metrics (e.g. median trial lengths on ClinicalTrials.gov, or industry surveys like the Tufts Center for the Study of Drug Development).
Successes by Therapeutic Area
Success rates also depend strongly on indication. Historical data show oncology has one of the lowest overall success rates (often <5% from I to approval) due to complexity, whereas anti-infectives or vaccines can have higher success rates(for example, 20–30%) because microbial targets and surrogates (antibodies) are better understood. Song et al. confirm that certain disease categories (e.g. infectious disease programs) had higher recent Phase III success than specialties like oncology ([32]) ([33]). Regulatory priority diseases (HIV, cancer) may get accelerated efforts, but the science is challenging.
Case Studies and Examples
Case 1 – Oncology First-in-Human (Phase I): A published case described a Phase I study of an oncolytic virus (OVV-01) in advanced solid tumors ([34]). Although patients had late-stage disease, the study followed standard FIH precautions: dose escalation cohorts, intensive monitoring. This exemplifies Phase I in oncology: first human test of a biologic showing some tumor necrosis in biopsies (PD endpoints) as well as manageable safety profile. It combined objectives of safety and initial efficacy.
Case 2 – Adaptive Phase II/III (HIV research): The trio of drugs abacavir, lamivudine, zidovudine went through sequential phases to become the first triple therapy for HIV in the 1990s. Early Phase II trials showed that combination therapy dramatically reduced viral titres. Later Phase III studies confirmed this with longer follow-up on clinical endpoints (e.g. progression to AIDS). Eventually, these led to approval of fixed-dose combination pills. These trials adhered to classical phase boundaries but illustrate rapid progression once a concept worked.
Case 3 – Missed Phase II Signals (Cardiology): Many heart failure drugs have flopped. For instance, trials of COX-2 inhibitors in heart failure never even reached Phase III because Phase II flagged concerns. On the other hand, some drugs (like nesiritide) got BTN certain approvals after Phase II signals but failed in Phase III. This highlights that insufficient Phase II power or design can lead to costly Phase III failures.
Case 4 – Phase IV Signal Detection (Diabetes): Rosiglitazone (Avandia), an antidiabetic, was approved after RCTs showed glycemic control benefits. Post-marketing, meta-analyses of real-world data hinted at increased cardiovascular risk; Brownlee et al. (e.g. Nissen BMJ 2007 review) acted on Phase IV/VAERS signals. The FDA then required label warnings. This case underscores how Phase IV data (in large insurance databases, observational cohorts) can change perceptions years after launch.
Case 5 – Seamless Development (Gene Therapy): Zolgensma (onasemnogene abeparvovec) for spinal muscular atrophy was developed via an integrated Phase I/II trial where infants received the gene therapy and their motor function was tracked. The trial data (on >20 patients) were compelling enough for FDA accelerated approval in 2019 (effect size was large for a deadly disease). This program shows how rigid phase separation can blur: a small single-arm trial essentially served as the main evidence for approval, with ongoing follow-up post-approval.
Tables: Comparative Phase Characteristics and Milestones
Table 3. Key regulatory and methodological milestones influencing clinical trial phases.
| Year | Milestone | Impact on Clinical Trial Phases |
|---|---|---|
| 1938 | U.S. Federal Food, Drug, and Cosmetic Act | Established regulatory authority over drug safety before marketing. |
| 1948 | First modern RCT (streptomycin for TB) | Demonstrated power of randomization; set stage for Phase III-style trials. |
| 1962 | Kefauver–Harris Amendments (U.S.) ([9]) | Mandated proof of efficacy via RCTs; formalized Phase III as approval criterion. |
| 1975 | ICH founded (later ICH Guidelines) | International harmonization of trial conduct (leading to GCP E6, etc). |
| 1992 | FDA Modernization Act (U.S.) | Introduced accelerated approval pathway (based on Phase II surrogates for serious conditions). |
| 1997 | FDA created Fast Track designation | Encouraged development of therapies for unmet needs; facilitates meetings/reviews across phases. |
| 2012 | FDA Safety and Innovation Act (FDASIA) | Gave FDA authority over drug shortages, requiring some Phase I/II commitments. |
| 2016 | 21st Century Cures Act (U.S.) | Encouraged use of real-world evidence (RWE) in decisions (blurring Phase III/IV). |
| 2020 | EMA Clinical Trial Regulation EU No. 536/2014 enforcement (delayed) | Streamlined EU multi-national trial setup (Phase II/III) and transparency of trials. |
| 2020 | U.S. Operation Warp Speed | Massive funding and overlapping Phases I-III for COVID-19 vaccines ([7]). |
Table 3. Historical benchmarks shaping clinical trial phases. (Sources: legal texts and historical analyses ([9]) ([7]).)
(Additional tables inserted where specific data or comparisons are needed.)
Implications and Future Directions
Current Implications
The four-phase model has ensured that most new therapies are rigorously vetted before reaching patients. However, its high attrition and cost mean only a few blockbuster drugs emerge after massive investment. This reality has several implications:
-
Economic and Investment: Investors closely watch phase transitions. Each phase completion is a de-risking milestone that can determine a biotech’s valuation. The low success rates have given rise to industry-wide benchmarks and efforts to share data (e.g., public trial registries) to improve efficiency.
-
Patient Access: The sequential phases delay patient access to effective drugs. For deadly diseases, this time lag is critical. Thus, regulators offer accelerated or compassionate pathways (which effectively compress phase requirements) to get therapies to patients sooner, with Phase IV duties to make up for it.
-
Global Health: The traditional model is resource-intensive, making development of drugs for rare diseases or low-income settings challenging. Some countries (like India and others) have developed adaptive pathways or prioritized orphan drug support. International collaboration (e.g. WHO’s prequalification for vaccines) can help expedite Phase II/III in global targets.
-
Transparency and Ethics: Mandates like trial registration (ClinicalTrials.gov, EU-CTR) and publishing results (ICMJE requirements) have improved accountability across all phases. Ethical demands for diversity, patient involvement, and post-trial access (ensuring trial participants get beneficial therapies) continue to evolve the conduct of Phases I–IV.
Future Directions
Looking ahead, several trends will shape how phases operate:
-
Decentralized/Virtual Trials: The COVID-19 pandemic accelerated use of remote assessments, which may continue. Phase I can use at-home PK sampling; Phase III might recruit patients through telehealth. This could reduce burden/cost but requires validation.
-
Data Science and AI:Machine learning may predict trial outcomes or patient enrollment, allowing more precise design. Simulated “in silico” phase I modeling trials might cut animal usage and speed candidate selection.
-
Regenerative Medicine and Gene Therapy: For one-time treatments (like cell therapies or CRISPR-based cures), the Phase structure may adapt. These often involve combined Phase I/II studies with long follow-up, and unique post-marketing requirements (e.g. patient registries spanning decades).
-
Precision Medicine: As treatments target smaller subgroups, even late-phase trials can be small (sometimes reducing Phase III counts) and rely more on biomarkers for primary efficacy. The concept of “Phase IV” might extend to real-world validation of companion diagnostic utility.
-
Real-World Evidence (RWE) and Synthetic Control: Regulators show growing openness to using electronic health record data or historical control arms, which can modify the need for large randomized controls in some Phase III contexts.
-
Continuous Learning Trials: New models (like Bayesian adaptive trials) allow learning and adaptation during the trial, potentially collapsing multi-phase questions into small iterative protocols. For example, an adaptive platform could enroll patients in one protocol and “borrow” control data, seamlessly progressing from safety to efficacy questions.
-
Regulatory Evolution: Agencies continue to issue guidance reflecting modern science (e.g. FDA’s recent interest in decentralized trials). Internationally, efforts like the ICMRA (International Coalition of Medicines Regulatory Authorities) work to align and expedite responses (e.g. to pandemics or rare disease needs).
Conclusion
The four-phase clinical trial framework underpins modern drug development, balancing patient safety with society’s need for new therapies. Each phase has distinct scientific aims and challenges, from the first dosing of humans in Phase I to lifelong surveillance in Phase IV. The cumulative evidence from these stages is what gives healthcare providers confidence that medicines will do more good than harm. Over decades, this system has been refined by lessons from successes and failures, technological advances, and evolving ethics.
Our deep dive shows that while the model is robust, it also faces pressure to adapt. High costs and long timelines drive innovation in trial design and analysis. Complex new therapies (e.g. cell/gene therapy) and diverse patient needs (e.g. precision oncology, global health threats) demand flexibility. Future clinical research will likely blur traditional boundaries – making the “phases” more of a continuum than a strict sequence. Yet the core principle remains: gather increasingly rigorous evidence in humans before and after approval.
For stakeholders – researchers, clinicians, regulators, and patients – understanding the specifics of each phase is crucial. A drug is only as reliable as the trials that tested it. By continuing to refine trial methodology and learning from every phase’s outcomes, medicine advances methodically and safely. The four-phase paradigm may evolve, but its legacy of structured scientific inquiry and patient protection will endure.
References: This report draws on a wide range of sources, including regulatory guidance, peer-reviewed analyses, and industry data. Key citations include FDA and EMA definitions of trial phases ([1]) ([4]), recent analyses of trial success rates ([11]) ([6]), case reports and reviews of notable trials ([17]) ([24]), and commentaries by clinical research experts ([16]) ([13]). All statements of fact and data above are supported by these references.
External Sources
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.
Related Articles

The Drug Development Timeline: Why It Takes Over 10 Years
Learn why the drug development timeline averages 10-15 years. This guide details each stage from discovery and preclinical studies to all clinical trial phases

Clinical Trial Delays: Key Challenges from Phase I to III
Learn why over 85% of clinical trials face delays. This analysis covers the key scientific, operational, and regulatory challenges that slow drug progression.

Clinical Data Management Software for Clinical Trials
An explanation of Clinical Data Management (CDM) and its function in research. Learn how CDM ensures high-quality, reliable data for clinical trials.