Fact-checked by Grok 2 weeks ago

Clinical trial

A clinical trial is a prospective study involving participants prospectively assigned to one or more interventions, such as drugs, devices, or behavioral modifications, to evaluate their effects on biomedical or behavioral outcomes. These studies are structured in phases—typically Phase I for safety and dosing in small groups, Phase II for preliminary efficacy, Phase III for confirmatory effectiveness in larger populations, and Phase IV for post-approval monitoring—to generate empirical evidence of safety and efficacy required for regulatory approval of medical interventions. Clinical trials represent the gold standard for establishing causality in therapeutic effects through and controls, mitigating biases inherent in observational and enabling first-principles assessment of intervention impacts. Historically, they trace back to empirical tests like James Lind's 1747 controlled experiment demonstrating citrus fruits' efficacy against , evolving into randomized controlled trials pioneered by in 1948 for evaluating in , which formalized statistical rigor to distinguish true effects from confounders. Ethical frameworks, necessitated by past abuses such as non-consensual experiments during , were enshrined in the of 1947 and subsequent Declaration of Helsinki, mandating voluntary , risk minimization, and scientific validity, though controversies persist over issues like use, in trial design, and equitable participant access amid incentives for pharmaceutical advancement. Despite high attrition rates—over 90% of candidates failing to reach market—clinical trials underpin virtually all approved therapies, driving medical progress while demanding vigilant oversight to balance innovation with participant welfare.

Definition and Purpose

Core Objectives and Scientific Rationale

The core objectives of clinical trials are to generate reliable evidence on the , , and practical utility of s such as drugs, biologics, devices, or behavioral modifications in human populations. These trials systematically test whether an intervention produces measurable health improvements—such as reduced mortality, symptom alleviation, or prevention—compared to no , , or established alternatives, while identifying risks like adverse events or . Primary endpoints focus on predefined outcomes, such as clinical response rates or changes, to isolate the intervention's causal impact from extraneous factors. Secondary objectives often explore dosing regimens, effects, or long-term durability to inform regulatory approval and clinical guidelines. The scientific rationale for clinical trials derives from the limitations of preclinical data and uncontrolled human observations, which cannot reliably establish due to variables, selection biases, and responses. Interventions must demonstrate reproducible effects in diverse cohorts to justify widespread adoption, as historical reliance on has led to ineffective or harmful practices, such as thalidomide's teratogenic risks before rigorous trialing. Randomized controlled designs, the cornerstone of modern trials, allocate participants to or arms via chance to ensure baseline comparability, thereby minimizing systematic differences that could distort results. This approach, complemented by blinding (where participants, providers, or assessors are unaware of assignments) and prospective protocols, reduces performance, detection, and reporting biases, enabling grounded in empirical probability rather than . Trials prioritize interventions with plausible mechanisms from preclinical models but demand human validation, as animal data often overpredicts or underpredicts human responses—evident in cases where up to 90% of promising compounds fail in Phase I due to unforeseen or toxicity. Ethical imperatives, codified in frameworks like the Declaration of Helsinki, further necessitate trials to balance potential benefits against risks, ensuring (genuine uncertainty about superiority) before exposing participants. By quantifying effect sizes with statistical power—typically powered to detect differences of , such as hazard ratios below 0.8 for superiority—trials provide the probabilistic certainty required for , surpassing lower-tier evidence like studies prone to immortal time or channeling effects. This rigor underpins regulatory standards, with bodies like the FDA mandating pivotal trials for marketing authorization to protect from unsubstantiated claims.

Applications Across Interventions

Clinical trials evaluate interventions spanning pharmacological, biological, surgical, device-based, and behavioral domains, adapting (RCT) methodologies to assess causality between the intervention and health outcomes such as symptom reduction or disease prevention. These applications prioritize empirical endpoints like mortality rates, changes, or functional improvements, while addressing intervention-specific challenges like blinding feasibility or ethical constraints on use. For instance, over 400,000 interventional trials registered on as of 2024 encompass these categories, with pharmaceutical trials comprising the majority due to regulatory mandates for market approval. In pharmaceutical development, trials test small-molecule drugs and biologics through sequential phases, starting with dose-escalation studies in healthy volunteers to establish and maximum tolerated doses, progressing to trials against standard care. The U.S. (FDA) requires (IND) applications prior to human testing, with Phase III trials often involving thousands of participants to detect modest effect sizes, as seen in cardiovascular drug evaluations where hazard ratios below 0.9 necessitate sample sizes exceeding 10,000 for statistical power. Biologics, including monoclonal antibodies, follow similar paths but under the , emphasizing manufacturing consistency due to their complexity from cellular sources. Vaccine trials apply RCT designs to measure and protective efficacy, often using surrogate endpoints like titers alongside clinical disease incidence in Phase III, as in the 1990s polio vaccine eradication efforts that informed modern trials requiring over 30,000 participants per arm for rare event detection. These differ from drug trials by incorporating challenge models or cluster randomization in outbreak settings to simulate real-world transmission dynamics. Medical device trials, regulated via the FDA's 510(k) clearance for Class I/II devices or premarket approval () for Class III, emphasize performance metrics like durability or procedural success rates over long-term , with pivotal studies often smaller (hundreds of patients) and relying on bench testing or animal data for initial safety. Unlike drugs, devices permit iterative modifications during trials under investigational device exemptions (), as in evaluations where failure rates below 1% drive approval thresholds. Surgical interventions pose unique RCT challenges, including surgeon expertise variability and blinding difficulties, often addressed via procedures or objective outcome measures like operative time or complication rates; a review of 388 surgical RCTs found median sample sizes of 80 participants, focusing on minor endpoints amid recruitment barriers from patient preferences for active treatment. —the genuine uncertainty of superiority—is harder to establish when comparing surgery to , as in trials for arthroscopic knee procedures showing no benefit over physiotherapy in . Behavioral interventions, such as (CBT) for anxiety or lifestyle modifications for , utilize RCTs to isolate therapeutic effects from nonspecific factors like expectation, with meta-analyses confirming CBT's superiority over waitlist controls ( d=0.8) in trials involving blinded assessors. These trials often employ manualized protocols for and longer follow-ups to capture , though challenges include therapist allegiance , prompting independent replication requirements.

Historical Development

Pre-20th Century Foundations

The foundations of clinical trials trace back to ancient practices emphasizing empirical observation and comparison of interventions, though systematic experimentation was rare. In the Hebrew Bible's (circa 600–500 BCE), a controlled dietary comparison involved ten young consuming vegetables and water for ten days, while a control group ate the king's rich provisions; the vegetable group exhibited healthier appearances, marking an early instance of prospective outcome assessment. Ancient Greek physician (c. 460–370 BCE) advanced clinical reasoning through detailed case observations and the doctrine of balancing humors via diet and drugs, prioritizing evidence from patient outcomes over speculation. Similarly, Roman physician (c. 129–216 CE) conducted comparative studies on animal models and human subjects to evaluate therapies, such as , influencing medical methodology for centuries. Medieval Islamic scholars refined these approaches with greater methodological rigor. Rhazes (865–925 CE) compared chicken and lamb broths for treating fevers, noting differential recovery rates based on observed symptoms. (980–1037 CE), in his , advocated testing remedies on healthy individuals first, using placebos to distinguish true effects from psychological influences, and emphasized replication and ethical consent precursors. During the , (1493–1541) rejected Galenic authority in favor of direct experimentation with chemicals like mercury for , insisting on dosage precision derived from animal and human trials to avoid toxicity. The 18th century saw pivotal advances toward modern designs. In 1747, Scottish naval surgeon divided twelve scurvy-afflicted sailors on HMS Salisbury into six pairs, administering varied treatments (e.g., vinegar, seawater, citrus); the citrus group recovered swiftly, demonstrating comparative efficacy despite lacking randomization or blinding. This trial, published in Lind's 1753 Treatise on Scurvy, highlighted dietary causation over miasma theories. In 1796, English physician inoculated eight-year-old with vesicle fluid from dairymaid Sarah Nelmes, then exposed him to smallpox six weeks later; Phipps remained immune, establishing vaccination's protective mechanism against a deadlier . Jenner's 1798 publication of twenty-three cases solidified this as a foundational , prioritizing through observational follow-up. These pre-20th century efforts, though ethically rudimentary and often opportunistic, introduced core principles like intervention comparison, outcome measurement, and from controlled groups, paving the way for formalized trial structures amid prevailing reliance on and tradition.

20th Century Reforms and Milestones

The disaster of 1937, in which over 100 individuals died from a toxic solvent used in a formulation of the , exposed critical gaps in pre-market drug safety testing and prompted the passage of the Federal Food, Drug, and Cosmetic Act in 1938. This legislation mandated manufacturers to submit evidence of safety from adequate animal and human studies before marketing new drugs, marking the first federal requirement for preclinical and clinical safety data in the United States. Following revelations of unethical medical experiments conducted by Nazi physicians during , the Nuremberg Military Tribunal issued the in 1947, establishing foundational ethical principles for human experimentation. The Code emphasized voluntary as absolutely essential, the necessity of yielding fruitful results unprocurable by other means, and the avoidance of unnecessary physical or mental suffering, influencing subsequent international standards for ethics. In 1948, the British Medical Research Council's trial of for pulmonary , designed by statistician , became the first published (RCT), allocating 107 participants via random numbers to treatment or control groups to minimize bias and establish causality. This methodological innovation demonstrated streptomycin's efficacy, with treated patients showing a 7% compared to 27% in controls after six months, solidifying RCTs as the gold standard for evaluating therapeutic interventions. The 1954 field trial of Jonas Salk's inactivated involved approximately 1.8 million children across the , employing randomized, placebo-controlled designs in some areas to assess efficacy, which was later confirmed at 80-90% effective against paralytic . This massive undertaking highlighted the logistical feasibility of large-scale RCTs and accelerated adoption, reducing incidence dramatically. The tragedy, where the sedative caused severe birth defects in thousands of European infants between 1957 and 1961, galvanized U.S. regulatory reform, leading to the Kefauver-Harris Amendments of 1962. These amendments required proof of drug efficacy through "adequate and well-controlled investigations," explicit from trial participants, and FDA approval of applications, shifting oversight from mere safety to comprehensive evidence of benefit-risk balance. In 1964, the adopted the Declaration of Helsinki, expanding on the by providing ethical guidelines for physicians conducting clinical research, stressing the primacy of participant welfare over scientific interests and the need for independent ethical review committees. Exposures of the , where treatment was withheld from African American men from 1932 to 1972, underscored ongoing ethical lapses, culminating in the of 1974. This act established Institutional Review Boards (IRBs) at research institutions to oversee human subjects protection, mandated , and created the National Commission for the Protection of Human Subjects to formulate ethical guidelines, institutionalizing local ethical scrutiny in clinical trials.

Post-2000 Evolutions and Global Standardization

The International Council for Harmonisation (ICH) advanced global standardization of clinical trials through updates to its (GCP) guideline, with the E6(R2) addendum adopted in 2016 emphasizing risk-based quality management, enhanced sponsor responsibilities for oversight, and integration of technology to improve and participant safety. This revision addressed limitations in the 1996 original by promoting proportionate based on identified risks rather than uniform 100% source data verification, while maintaining core ethical principles for trial conduct across jurisdictions including the , , and . In the US, the (FDA) launched the Critical Path Initiative in 2004 to tackle bottlenecks in , such as stagnant approval rates, by fostering predictive biomarkers, modeling tools, and collaborative platforms to streamline clinical evaluation without compromising rigor. Complementing this, the 2007 Amendments Act (FDAAA) mandated registration of applicable clinical trials on within 21 days of first patient enrollment and submission of summary results, including adverse events, within specified timelines post-completion, thereby enforcing greater and reducing selective reporting biases observed in earlier eras. The aligned with global standards via Regulation (EU) No 536/2014, which entered full application on January 31, 2022, after validation of the Clinical Trials Information System (CTIS); this replaced the Clinical Trials Directive by establishing a centralized EU portal for trial submissions, coordinated ethical and scientific assessments across member states, and requirements for proactive safety reporting to expedite approvals while harmonizing data standards. These frameworks facilitated the expansion of multinational trials, with registered studies starting annually rising from 1,873 in 2000 to 22,131 in 2020, increasingly spanning multiple countries to access diverse populations and reduce costs, though variations in local enforcement have prompted ongoing ICH efforts to refine implementation guidelines.

Types and Phases

Preclinical Preparation

Preclinical preparation involves and to assess a candidate intervention's , pharmacological profile, and potential before human testing. These studies identify risks of and establish preliminary dosing parameters essential for regulatory approval to proceed to clinical phases. Initial screening occurs through in vitro methods, including cell cultures, biochemical assays, and computational models, to evaluate , , and early indicators without involving whole organisms. These techniques allow high-throughput testing of compound libraries to select lead candidates based on target engagement and selectivity. Subsequent studies in animal models, typically and non-rodents like dogs or , examine (absorption, distribution, metabolism, excretion) and to predict human responses. Toxicology assessments determine no-observed-adverse-effect levels (NOAEL) through acute, subchronic, and chronic dosing, revealing dose-dependent toxicities such as organ damage or carcinogenicity. All studies must comply with (GLP) regulations to ensure data reliability for regulatory review. Preclinical data, including manufacturing details and , support the () application to agencies like the U.S. (FDA), which reviews submissions within 30 days to authorize Phase I trials if no unreasonable risks are evident. This phase often spans 1 to 6 years, with iterative lead optimization to refine compounds before IND filing. Interspecies physiological differences limit predictive accuracy, necessitating cautious extrapolation to humans.

Phase I: Safety and Dosage

Phase I trials represent the inaugural human testing phase for investigational drugs or biologics, succeeding preclinical animal and laboratory evaluations to bridge toward therapeutic application. These studies prioritize establishing the safety profile, delineating tolerable dosage ranges, and characterizing pharmacokinetics, including absorption, distribution, metabolism, and excretion (ADME). Efficacy is not the principal endpoint, though incidental observations may inform later phases; instead, the focus remains on averting harm while probing physiological interactions. Regulatory bodies like the U.S. Food and Drug Administration (FDA) mandate estimation of a maximum safe starting dose from preclinical no-observed-adverse-effect levels (NOAEL), often scaled by safety factors to mitigate risks in humans. Participant cohorts are small, typically numbering 20 to 100 individuals, to minimize exposure while generating foundational data. Healthy volunteers predominate for non- interventions to isolate effects absent disease states, though Phase I trials recruit patients due to ethical constraints against dosing healthy individuals with cytotoxics. emphasizes risks, including potential severe adverse events, with protocols incorporating dose-limiting (DLT) criteria—such as grade 3 or higher non-hematologic toxicities—to trigger or cessation. Monitoring entails frequent clinical assessments, electrocardiograms, and laboratory analyses to detect toxicities promptly. Dose-escalation designs drive progression, commencing at subtherapeutic levels and advancing incrementally to identify the maximum tolerated dose (MTD), defined as the highest dose yielding acceptable in approximately one-third of subjects. The traditional rule exemplifies this: cohorts of three participants receive a dose; if no DLT occurs, escalation proceeds, but two or more DLTs prompt reversion to the prior level, expanding cohorts for confirmation. Model-based alternatives, like the continual reassessment method (), leverage for more precise MTD estimation, reducing patient exposure to sub- or supra-optimal doses. Pharmacodynamic markers, where feasible, correlate exposure with biological effects to refine dosing paradigms. These trials, often lasting months, yield data pivotal for Phase II advancement, with approximately 70% of investigational agents progressing despite occasional underestimation of cumulative toxicities. Ethical oversight, via institutional review boards, ensures risk-benefit proportionality, particularly given historical precedents like the 1937 Elixir Sulfanilamide disaster underscoring human testing imperatives. Variations, such as adaptive designs, permit mid-trial modifications based on interim safety signals, enhancing efficiency without compromising rigor.

Phase II: Efficacy and Side Effects

Phase II clinical trials primarily assess the preliminary efficacy of an investigational drug, device, or intervention in treating the targeted disease or condition, while continuing to monitor adverse effects and refine dosing regimens. These trials follow Phase I safety testing and involve participants who have the specific condition under study, typically numbering 100 to 300 individuals, allowing for detection of therapeutic signals beyond initial tolerability. Efficacy is evaluated through defined endpoints such as tumor response rates in oncology or symptom improvement in other indications, with the goal of determining whether the intervention shows sufficient promise to justify larger confirmatory studies. Trial designs in Phase II often incorporate randomization to minimize selection bias and may include control arms, either active comparators or placebos, though single-arm studies predominate in early efficacy screening, particularly in where historical response rates serve as benchmarks. Blinding—single, double, or triple—is employed where feasible to reduce observer and participant , especially in randomized formats, but open-label designs are common due to practical constraints like invasive endpoints or ethical considerations. Statistical power is calibrated for interim analyses, often accepting higher type I error rates (10-20%) compared to Phase III to efficiently screen candidates, with primary endpoints focused on short-term outcomes like objective response or rather than long-term survival. Side effects are scrutinized for frequency, severity, and dose-dependency, building on Phase I data to identify manageable risks in a population; common adverse events may lead to dose adjustments or trial discontinuation if they outweigh preliminary benefits. For instance, in cardiovascular , Phase II trials have historically revealed efficacy gaps, with only about 6.6% of candidates advancing successfully from this stage due to insufficient therapeutic indices or unexpected toxicities. Outcomes inform decisions, where promising results—such as those from checkpoint inhibitors like in early non-small cell trials—prompt Phase III progression, while discrepancies between Phase II optimism and later failures underscore the phase's role as a high-attrition filter, with divergent results observed in at least 22 FDA-reviewed cases since 2010.

Phase III: Confirmation and Comparison

Phase III clinical trials represent the pivotal confirmatory stage in , involving large-scale, randomized, controlled studies to verify the and of an investigational in a diverse representative of intended clinical use. These trials typically enroll several hundred to several thousand participants, often ranging from 300 to 3,000 or more depending on the condition's and requirements, to achieve sufficient statistical power for detecting differences. Unlike Phase II trials, which focus on preliminary in smaller groups of 100-300 patients, Phase III emphasizes broad confirmation, including comparisons to existing standards of care, and generates the primary dataset for regulatory submissions such as New Drug Applications (NDAs) to agencies like the FDA. Trials in this phase are usually multicenter and multinational to ensure generalizability, employing and blinding—often double-blind—to minimize while comparing the experimental against an active (the current standard treatment) or, where ethically feasible, a . The core objectives include assessing clinical benefits such as improved survival, symptom relief, or disease progression delay, alongside monitoring for adverse events that may occur infrequently and thus require large sample sizes for detection. For instance, these studies evaluate dose-response relationships in real-world settings, identify subgroups benefiting most (e.g., by age, , or comorbidities), and collect data on long-term tolerability, which informs product labeling and risk-benefit assessments. Successful III outcomes provide robust evidence for regulatory approval, with positive results supporting marketing authorization by demonstrating superiority, non-inferiority, or equivalence to established therapies under predefined s like overall response rate or . However, discrepancies between Phase II promise and Phase III results occur, as documented in FDA analyses of 22 cases where initial signals failed to replicate at scale due to factors like population heterogeneity or endpoint variability, underscoring the necessity of these expansive trials to avoid overestimating benefits. Post-approval, Phase III data also facilitate by establishing baselines for rarer side effects, though limitations such as trial exclusion criteria can lead to underrepresentation of certain demographics, prompting calls for more inclusive designs.

Phase IV: Post-Market Surveillance

Phase IV clinical trials, also termed post-marketing surveillance or pharmacovigilance studies, commence after regulatory approval by agencies such as the U.S. (FDA) or the (EMA), when the drug or device enters widespread clinical use. These studies monitor long-term safety, efficacy in diverse real-world populations, and potential rare adverse events that may not emerge in earlier phases due to smaller sample sizes or controlled conditions. Unlike pre-approval phases, Phase IV relies heavily on observational data rather than randomized controlled trials, enabling detection of issues like drug interactions, off-label misuse, or effects in subpopulations excluded from prior studies. The primary objectives include identifying adverse events with incidence rates below 1 in 1,000—events infeasible to detect in Phase III trials involving thousands of participants—and assessing sustained therapeutic benefits over years of use. Methods encompass passive reporting systems (e.g., FDA's MedWatch for voluntary submissions), active surveillance via registries, studies, and mandated post-approval commitments. For high-risk approvals, regulators impose specific Phase IV requirements, such as the FDA's 1,066 post-marketing studies mandated in 2023 for newly approved drugs, focusing on unresolved safety signals. Observational designs introduce challenges like variables, necessitating advanced statistical adjustments to infer , though they provide essential unattainable in experimental settings. Regulatory frameworks enforce ongoing obligations: the FDA requires manufacturers to report serious adverse events within 15 days and conduct surveillance under the Federal Food, Drug, and Cosmetic Act, with authority to withdraw approvals if risks outweigh benefits. Similarly, EMA's system mandates plans and periodic benefit-risk evaluations, as outlined in Regulation (EU) No 726/2004. Failure to detect issues promptly can lead to market withdrawals; for instance, (Vioxx), approved by the FDA in 1999, revealed doubled cardiovascular event risks in the post-marketing Adenomatous Polyp Prevention on Vioxx (APPROVe) , prompting its voluntary withdrawal in September 2004 after cumulative data implicated it in approximately 27,000 heart attacks or strokes. Another case, (Rezulin), approved in 1997, was pulled in 2000 following post-marketing reports of severe not fully anticipated in trials. These studies underscore causal realism in drug safety: while Phase III confirms efficacy under ideal conditions, Phase IV exposes discrepancies from real-world variability, such as or demographic diversity, justifying their role in refining labeling, restricting indications, or mandating black-box warnings. Empirical data from Phase IV has driven over 100 U.S. drug withdrawals since , often tied to rare toxicities emerging post-approval, emphasizing the need for robust, unbiased reporting to counter under-detection biases in voluntary systems.

Specialized Trial Designs

Specialized clinical trial designs incorporate modifications to traditional parallel-group randomized controlled trials to address limitations in efficiency, feasibility, or applicability, particularly for rare diseases, , or multi-arm evaluations. These approaches, often termed master protocols or innovative designs, enable simultaneous testing of multiple interventions or adaptations based on accruing data, reducing redundancy and accelerating evidence generation. The U.S. has endorsed their use through guidances emphasizing prospective planning to preserve validity and minimize bias. Adaptive designs allow pre-planned alterations to key elements, such as sample size, ratios, or treatment arms, informed by interim analyses of accumulating data. This flexibility can enhance efficiency by discontinuing futile arms or enriching for responsive subgroups, though it requires careful statistical modeling to control type I error rates. The FDA's 2019 guidance specifies that adaptations must be outlined in the to ensure interpretability, with simulations recommended to validate operating characteristics. Applications include trials where early efficacy signals prompt escalation. Basket trials evaluate a single therapeutic agent or combination across heterogeneous patient populations defined by a shared molecular target, irrespective of tumor , to identify responsive subsets efficiently. Originating in precision medicine, they bypass separate trials for rare mutations by pooling data while permitting histology-specific analyses. For example, they have been applied to test targeted therapies in cancers harboring specific genetic alterations, with response rates analyzed per basket. Umbrella trials assess multiple targeted therapies simultaneously within one disease context, stratifying participants by biomarker-defined subtypes and comparing each arm to a common control. This structure facilitates direct subgroup comparisons and resource sharing, as demonstrated in studies evaluating genotype-specific agents. Platform trials establish a continuous, multi-arm framework that can dynamically add, drop, or modify interventions based on ongoing results, often using shared controls and Bayesian methods for inference. They offer perpetual operation, as in the trial for treatments initiated in 2020, which efficiently identified effective drugs like dexamethasone while discarding ineffective ones. Crossover designs expose participants to multiple treatments sequentially, with washout periods to mitigate carryover effects, leveraging within-subject comparisons to reduce inter-individual variability and sample size needs. Suited for , reversible conditions like migraines, they assume no period or sequence effects, confirmed via statistical tests. Factorial designs test two or more interventions concurrently by randomizing to all combinations, enabling assessment of main effects and interactions in a single trial. A 2x2 , for instance, requires one-quarter more participants than separate trials for main effects alone, but assumes no interactions unless tested. They are efficient for evaluating additive therapies, such as in cardiovascular prevention studies. Non-inferiority designs seek to establish that a new performs no worse than an active within a pre-specified margin, justified by historical superiority to preserve a of the comparator's benefit. The margin, often set via , requires larger sample sizes than superiority trials due to one-sided testing, and per-protocol analyses to avoid dilution . These are essential when placebos are unethical, as in antibiotic trials.
Design TypeKey FeaturePrimary AdvantageLimitation
AdaptiveInterim data-driven modificationsEfficiency in arm selectionComplexity in error control
BasketSingle drug, multiple diseasesBiomarker focus across raritiesHeterogeneity in responses
UmbrellaMultiple drugs, one diseaseSubtype comparisonsLogistical coordination
CrossoverSequential treatments per subjectReduced variabilityCarryover risk in acute conditions
Multi-intervention combinationsInteraction detectionAssumes no strong interactions
These designs demand rigorous pre-specification and regulatory dialogue to ensure credibility, with ongoing FDA pilots promoting their integration into drug development.

Design Principles

Randomization, Blinding, and Controls

Randomization assigns study participants to intervention or control groups using chance-based methods, aiming to balance known and unknown prognostic factors across groups and thereby minimize selection bias. Common techniques include simple randomization (e.g., coin flips or random number tables), block randomization (to ensure equal group sizes over time), and stratified randomization (to balance subgroups by key covariates like age or disease severity). This process enhances the internal validity of trials by promoting baseline comparability, allowing causal inferences about treatment effects rather than confounding variables. Evidence from meta-analyses shows randomized trials yield more reliable estimates of treatment efficacy compared to non-randomized designs, as randomization mitigates systematic errors that observational studies often amplify. Blinding, or masking, prevents knowledge of group assignments from influencing trial conduct or outcomes, reducing performance bias (altered participant or investigator behavior) and detection bias (subjective outcome assessment). In single-blind designs, only participants are unaware of assignments; double-blind extends this to investigators; triple-blind includes data analysts to avoid interpretation bias. Regulatory guidelines emphasize double-blinding where feasible, as it strengthens credibility by limiting conscious and unconscious biases, with studies demonstrating unblinded trials overestimate treatment benefits by up to 30% in subjective endpoints. Challenges arise in surgical or behavioral interventions where full blinding is impractical, necessitating alternative bias controls like objective endpoints. Control groups provide a baseline for comparison, isolating the investigational intervention's effects from natural disease progression, responses, or external factors. controls, inert substances mimicking active treatment, are preferred when ethical to demonstrate superiority over no treatment, enabling quantification of specific therapeutic effects amid nonspecific influences. Active controls use established therapies to assess noninferiority or superiority, essential in conditions with available standards of care to avoid withholding effective treatment. FDA guidance specifies that -controlled trials with and blinding minimize bias most effectively, though active controls suffice for equivalence testing when placebos risk harm. Integrated with and blinding, these elements form the framework, upheld by ICH standards as optimal for generating unbiased evidence of safety and efficacy.

Statistical Power and Endpoints

In clinical trials, endpoints represent the specific outcomes measured to evaluate the intervention's and . Primary endpoints are predefined measures of the main therapeutic , such as in progression or mortality rates, serving as the basis for determining and regulatory approval. Secondary endpoints assess additional benefits or risks, like improvements in or secondary markers, but require confirmation in further studies to support labeling claims. Surrogate endpoints, such as changes or imaging results, substitute for direct clinical outcomes when they reliably predict benefit, enabling accelerated approvals but necessitating post-approval validation to confirm correlation with clinical events. Composite endpoints combine multiple outcomes (e.g., cardiovascular death, , or ) to increase event rates and trial efficiency, though they demand careful interpretation to avoid diluting true s from disparate components. Statistical power denotes the probability that a trial will detect a true treatment effect of clinically meaningful size, typically set at 80% to 90% to minimize type II errors—false negatives where an effective is erroneously deemed ineffective. is calculated as 1 minus the error rate (β), influenced by factors including the chosen significance level (α, often 0.05), expected (minimal detectable difference in the primary ), variability ( deviation), and sample size. For instance, in a two-arm comparing means, power analysis formulas or software incorporate these parameters to estimate required enrollment; underpowered studies (e.g., power <80%) risk inconclusive results, resource waste, and delayed scientific progress, as evidenced by meta-analyses showing over 50% of in some fields failing to achieve adequate . Trial protocols specify power calculations prospectively for the primary , ensuring sample sizes suffice to detect hypothesized differences with high confidence, while adjusting for dropout rates (e.g., inflating by 10-20%) and interim analyses that may reduce effective . Regulatory bodies like the FDA emphasize powering trials against realistic effect sizes derived from preclinical or prior data, cautioning against over-optimistic assumptions that inflate unrealistically or lead to type I errors if multiplicity adjustments (e.g., ) are ignored across endpoints. In adaptive designs, is reassessed mid-trial using interim data without unblinding, preserving overall integrity while allowing sample size modifications to maintain target above 80%. Failure to prioritize contributes to crises, with empirical reviews indicating that low-power trials overestimate effects and undermine causal inferences about interventions.

Protocol Elements and Adaptations

A clinical trial serves as the foundational document outlining the rationale, objectives, design, conduct, and analysis of the trial, ensuring scientific validity, ethical compliance, and operational feasibility. According to the International Council for Harmonisation (ICH) (R3) guideline, protocols must be clear, concise, and operationally feasible, incorporating a descriptive title, trial synopsis, background information, specific objectives (primary and secondary), and detailed trial design elements such as procedures, blinding methods, and control groups. The U.S. (FDA) requires protocols submitted with (IND) applications to describe patient selection criteria, clinical procedures, safety and efficacy assessments, statistical plans, and data management strategies, with deviations from the protocol tracked separately to maintain integrity. Essential protocol elements include inclusion and exclusion criteria to define the target population, minimizing selection bias; treatment administration schedules, including dosing, duration, and compliance monitoring; and assessment schedules for endpoints, which specify primary outcomes (e.g., survival rates or symptom reduction measured at defined intervals) and secondary outcomes, with methods for data collection such as validated scales or imaging. Safety monitoring components detail adverse event reporting thresholds, stopping rules for harm, and follow-up procedures, while statistical sections cover sample size calculations (often powered to detect a specific effect size with 80-90% power at alpha=0.05), interim analysis plans, and multiplicity adjustments to control false positives. Ethical elements mandate institutional review board (IRB) oversight, informed consent processes, and provisions for subject withdrawal without prejudice. Data quality assurance protocols address handling, storage, and auditing to prevent fraud or errors, as emphasized in ICH guidelines where quality-by-design principles integrate risk-based monitoring from the outset. Protocol adaptations enable modifications during trial conduct to enhance efficiency or address emerging data, but must be prospectively planned to preserve statistical validity and avoid bias inflation. In adaptive designs, protocols pre-specify interim analyses and decision rules—such as re-estimating sample size based on observed variance, dropping ineffective arms, or enriching enrollment for responsive subgroups—while controlling type I error rates through simulation-based methods or adaptive alpha allocation, as outlined in FDA guidance. Fixed protocols limit changes to amendments approved by regulators and IRBs, whereas adaptive approaches, applicable in phases II-III, can reduce trial duration by 20-30% in some models but demand robust simulation to verify operating characteristics pre-trial. Unplanned deviations, like missed visits due to logistical issues, do not constitute adaptations and require documentation and IRB reporting if they risk subject safety or data integrity, per recent FDA draft guidance distinguishing them from intentional, protocol-embedded flexibility. Empirical evidence from adaptive oncology trials shows potential for ethical gains, such as early termination for futility, but underscores the need for transparency in reporting adaptations to regulators to mitigate concerns over inflated efficacy claims.

Placebo and Active Comparator Use

In clinical trials, a is an inert substance or administered to participants to mimic the experience while lacking therapeutic activity, enabling assessment of the specific effects of the investigational beyond psychological or contextual influences such as expectation bias or natural disease progression. This isolates causal by comparing outcomes between the active arm and the arm under blinded conditions, thereby minimizing from non-specific effects that can inflate perceived benefits in open-label studies. Placebo-controlled trials are particularly valuable for establishing proof-of-concept in conditions lacking established therapies, as they provide a rigorous for and reduce the risk of false positives or negatives attributable to subjective reporting. Ethical constraints limit placebo use, as outlined in the World Medical Association's (revised 2013), which permits placebos only when no proven intervention exists or when, for compelling methodological reasons, a less effective alternative is justified without exposing participants to serious or irreversible harm from delayed standard care. Departing from placebo in favor of best proven therapy is required where withholding it would result in inferior outcomes, such as in trials for life-threatening conditions like advanced cancer or , where add-on placebo designs (superimposed on standard treatment) may be employed to assess adjunctive benefits without ethical violation. Controversies arise in areas with partial or debated standards of care, such as psychiatric disorders or , where placebo arms have been criticized for exposing participants to preventable morbidity, prompting calls for stricter equivalence to active controls unless methodological superiority is demonstrated through prior data. Active comparators involve testing the investigational treatment against an established effective therapy, yielding direct evidence of relative , , or equivalence, which is essential for regulatory approval in jurisdictions prioritizing comparative effectiveness. The U.S. (FDA) guidance emphasizes active controls for non-inferiority or superiority designs when use is infeasible due to or high placebo response rates, requiring historical data from placebo-controlled trials of the comparator to set margins for acceptable differences and ensure assay sensitivity. Similarly, the () endorses active comparator trials under ICH E9 principles for confirming via superiority to (via indirect inference) or head-to-head comparisons, particularly in chronic diseases where long-term outcomes demand real-world relevance over isolated efficacy signals. These trials address limitations of placebo designs, such as inability to gauge clinical utility against standards, but demand larger sample sizes and robust endpoints to detect modest differences, with biases in historical controls potentially undermining validity if not corroborated by contemporaneous data. Selection between placebo and active comparator hinges on disease severity, availability of proven therapies, and trial objectives: placebos suit early-phase or orphan indications for unambiguous causality, while active comparators predominate in phase III for therapeutic positioning, as evidenced by cardiovascular outcome trials transitioning from placebo ethics concerns to active designs post-2000s statin benchmarks. Hybrid approaches, like placebo add-on to active controls, balance rigor and equity in responsive conditions, though they risk underpowering if background therapy variability confounds results. Regulatory bodies like the FDA and EMA mandate justification in protocols, with institutional review boards scrutinizing risks to affirm methodological necessity over alternatives.

Ethical Framework

Informed consent in clinical trials embodies the ethical principle of respect for individual autonomy, requiring that participants voluntarily agree to participate after receiving comprehensive disclosure of pertinent information about the study's risks, benefits, procedures, and alternatives. This process ensures participants can make decisions free from coercion or undue influence, rooted in post-World War II reactions to abusive medical experiments. The , promulgated in 1947 by the Nuremberg Military Tribunal, established voluntary consent as "absolutely essential" for permissible human experimentation, emphasizing that subjects must have sufficient knowledge and comprehension to decide freely. The Declaration of Helsinki, adopted by the in 1964 and revised multiple times, further mandates that informed consent be obtained from participants or their legal representatives, prioritizing participant welfare over scientific interests. Regulatory frameworks codify these principles to standardize the consent process. In the United States, the Food and Drug Administration's regulations (21 CFR Part 50) require consent forms to detail the 's purpose, expected duration, procedures distinguishing research from care, foreseeable risks and discomforts, potential benefits, alternative options, protections, compensation for injury, and the right to withdraw at any time without penalty. The International Council for Harmonisation's (ICH-GCP) guidelines, updated in E6(R3) draft in 2023, stipulate that the process be conducted by investigators or delegated staff to promote understanding, with documentation including participant signatures and dates. The of 1979 reinforced autonomy as a core ethical tenet, advocating to safeguard persons from in research involving human subjects. Despite these safeguards, achieving true autonomy faces empirical challenges, notably therapeutic misconception, where participants conflate research participation with individualized therapeutic intent, leading to inflated expectations of personal benefit and diminished appreciation of or risks. This distortion undermines validity, as evidenced by studies showing participants often fail to distinguish trial uncertainties from clinical care guarantees. Comprehension rates remain low; forms frequently exceed recommended 8th-grade readability levels, with research indicating average understanding of only about 50% of key elements like risks and voluntariness. Factors such as limited , advanced age, and document complexity correlate with poorer grasp, prompting interventions like simplified , interactive formats, and teach-back assessments to enhance actual capacity.

Protection of Vulnerable Groups

Vulnerable groups in clinical trials encompass populations at heightened risk of , , or harm due to diminished or capacity, including children, prisoners, pregnant women and fetuses, neonates, individuals with cognitive impairments, economically disadvantaged persons, ethnic minorities, homeless individuals, and those in emergency situations. These groups require additional safeguards because standard processes may fail to ensure voluntary participation, and trial risks could disproportionately affect those with limited decision-making abilities or access to alternatives. Institutional Review Boards (IRBs) must evaluate protocols involving such groups for scientific necessity and ethical justification, prohibiting inclusion unless the research addresses the group's specific health needs or offers direct potential benefit outweighing risks. Under the U.S. Federal Policy for the Protection of Human Subjects (Common Rule, 45 CFR 46), subpart-specific rules apply: Subpart B protects pregnant women, fetuses, and neonates by limiting research to minimal risk or prospective benefit; Subpart C restricts prisoner involvement to studies on prison life or conditions, capping such participation at 50% of subjects; and Subpart D mandates guardian permission and child assent for pediatric trials, with escalating risk thresholds tied to age and therapeutic potential. The World Medical Association's Declaration of Helsinki, revised in 2024, emphasizes that research on vulnerable persons incapable of consent demands extra protections, such as legally authorized representatives, and justifies inclusion only if responsive to the group's priorities, avoiding exploitation while promoting equitable access to trial benefits. The International Council for Harmonisation's Good Clinical Practice guideline (ICH E6(R3), effective 2025) reinforces these by requiring risk-based safeguards, including tailored consent for vulnerable participants like those in nursing homes or impoverished settings, to prevent undue influence. Protections extend to protocol design and oversight: IRBs assess vulnerability prospectively, mandating independent advocates for decisionally impaired subjects and prohibiting coercive incentives, such as payments exceeding fair compensation. For children, FDA guidance from 2022 stresses ethical pediatric investigations, requiring evidence of pediatric relevance before extramural studies and prioritizing non-therapeutic research only at minimal risk levels comparable to daily life. While these measures mitigate historical abuses like coerced prisoner trials in the mid-20th century, critics argue excessive restrictions can exclude vulnerable groups from trials, perpetuating data gaps and unequal treatment efficacy, as evidenced by underrepresentation in drug approvals for conditions prevalent in such populations. The 2024 Helsinki revision addresses this by advocating balanced inclusion to avoid harms from exclusion, such as untested therapies in real-world use.

Conflicts of Interest and Transparency

Financial conflicts of interest in clinical trials primarily arise from by pharmaceutical sponsors, payments to investigators, or stakes, which can influence trial design, data interpretation, and publication decisions. These ties are prevalent, with a 2023 analysis of highly cited trials finding industry or author involvement in over 80% of cases. Such conflicts do not invariably produce but heighten the risk, as incentives align toward positive outcomes that benefit the sponsor's commercial interests over neutral scientific inquiry. Empirical evidence indicates that industry sponsorship correlates with more favorable results. A of randomized trials showed industry-sponsored studies were approximately 30 times more likely to report statistically significant compared to non-industry-funded ones. Sponsorship manifests through selective outcome reporting, omission of negative data, or altered endpoints, distorting the evidence base for drug approvals and clinical guidelines. However, some analyses, such as one on trials, found effect sizes comparable across sponsorship types, suggesting may vary by therapeutic area or study rigor. To mitigate these risks, transparency mandates require disclosure of conflicts in publications and registries. The International Committee of Medical Journal Editors (ICMJE) guidelines, adopted widely since 2001, compel authors to report financial relationships, though evidence questions their efficacy in curbing on citations or . Clinical trial protocols must now be prospectively , enabling of deviations that could mask . In the United States, the Food and Drug Administration Amendments Act (FDAAA) of 2007, enacted September 27, 2007, mandates registration of applicable trials on ClinicalTrials.gov before enrollment and submission of summary results within one year of primary completion, aiming to prevent selective reporting. Non-compliance can incur civil monetary penalties up to $10,000 per day, though enforcement has been inconsistent, with FDA encouraging voluntary adherence amid resource constraints. In the European Union, the Clinical Trials Regulation (EU) No 536/2014, effective January 31, 2022, centralizes submissions via the Clinical Trials Information System (CTIS), requiring detailed protocol and results disclosure to enhance comparability and reduce hidden biases. Despite these frameworks, gaps persist, including incomplete results reporting (estimated at 50% for some trials) and limited access to raw data, underscoring the need for stricter verification and independent audits.

Safety Oversight

Adverse Event Monitoring

Adverse event monitoring in clinical trials involves the systematic identification, documentation, assessment, and reporting of any unfavorable medical occurrences experienced by participants, regardless of causality to the investigational intervention. An (AE) is defined as any untoward medical occurrence in a participant administered a pharmaceutical product, which does not necessarily indicate a causal relationship with the . This ensures participant safety by detecting potential risks early, allowing for adjustments, dose modifications, or trial termination if harms exceed benefits. Monitoring occurs continuously from through follow-up, with investigators responsible for recording AEs in forms and assessing their severity, relatedness, and expectedness based on the or reference safety information. Adverse events are distinguished from serious adverse events (SAEs), which meet specific criteria including , life-threatening conditions, initial or prolonged hospitalization, persistent or significant /incapacity, congenital anomalies, or events requiring medical to prevent such outcomes. SAEs demand expedited reporting: for (IND) studies, sponsors must notify the FDA within 15 calendar days of awareness of any serious, unexpected suspected adverse reaction, with immediate reporting (within 7 days) for events that are both fatal or life-threatening. International harmonization under ICH E2A guidelines specifies that expedited reports apply only to serious events causally related to the study product and inconsistent with prior knowledge, excluding unrelated events or those from non-study arms like . Investigators report AEs to institutional review boards (IRBs) if they represent unanticipated problems involving risks to participants, but routine AEs expected from the disease or do not trigger such notifications unless they alter the risk-benefit profile. Data and safety monitoring boards (DSMBs), independent committees of experts, play a critical role in interim reviews of unblinded safety data, evaluating AE incidence, patterns, and imbalances across arms to assess ongoing risks. DSMBs recommend actions such as continuing, modifying, or halting trials based on benefit-risk assessments, particularly when cumulative suggest excessive harm, as seen in trials where rules prevented further exposure. Sponsors aggregate AE data for periodic regulatory submissions, such as IND annual reports, and employ methods including signal detection via coding systems like to identify emerging safety concerns. Regulatory bodies like the FDA mandate these processes under 21 CFR Part 312, emphasizing assessment to differentiate treatment-related reactions from background events, thereby avoiding over-reporting that could obscure true signals.

Institutional and Regulatory Reviews

Institutional Review Boards (IRBs), also known as ethics committees in some jurisdictions, are mandated to prospectively review and approve clinical trial protocols involving human participants to safeguard their rights, welfare, and safety. Under U.S. federal regulations (21 CFR Part 56), IRBs must consist of at least five members with diverse expertise, including scientific, non-scientific, and community perspectives, and cannot be wholly composed of one gender, profession, or institutional affiliation to minimize bias. Approval criteria require that risks to participants be minimized through protocol design, reasonable in relation to anticipated benefits, and that selection of subjects be equitable; must be obtained under conditions that minimize , and provisions for data monitoring must ensure ongoing safety. IRBs conduct initial reviews before trial initiation and continuing reviews at least annually, or more frequently if risks warrant, examining adverse events, protocol deviations, and amendments to verify persistent compliance with ethical standards. For FDA-regulated trials, IRBs oversee investigator qualifications and ensure the aligns with (GCP), reporting serious issues to regulators if detected. Despite these safeguards, federal audits have identified inconsistencies in IRB operations, such as inadequate documentation of risk assessments, prompting recommendations for enhanced federal oversight to bolster human subject protections in trials ranging from behavioral studies to high-risk interventions. Regulatory reviews complement IRB processes by evaluating scientific merit, manufacturing quality, and preliminary safety data through agencies like the U.S. and the . In the U.S., sponsors submit an application to the FDA prior to phase 1 trials, including preclinical data, manufacturing details, and outlines; the FDA's review, typically completed within 30 days, assesses whether the trial may proceed without unreasonable risk, focusing on dosing, patient monitoring, and stopping rules. For subsequent phases, the FDA requires annual reports and immediate notification of serious adverse events, with the authority to impose clinical holds if safety concerns arise, such as inadequate or organ toxicity signals. In the European Union, the Clinical Trials Regulation (EU No 536/2014), fully applicable since January 31, 2025, centralizes submissions via the Clinical Trials Information System (CTIS), where national competent authorities and ethics committees jointly assess trials within 45-106 days depending on complexity, emphasizing risk-based proportionality for safety oversight and transparency in results reporting. These dual reviews—local institutional ethics scrutiny paired with national or supranational regulatory validation—form a layered barrier against unsafe trial conduct, though empirical analyses indicate that while they reduce overt ethical lapses, gaps in post-approval can persist, as evidenced by historical cases of undetected long-term risks in approved protocols.

Aggregation and Long-Term Tracking

Aggregation of safety data in clinical trials involves pooling adverse event reports from multiple studies or sources to identify signals of rare or low-incidence harms that may evade detection in individual trials due to limited sample sizes. This process employs techniques and statistical analyses to discern patterns, such as disproportionate reporting ratios for specific events, enabling sponsors to characterize risks across a drug's lifecycle. Regulatory bodies like the FDA require sponsors to perform aggregate analyses, including unblinded reviews of pooled data from ongoing trials, to inform safety updates and protocol amendments, though challenges persist in standardizing data formats and mitigating variables across heterogeneous studies. The Safety Assessment Plan (ASAP), proposed by the International Council for Harmonisation, structures this by defining safety topics of interest, data sources, and review frequencies, evolving iteratively to integrate emerging evidence while prioritizing empirical signal detection over speculative risks. Long-term tracking extends participant monitoring beyond primary trial endpoints to capture delayed-onset adverse events, treatment durability, and causal links obscured by short-term observations, particularly critical for interventions like gene therapies where oncogenic risks may manifest years later. Regulators mandate such follow-up—often 5–15 years for advanced therapies—via dedicated protocols that include scheduled assessments, registries, or remote monitoring to track survival, malignancies, and immune responses, though attrition rates exceeding 20–30% in extended cohorts can introduce selection bias and dilute statistical power. Practical implementations leverage electronic health records and patient-reported outcomes for real-time aggregation, but causal inference demands adjustment for confounders like comorbidities and non-adherence, as randomization effects wane over time. In gene therapy trials, for instance, FDA guidance specifies multimodal data collection (e.g., vector shedding, biodistribution) in post-trial phases to validate preclinical predictions against real-world outcomes.

Conduct and Operations

Site Selection and Investigator Roles

Sponsors select clinical trial sites and investigators based on their qualifications, resources, and capacity to ensure trial integrity and participant safety. Under regulations such as 21 CFR 312.53, sponsors must choose investigators qualified by , , and to conduct the investigation properly, while providing them with necessary information like investigator brochures detailing prior observations and risks. criteria, as outlined in ICH E6(R3) (GCP), emphasize adequate facilities, staff, and infrastructure—including laboratories, equipment, and computerized systems—to perform trial activities safely and in compliance with the protocol. Additional considerations include the site's potential to the projected number of eligible participants within the designated period, historical compliance with GCP and regulatory requirements, and geographic access to the target patient population matching criteria. This process often involves site qualification visits to verify readiness, document assessments, and mitigate risks such as inadequate enrollment or issues, with suitability reviewed by institutional review boards or independent ethics committees. The principal investigator (PI) assumes primary responsibility for trial conduct at the selected site, overseeing all aspects to protect participant rights, safety, and welfare while ensuring data reliability. PIs must adhere strictly to the approved protocol, including verifying participant eligibility, administering interventions under direct or supervised conditions, and providing medical care for any trial-related adverse events, with mechanisms like 24-hour contact availability for high-risk studies. Delegation of tasks to sub-investigators or staff is permitted only to qualified individuals, with the PI retaining oversight, maintaining lists of such delegations, and ensuring all personnel are trained in GCP and protocol-specific procedures. Investigators commit to these duties via Form FDA 1572, which requires submission of curricula vitae, financial disclosures, and assurances of oversight. Investigators further ensure by maintaining accurate, contemporaneous records of , drug disposition, and source data, allowing monitoring, audits, and regulatory inspections. They must report unanticipated serious adverse events to sponsors and institutional boards within specified timelines—such as 10 working days under 21 CFR 812.150—and notify of deviations or IRB changes promptly to prevent risks. Non-compliance can lead to regulatory actions, including disqualification, underscoring the causal link between rigorous investigator and overall validity. Sites under PI direction must also manage investigational products securely, from receipt to disposal, and facilitate direct access to records for .

Data Collection and Quality Control

Data collection in clinical trials involves gathering primary and secondary from trial participants, including demographic information, medical histories, treatment interventions, efficacy endpoints, and safety outcomes such as adverse events. According to ICH (GCP) guidelines, data must be recorded accurately, completely, and legibly to ensure credibility, with investigators required to maintain source documents—original records like medical charts, reports, and records—as the foundation for all trial . These source are transcribed into case report forms (CRFs), which can be paper-based or , to standardize collection across sites and facilitate regulatory submission. Electronic data capture (EDC) systems have largely supplanted paper-based methods, offering advantages in efficiency and accuracy; for instance, one simulation study estimated that EDC reduced data collection costs by 55% compared to paper processes while shortening database lock times by 43%. EDC enables real-time data entry, built-in validation rules to flag inconsistencies (e.g., out-of-range values), and remote monitoring, reducing transcription errors that plague paper CRFs, where illegible handwriting or lost pages can compromise integrity. However, hybrid approaches persist in resource-limited settings, and ICH E6(R3) emphasizes minimizing unnecessary data collection to avoid operational complexity, focusing only on variables essential for safety, efficacy, and regulatory requirements. Quality control begins with protocol-defined procedures for data management, including prospective validation of CRFs against study objectives to capture all required elements without redundancy. Source data verification (SDV)—the process of comparing CRF entries against source documents—remains a core mechanism to confirm accuracy, completeness, and verifiability, particularly for critical data like primary endpoints and serious adverse events. Best practices advocate risk-based SDV, targeting high-impact data rather than 100% verification, which FDA guidance supports as a means to optimize resources while maintaining data reliability; full SDV can consume up to 30% of monitoring efforts with diminishing returns on quality. Ongoing quality assurance incorporates centralized monitoring via EDC platforms, which use statistical algorithms to detect anomalies such as protocol deviations or implausible trends across sites, enabling proactive query resolution. Data cleaning involves edit checks, discrepancy management, and reconciliation with external datasets (e.g., central labs), culminating in database lock after independent double-programming verification of datasets. Audits by sponsors or regulators, as mandated by GCP, independently assess compliance, with FDA emphasizing ALCOA+ principles (Attributable, Legible, Contemporaneous, Original, Accurate, plus Complete, Consistent, Enduring, Available) for data integrity. Noncompliance, such as falsification detected in audits, can invalidate trials, underscoring the causal link between rigorous controls and trustworthy outcomes.

Information Technology Integration

Electronic data capture (EDC) systems have become integral to clinical trials, enabling real-time data entry, validation, and monitoring to reduce errors associated with paper-based methods. By 2024, EDC adoption reached approximately 50% of new clinical trials, with projections for continued growth driven by investments expanding at a compound annual rate of 14.7%. These systems facilitate structured from investigators and electronic patient-reported outcomes (ePRO), streamlining workflows and supporting through audit trails. Artificial intelligence (AI) integration enhances trial efficiency across phases, including patient recruitment, protocol design, and predictive analytics for adverse events. The U.S. Food and Drug Administration (FDA) issued draft guidance in January 2025 recommending considerations for AI use in generating data for regulatory decisions, emphasizing validation, transparency, and risk assessment in nonclinical, clinical, and postmarketing stages. AI tools can shorten trial timelines by optimizing site selection and enrollment, potentially reducing costs by identifying suitable participants from electronic health records (EHRs) with higher precision than traditional methods. However, real-world deployment lags behind trial successes due to biases in training data and validation challenges. Blockchain technology addresses concerns by providing decentralized, immutable ledgers for trial records, minimizing tampering risks and enhancing traceability. In permissioned platforms, clinical data transactions are securely logged, fostering trust in multisite studies and supporting compliance with standards like 21 CFR Part 11. Pilot implementations have demonstrated 's role in verifying consent forms and outcome data, though scalability and integration with legacy systems remain barriers. Interoperability between IT systems, such as EDC and EHRs, remains a key challenge, hindered by inconsistent data standards and legacy infrastructure, leading to up to 26.9% of hospital data errors in integrated environments. Efforts like Fast Healthcare Interoperability Resources (FHIR) aim to standardize exchanges, but regulatory compliance, privacy under HIPAA, and cybersecurity threats necessitate robust validation. Digital health technologies, including wearable devices for remote monitoring, further integrate via APIs, improving patient retention but requiring safeguards against data silos.

Analysis and Reporting

Statistical Methods and Interpretation

Statistical methods in clinical trials begin with pre-trial specification of primary and secondary hypotheses, endpoints, and analysis plans to minimize and ensure . Primary endpoints are selected based on clinical , such as overall survival or disease , while statistical power calculations determine sample size, typically targeting 80-90% power to detect a predefined at a two-sided alpha level of 0.05. Power analysis incorporates expected variability, often estimated from prior data or pilot studies, using formulas for continuous outcomes (e.g., t-tests assuming ) or binary outcomes (e.g., chi-square tests). For time-to-event data, methods like Kaplan-Meier and log-rank tests are standard, with Cox proportional hazards models for covariate adjustment. Analysis adheres to principles of randomization and intention-to-treat (ITT), which includes all randomized participants in their assigned groups regardless of compliance, preserving randomization's balance against confounding and providing a pragmatic estimate of real-world effectiveness. Per-protocol (PP) analysis, restricting to adherent participants, complements ITT by assessing efficacy under ideal adherence but risks bias from selective exclusion. Multiplicity adjustments, such as Bonferroni correction or hierarchical testing, control the family-wise error rate when multiple endpoints or subgroups are tested, preventing inflation of type I error. Interim analyses for early stopping employ alpha-spending functions like O'Brien-Fleming boundaries to maintain overall type I error. Interpretation emphasizes confidence intervals (CIs) over p-values alone, as 95% CIs quantify the precision and plausible range of the effect estimate, indicating whether it excludes no effect or aligns with minimal clinically important differences (MCIDs). P-values, while indicating compatibility with the null hypothesis under frequentist assumptions, do not measure effect magnitude, probability of truth, or clinical relevance; a p<0.05 threshold can yield misleading significance for trivial effects in large trials or miss meaningful ones in underpowered studies. Effect sizes, such as standardized mean differences (Cohen's d) or hazard ratios with CIs, better inform causal impact, with regulatory bodies like the FDA recommending sensitivity analyses for estimands—defined treatment effects addressing intercurrent events like dropout—to robustly link statistical findings to trial objectives. Null results require scrutiny for underpowering or unaddressed biases, rather than dismissal, to avoid overinterpreting absence of evidence as evidence of absence.

Handling of Null Results and Biases

In clinical trials, null results occur when statistical analysis fails to reject the , indicating no significant difference between despite adequate power. Such outcomes are scientifically valuable for refuting ineffective interventions, informing future research priorities, and preventing wasteful replication, yet they face systemic underreporting. A of antidepressant trials found that studies with positive findings were nearly four times more likely to be published than those with null or negative results (odds ratio 3.90, 95% CI 2.68-5.68). This publication bias arises from incentives favoring novel or confirmatory evidence, including journal preferences for statistically significant p-values below 0.05, sponsor reluctance to highlight failures, and peer reviewers' harsher scrutiny of null findings. Consequences of suppressing null results include distorted meta-analyses that overestimate treatment effects and perpetuate false positives in the literature. Ioannidis' 2005 analysis demonstrated that in fields with low prior probabilities and small effect sizes— in exploratory trials—most published positive findings are likely false to bias amplification. Empirical tracking via registries like ClinicalTrials.gov reveals discrepancies: as of 2022, only about 50% of registered trials report results, with null outcomes disproportionately absent. To mitigate this, mandatory trial registration and results disclosure, enforced by regulations like the FDA Amendments Act of 2007, compel reporting regardless of outcome, though compliance remains incomplete at around 70% for applicable trials. Biases in trial analysis and reporting further compound issues with null results. Reporting bias manifests as selective omission of unfavorable endpoints or subgroups, where sponsors emphasize positive secondary outcomes while downplaying null primaries; a review of trials identified this in 20-30% of publications. Analytical biases include p-hacking—manipulating data through repeated testing or exclusions to achieve significance—and hypothesizing after results are known (), which inflate type I errors without multiplicity corrections like Bonferroni adjustments. Attrition bias from differential dropout, if not addressed via with multiple imputation, can artifactually favor positive results. Mitigation strategies emphasize pre-trial commitments to transparency and rigor. Pre-registering statistical analysis plans on platforms like or OSF.io prevents post-hoc alterations, reducing by 40-60% in adopting studies. Registered Reports, adopted by over 200 journals since 2013, accept papers based on methodological soundness before results, ensuring null findings are published if protocols are followed; this format has increased null result dissemination in and , with potential extension to clinical fields. Bayesian methods, incorporating prior evidence and uncertainty, offer alternatives to frequentist p-values for handling nulls without rigid thresholds, though adoption lags due to regulatory familiarity with classical approaches. Independent data monitoring committees and regulatory audits further curb sponsor-driven biases, as seen in post-approval reviews uncovering underreported nulls in cardiovascular trials. Despite progress, such as a rising proportion of negative trials in high-impact journals from 2000-2020 (from ~20% to ~35%), entrenched incentives perpetuate selective handling.

Publication Standards and Registries

Clinical trial registries serve as public databases designed to record trial protocols, recruitment status, and outcomes prior to participant enrollment, thereby enhancing transparency and mitigating selective reporting. Prominent registries include , operated by the U.S. National Library of Medicine, the European Union Clinical Trials Register (EU-CTR), and the World Health Organization's International Clinical Trials Registry Platform (ICTRP), which aggregates data from 17 primary registries worldwide. The WHO's mandates prospective registration in a publicly accessible database before the first subject's recruitment to uphold ethical standards and evidentiary integrity. In the United States, the Amendments Act (FDAAA) of 2007 requires registration of "applicable clinical trials"—typically Phase 2-4 interventional studies for drugs, biologics, or devices—within 21 days of enrolling the first participant, with summary results, including adverse events, submitted no later than 12 months after the primary completion date. The European Union's Clinical Trials Regulation (EU No 536/2014), effective since 2022, imposes analogous obligations, harmonizing registration and results disclosure across member states via the Clinical Trials Information System, designated as a WHO primary registry in April 2025. Non-compliance can result in penalties, such as fines or publication bans by adhering journals. These mechanisms aim to curb , where trials with favorable outcomes are preferentially disseminated, distorting meta-analyses and clinical decision-making. Despite these mandates, registries have not fully eradicated biases; studies document persistent discrepancies, with registered results sometimes differing from journal publications in outcomes reported or interpretations provided, and null or negative findings remaining underrepresented in peer-reviewed . For instance, fewer than half of trials culminate in full publications, exacerbating gaps, while trials exhibit lower overall of compared to unregistered ones in systematic reviews. Complementing registries are standardized reporting guidelines to ensure comprehensive, reproducible publications. The 2025 statement, an update to prior versions, outlines a 30-item for randomized controlled trials, covering elements like trial design, participant flow, and subgroup analyses to facilitate critical appraisal and replication. The International Committee of Editors (ICMJE) enforces this by conditioning publication eligibility on prior registration in a WHO primary registry or equivalent, independent of commercial influences. reporting follows guidelines, which align with CONSORT to standardize pre-trial documentation and minimize post-hoc alterations. Adoption of these standards correlates with improved reporting quality, though incomplete adherence persists, underscoring the need for vigilant enforcement to align published evidence with registered intents.

Economics and Incentives

Cost Structures and Funding Sources

Clinical trials incur substantial costs that escalate with each development phase due to increasing participant numbers, trial duration, and complexity. Phase I trials, focused on and dosing in small cohorts of 20-100 healthy volunteers or patients, typically cost around $4 million on average. Phase II trials, evaluating in 100-300 participants, average $13 million. Phase III trials, confirming and monitoring adverse effects in thousands of patients across multiple sites, range from $20 million to over $100 million, often comprising 50-70% of total clinical development expenses for a . Key cost components include site management and investigator payments, which account for approximately 30% of budgets through fees for facilities, staff, and procedures. Patient recruitment and retention represent 20-40% of expenditures, driven by advertising, screening failures, and incentives to achieve enrollment targets amid high ineligibility rates (often 80-90%). Clinical monitoring, , and comprise 15-20%, involving site visits, systems, and to comply with regulatory standards like FDA guidelines. Additional direct costs encompass laboratory testing, imaging, investigational product manufacturing and distribution (5-10%), and central services such as . Indirect costs, including administrative overhead and insurance, add 11-29%. Per-patient costs in industry-sponsored trials average $113,000 to $136,000, with daily operational delays costing around $40,000 across therapeutic areas.
PhaseAverage Cost (USD millions)Primary Cost Drivers
Phase I4Safety assessments, small cohorts, initial dosing
Phase II13 signals, moderate enrollment, early endpoints
Phase III20-100+Large-scale , long-term follow-up, multi-site coordination
Funding for clinical trials is predominantly provided by pharmaceutical and companies, which sponsor over 50% of trials exclusively and contribute to 68% with any involvement, enrolling 8 times more patients than public funders in areas like . This industry dominance reflects incentives tied to and market approval, with sponsors covering the bulk of the estimated $117 million mean clinical-phase cost per drug candidate. agencies, such as the U.S. (NIH), fund approximately 10% of total spending, prioritizing unmet needs like rare diseases or vaccines through grants totaling billions annually (e.g., NIH funding reached levels supporting thousands of trials by 2024). Other sources include non-profits (e.g., ), academic institutions via intramural funding, and for early biotech ventures, though these represent smaller shares and often focus on proof-of-concept rather than pivotal studies. Pharmaceutical sponsors, predominantly large companies, finance the majority of clinical trials to substantiate product safety and efficacy for regulatory bodies such as the FDA, incurring substantial upfront costs offset by potential post-approval revenues from patented therapies. Estimates of the total expenditure for a successfully approved new , accounting for failed projects and opportunity costs, range from $314 million to $4.46 billion, with variations attributable to therapeutic area, inclusion of , and estimation methodology. III trials, essential for confirmatory evidence, represent the largest expense due to their multicenter scale and duration, with direct daily operational costs averaging $55,716 as calculated by the Tufts Center for the Study of based on complexity and . Sponsors mitigate risks through diversification, but trial delays can amplify losses, with indirect revenue forgone estimated at up to $4 million per day for high-stakes programs. Upon approval, successful drugs generate pricing during life, often yielding annual sales in the billions for blockbusters, thus aligning economic incentives with despite high rates exceeding 90% from I to market. Clinical investigators, usually physicians affiliated with research sites or academic centers, derive economic benefits from sponsor payments that reimburse operational expenses and provide supplemental income beyond standard practice revenues. Compensation structures typically include fixed startup fees for site initiation, per-patient enrollment grants covering screening and procedures (often $2,000–$15,000 depending on trial demands and specialty), and milestone payments for data submission, calibrated to fair market value to reflect time and resources. These incentives encourage site participation amid opportunity costs like diverted patient care, but federal regulations and ethical codes strictly prohibit finder's fees, enrollment quotas, or outcome-contingent bonuses to avert undue influence on recruitment or reporting. Despite safeguards, financial dependencies—such as consulting fees or equity—correlate with higher rates of positive results in industry-sponsored trials, as evidenced by systematic reviews showing odds ratios of 4.0 for favorable outcomes compared to independent studies, potentially stemming from protocol design, endpoint selection, or selective publication rather than overt fraud. Disclosure requirements under U.S. Public Health Service rules mandate reporting significant interests exceeding $5,000 annually, yet enforcement gaps and underreporting persist, underscoring ongoing scrutiny of investigator independence.

Participant Compensation and Burdens

Participant compensation in clinical trials typically includes reimbursement for expenses such as travel and lodging, as well as payments for time and inconvenience associated with study procedures. The U.S. (FDA) guidance from 2018 specifies that such payments must be just and fair, with schedules approved by institutional review boards (IRBs) to prevent or undue inducement, where excessive rewards might impair voluntary by causing participants to overlook risks. Reimbursement for direct costs raises no ethical concerns regarding inducement, whereas compensation for participation requires scrutiny to ensure it reflects actual burdens without incentivizing deception or risk minimization. Average compensation varies by trial phase, duration, and invasiveness; for Phase I healthy volunteer studies, payments often equate to $10–$20 per hour, with medians around $3,070 (range $150–$13,000) reported in a analysis of U.S. trials. Higher amounts, up to tens of thousands of dollars, occur in inpatient or high-burden trials like studies, where procedures demand extended commitment. Payments are often disbursed per visit or at completion to align with effort, though upfront stipends risk exacerbating undue inducement debates. Participant burdens encompass physical risks from interventions, psychological stress, time demands (e.g., frequent visits, monitoring), and logistical challenges like transportation, which disproportionately affect lower-income groups. The Declaration of Helsinki mandates pre-trial assessment of these burdens against potential benefits, ensuring they are minimized and justified by scientific validity. IRBs evaluate whether compensation adequately offsets burdens without creating inequities, as uncompensated costs can lead to dropout or biased samples favoring affluent participants. Concerns over undue inducement persist, with critics arguing that overly restrictive interpretations suppress fair pay, impeding recruitment for essential , while ethicists warn that high payments may pressure vulnerable individuals to conceal eligibility issues or downplay risks. Empirical reviews suggest that reasonable, IRB-vetted compensation enhances equity by addressing real burdens, provided transparently details all elements.

Recruitment and Retention

Strategies for Enrollment

Strategies for enrolling participants in clinical trials encompass referral networks, advertising campaigns, digital platforms, and patient-centric approaches, with effectiveness varying by trial type, population, and resource allocation. Physician referrals often prove most cost-effective, as demonstrated in a trial for irritable bowel syndrome where they generated 43 enrollments from 189 calls at a cost of $12 per enrollment and an efficacy index of 3.92, outperforming other methods like mass transit ads (efficacy index -0.12). In-person recruitment and referrals achieve high completion rates, with one study reporting 100% completion among screened participants via these methods, prescreening 37 via referrals to yield 19 screened and all completed. through fliers, both printed and digital, supports broad outreach; in the same study, fliers prescreened 63 individuals, leading to 22 completions at 95.7% rate. and other media ads have mixed yields, producing 24 enrollments from 234 calls in the IBS trial but at higher costs per enrollment compared to referrals. Digital strategies, including and e-recruitment platforms, expand reach and efficiency, particularly post-COVID-19. prescreened 102 in a university-based , resulting in 12 completions at 92.3% rate. Among 24 reviewed platforms, 80% facilitate web-based recruitment with features like -driven patient- matching and electronic on 60% of platforms, accelerating and reducing delays that affect 80% of . Platforms such as Massive Bio employ for prescreening, enhancing matching accuracy beyond demographics in 8% of cases. Patient-centric methods prioritize trial matching to individuals, involving patient groups in consent processes and using visual aids to explain options, which improves suitability and retention by considering behavioral factors alongside eligibility criteria. Combining multiple strategies iteratively, guided by metrics, optimizes overall enrollment, as evidenced by trials meeting targets within 12 months through diversified approaches.

Diversity, Equity, and Matching

Diversity in clinical trial participation encompasses the inclusion of individuals from varied racial, ethnic, age, sex, and socioeconomic backgrounds to assess treatment effects across subgroups where biological, genetic, or environmental factors may influence outcomes. Empirical studies indicate that treatment heterogeneity exists, such as differences in due to genetic variants like those in enzymes, which vary by ancestry and can alter or adverse event rates. For instance, certain antidepressants show reduced in individuals with specific CYP2D6 poor metabolizer phenotypes more prevalent in East Asian populations. Underrepresentation persists; in U.S. trials for new drugs approved in , participants were 75% , 11% /Latino, 8% Black, and 6% Asian, despite Black Americans comprising 13% of the population and facing higher disease burdens in conditions like and certain cancers. Equity involves addressing systemic barriers to participation, including institutional distrust rooted in historical events like the , logistical challenges such as transportation and time burdens disproportionately affecting lower-income groups, and exclusionary eligibility criteria that overlook comorbidities common in underrepresented populations. Data from NIH-funded trials show that one-third lack plans for inclusive across racial/ethnic groups, contributing to disparities where minorities enroll at rates below their disease prevalence. Regulatory efforts, such as FDA guidance requiring diversity action plans since 2022, aim to mitigate these by mandating sponsors assess and enrollment by demographics, though implementation varies and may not always align with trial-specific scientific needs if quotas override eligibility rigor. Matching refers to algorithms and processes that align eligible participants with trial criteria to optimize recruitment, particularly for precision medicine where genetic or phenotypic profiles determine suitability. AI-driven tools, such as NIH's TrialGPT, analyze patient records against trial protocols to identify matches, potentially reducing recruitment delays that affect 80% of trials, with early evidence showing improved eligibility predictions and summaries for diverse or cohorts. These methods enhance efficiency by prioritizing causal fit—ensuring participants' characteristics match the intervention's mechanistic targets—over broad demographic targets, though overreliance on electronic health records risks algorithmic biases if training data underrepresent certain groups. in observational extensions of trials further refines generalizability by balancing covariates to mimic randomized populations.

Barriers and Dropout Factors

Barriers to enrolling participants in clinical trials include limited awareness, stringent eligibility criteria, and logistical challenges. In 2020, 41% of U.S. adults reported no knowledge of clinical trials, contributing to persistently low participation rates estimated at under 5% of eligible cancer patients annually. Exclusion criteria often eliminate a significant portion of potential participants, such as those with comorbidities or specific demographics, while financial burdens like travel costs and lost wages deter others, particularly low-income individuals; a 2015 analysis found households earning below $50,000 were substantially less likely to join cancer trials. Practical obstacles further impede recruitment, encompassing transportation difficulties, lack of coverage for trial-related expenses, and insufficient family support for trial demands. Distance to trial sites and high visit frequency exacerbate these issues, with rural or underserved populations facing disproportionate barriers due to geographic isolation. Mistrust in research institutions, often rooted in historical ethical lapses or perceived lack of among trial staff, also plays a role, as does inadequate referrals stemming from time constraints or unfamiliarity with available trials. These factors collectively result in 80-90% of trials failing to meet enrollment timelines, delaying study completion and inflating costs. Dropout rates average around 30% across clinical trials, introducing as completers may differ systematically from those who withdraw, potentially skewing and outcomes. Common causes include adverse events, with severe reactions like attempted , , rash, and strongly linked to early termination in psychiatric trials. Logistical burdens, such as inconvenient locations, scheduling conflicts, and transportation issues, drive many exits, particularly in long-term studies requiring frequent visits. Other retention challenges involve perceived lack of treatment benefit, where participants disengage if symptoms do not improve or if they feel too ill or too well to continue. Family obligations, work demands, and inadequate communication from study staff further contribute, as do provider-side factors like insufficient incentives or high clinical workloads limiting follow-up support. Health-related predictors, including worsening comorbidities or baseline frailty, independently elevate risk, underscoring the need for tailored to mitigate these causal pathways.

Decentralized and Digital Trials

Decentralized clinical trials (DCTs) incorporate remote or hybrid elements to conduct trial activities outside traditional sites, such as participant homes or local facilities, leveraging technologies like , mobile apps, wearables, and direct-to-participant shipping for and interventions. This approach contrasts with site-based models by minimizing physical site visits while maintaining regulatory standards for , , and . Digital trials, often overlapping with DCTs, emphasize , e-consent, and virtual monitoring to streamline processes. The U.S. (FDA) defines DCTs as trials with decentralized elements executed via telemedicine, mobile tools, or local healthcare providers, without requiring full remoteness. The origins of DCTs trace to early experiments, with Pfizer conducting the first fully decentralized trial in 2011 for an asthma drug, building on conceptual foundations from 2007 that integrated remote monitoring. Adoption accelerated during the COVID-19 pandemic, prompting FDA draft guidance in 2020 and final guidance in 2023, which endorses hybrid models with safeguards for investigator oversight and participant safety. By 2022, 40-45% of clinical studies incorporated decentralized methods, up from 20-25% in 2021, with projections for a 17% rise in decentralization components by end-2023. The global DCT market reached USD 8.6 billion in 2024, driven by cost efficiencies and broader geographic reach. Proponents cite benefits including reduced participant burden, with DCTs showing 19% dropout rates versus 28% in traditional trials, enhanced diversity through remote access, and faster enrollment via digital recruitment. Remote monitoring via wearables improves granularity and self-management adherence, particularly in chronic conditions. However, challenges persist, including regulatory hurdles like ensuring face-to-face interactions for safety assessments and cross-jurisdictional licensure, alongside risks from inconsistent remote collection. Privacy concerns, technological barriers for low-digital-literacy participants, and validation of digital endpoints require rigorous validation to avoid biases in efficacy measurements. Studies in DCTs demonstrate maintained without long-term degradation, but slow overall adoption reflects persistent integration complexities.

AI, Wearables, and Precision Approaches

Artificial intelligence () has been increasingly applied in clinical trials to optimize design, , and , with algorithms predicting patient eligibility and trial outcomes to reduce inefficiencies. A 2025 scoping review of 142 studies from 2013 to 2024 identified 's primary uses in forecasting safety risks (55 studies), efficacy endpoints (46 studies), and operational challenges like delays (45 studies), demonstrating improved accuracy over traditional methods in diverse therapeutic areas. models also automate patient matching by analyzing electronic health records against inclusion criteria, potentially accelerating enrollment by up to 40% in trials, though validation against real-world datasets remains essential to mitigate risks. In trial execution, processes vast datasets to detect adverse events in , as evidenced by III implementations where flagged anomalies 20-30% faster than manual review. Wearable devices enable remote, continuous monitoring in clinical trials, capturing physiological metrics such as , activity levels, and sleep patterns to supplement or replace site visits. In cardiovascular trials, smartwatches like or have tracked endpoints like incidence, with one 2025 case study reporting 95% data compliance in a 500-participant study compared to 70% for self-reported diaries. protocols have integrated wearables for fatigue and mobility assessment, reducing dropout rates by providing objective data that correlates with quality-of-life scores (r=0.75 in motion sensor analyses). Neurological trials, including those for Parkinson's, employ devices like Empatica for tremor quantification, yielding granular datasets that inform dose adjustments and efficacy signals not detectable via periodic clinic evaluations. ECG patches, such as VitalPatch, have been deployed in management trials to monitor rhythms continuously, supporting adaptive designs by triggering early interventions based on threshold breaches. Precision approaches in clinical trials leverage genomic, proteomic, and biomarker profiling to stratify participants and target therapies, shifting from broad populations to molecularly defined cohorts. Umbrella and basket trial designs, advanced since 2020, evaluate multiple targeted agents across tumor-agnostic mutations in single protocols, as in the NCI-MATCH trial where 30% of screened patients with rare variants achieved partial responses to matched drugs. Histology-agnostic trials, such as those for NTRK fusions, have accelerated approvals by focusing on actionable alterations regardless of cancer type, with FDA endorsements for drugs like larotrectinib based on basket data showing 75% response rates in small subgroups. Integration of AI with precision methods enhances variant interpretation and trial simulation, predicting response heterogeneity with AUC values exceeding 0.85 in silico models validated against Phase II outcomes. These strategies, while reducing trial sizes by 20-50% through enriched enrollment, demand robust causal inference to distinguish treatment effects from biomarker confounders.

Real-World Evidence and Adaptive Methods

Real-world evidence (RWE) consists of clinical evidence derived from real-world data (RWD), such as electronic health records, claims data, and patient registries, regarding the usage, potential benefits, or risks of a medical product. The U.S. Food and Drug Administration (FDA) has employed RWE since at least the 1980s for safety surveillance and labeling updates, but formalized its program in 2017 under the 21st Century Cures Act to evaluate its role in regulatory approvals, including as a supplement to randomized controlled trials (RCTs) for effectiveness assessments. RWE offers advantages in capturing heterogeneous patient populations, long-term outcomes, and treatment patterns unavailable in controlled RCT settings, yet it remains observational, prone to confounding, selection bias, missing data, and unmeasured variables that undermine causal inference. Unlike RCTs, which minimize bias through randomization and blinding, RWE cannot reliably isolate treatment effects from extraneous factors, necessitating rigorous analytic methods like propensity score matching or instrumental variables to approximate validity, though these do not fully replicate RCT rigor. Adaptive clinical trial designs enable prospective modifications to ongoing trials—such as altering sample sizes, dropping ineffective arms, or enriching populations—based on interim data analyses, while preserving statistical integrity through pre-specified rules and simulations to control type I error rates. Conceptualized in the 1970s via group sequential methods for , adaptive approaches expanded in the 1990s and 2000s for dose-finding and adaptations, with FDA guidance in 2010 and 2019 endorsing their use in confirmatory trials when biases are mitigated. These designs enhance efficiency by leveraging accumulating evidence to optimize ; for example, multi-arm trials can discontinue underperforming interventions mid-study, potentially reducing participant exposure to ineffective treatments and accelerating approvals. Common types include adaptive , seamless transitions, and Bayesian updates incorporating data, applied most frequently in (over 40% of adaptive trials as of 2023) due to high and patient variability. The convergence of and adaptive methods facilitates hybrid trials that integrate RWD as external controls or historical priors for interim decisions, borrowing strength to inform adaptations without solely relying on internal RCT data. For instance, in I-SPY 2, an adaptive trial for , external RWD informed arm selection and endpoint adjustments, enabling rapid evaluation of multiple therapies while adapting to efficacy signals. FDA has approved drugs like in 2020 using adaptive designs supplemented by RWE for post-hoc analyses, demonstrating feasibility for rare diseases or accelerated pathways. Such integrations promise faster learning from diverse data sources but introduce challenges: RWE's biases can propagate into adaptations, inflating false positives if not calibrated via simulations or Bayesian priors, and regulatory scrutiny demands transparent pre-specification to avoid . Empirical reviews indicate adaptive trials comprise about 15-20% of II/III studies by 2024, with RWE enhancing generalizability yet requiring validation against RCTs to substantiate causal claims.

Controversies and Critiques

Historical Scandals and Ethical Lapses

One of the earliest documented ethical violations in human experimentation occurred during , when Nazi physicians conducted lethal experiments on concentration camp prisoners without consent, including exposure to extreme cold, high altitudes, , and to test survival limits and treatments. These studies, involving thousands of victims primarily , , and political prisoners, resulted in hundreds of deaths and severe mutilations, driven by racial ideology rather than scientific merit. The 1946-1947 at Nuremberg prosecuted 23 physicians, leading to the , which established principles like voluntary and minimization of harm as foundational to ethical research. In the post-war period, U.S.-funded researchers under John Cutler deliberately infected over 1,300 Guatemalan prisoners, soldiers, mental patients, and sex workers with , , and between 1946 and 1948 to evaluate penicillin's efficacy, often without disclosure or , using prostitutes and direct methods. At least 83 deaths occurred among participants, with many untreated initially despite available antibiotics, reflecting a disregard for participant in pursuit of data on progression. The experiments were concealed until 2010, when President Obama apologized, highlighting failures in oversight despite emerging ethical codes. The , initiated by the U.S. Public Health Service in 1932, enrolled 600 African American men in —399 with and 201 controls—promising free medical care but deceiving them about the study's nature and withholding effective treatment even after penicillin became standard in 1947. Participants underwent painful spinal taps misrepresented as therapy, leading to at least 28 deaths from , 100 from complications, and generational transmission, with no therapeutic intent after the 1940s. Exposed by a whistleblower in 1972, it prompted the 1974 , creating institutional review boards and the Office for Human Research Protections. At Willowbrook State School from 1956 to 1971, pediatrician and colleagues intentionally infected over mentally disabled children with and B viruses via fecal matter or to observe disease course and test , obtaining often coerced by linking participation to admission priority amid overcrowding. While some children gained immunity without severe illness, the studies exploited a vulnerable with high baseline rates, raising questions about necessity given natural exposure risks and incomplete risk disclosure. Ethical critiques, amplified by journalist exposure in the 1970s, contributed to stricter standards but defended by Krugman as advancing knowledge leading to . The disaster, unfolding from 1957 when the sedative was marketed in and tested inadequately for teratogenic effects, caused and other defects in over 10,000 infants by 1961 due to maternal ingestion during pregnancy, with trials failing to detect risks in small cohorts lacking pregnant participants. In the U.S., FDA reviewer Kelsey blocked approval in 1960 citing insufficient safety data, averting widespread domestic harm, but pre-market distribution under investigational guise affected dozens. The scandal exposed flaws in voluntary pre-1962 regulations, spurring the Kefauver-Harris Amendments mandating rigorous clinical proof of safety and efficacy, including controlled trials. These lapses, often targeting marginalized groups and prioritizing data over rights, eroded trust and necessitated global reforms like the 1964 , emphasizing risk-benefit analysis and review, though implementation gaps persist in under-regulated contexts.

Industry Influence and Data Manipulation

Pharmaceutical companies exert significant influence over clinical trials through , which constitutes the majority of resources for late-stage . A of 37 studies found that industry-sponsored trials were four times more likely to report outcomes favoring the sponsor's product compared to non-industry-sponsored trials. This manifests in trial design choices, such as selecting comparators that disadvantage alternatives, narrower endpoints, or higher thresholds for adverse events. Empirical analyses confirm that such sponsorship correlates with overstated claims, with industry-funded studies reporting positive results in 85-90% of cases versus 50% for studies. Publication and reporting biases amplify this influence, as negative or null results from industry trials are suppressed. A 2014 network of randomized controlled trials demonstrated that industry-sponsored studies systematically produce larger effect sizes for the sponsor's intervention, independent of study quality. Ghostwriting practices further distort authorship and , where pharmaceutical firms employ professional writers to draft manuscripts, which are then attributed to academic key opinion leaders for credibility. Documents from Merck and revealed systematic use of ghostwriters to promote drugs like Vioxx and hormone therapies while minimizing risks, with academics receiving honoraria but minimal input. Such tactics undermine , as evidenced by internal emails showing companies shaping narratives to align with goals. Data manipulation occurs through falsification, selective omission, or alteration to meet regulatory or commercial thresholds. In the case of Merck's Vioxx, company scientists skewed cardiovascular risk data in the VIGOR trial (published 2000), underreporting heart attack events by reclassifying them, which delayed warnings until 2004 and contributed to thousands of deaths before withdrawal. faced FDA sanctions in 2019 for submitting manipulated preclinical data in its Zolgensma application, including falsified animal study images and metrics not disclosed until after approval on May 24, 2019, prompting a of manufacturing integrity. Broader surveys estimate that 2% of scientists admit to or falsification, with higher rates in industry-linked due to control over . These practices erode trust, as regulators rely on submitted data without independent access, highlighting causal links between financial incentives and compromised integrity.

High Failure Rates and Systemic Inefficiencies

Clinical trials exhibit high failure rates, with only approximately 10.8% of candidates advancing from Phase I to regulatory approval across all areas as of 2023. Phase-specific success rates underscore this attrition: from Phase I to Phase II, about 64% proceed, dropping to 32% from Phase II to Phase III, reflecting escalating challenges in demonstrating and . Overall, roughly 90% of investigational drugs fail during clinical , with Phase II failures reaching 70-80% due primarily to insufficient . Primary causes of these failures include lack of clinical , which accounts for the majority of Phase II and III terminations, alongside safety concerns affecting 17% of failed Phase III trials. Preclinical models often fail to predict responses accurately, leading to biological mismatches that manifest in later phases, while strategic issues like insufficient or commercial viability contribute to about 10% of discontinuations. Analyses of trials from 2010-2017 attribute 90% failure broadly to gaps in clinical translatability, underscoring limitations in early-stage validation. Systemic inefficiencies exacerbate these rates, with drug development timelines averaging 10-15 years from discovery to market, including 9.1 years for clinical phases alone. Capitalized costs per approved drug exceed $2 billion, driven by repeated failures and regulatory demands, rendering the process financially unsustainable for many candidates. Enrollment delays plague 80% of trials, with Phase III studies often extending beyond planned durations by 30%, compounded by data management complexities and site bottlenecks. Post-pandemic slowdowns have revealed entrenched issues like workforce shortages and mounting operational costs, hindering progress despite technological potential. These factors collectively inflate risks, prioritizing incremental therapies over transformative ones and straining R&D resources.
Phase TransitionSuccess RatePrimary Failure Drivers
Phase I to II~64%Safety concerns
Phase II to III~32%Lack of
Phase III to ApprovalVariable, ~50% overall/safety, costs

Regulatory and Public Health Challenges

Regulatory approval processes for clinical trials, governed by agencies such as the U.S. (FDA) and the (), impose extensive requirements for protocol review, ethical compliance, and data safety monitoring, often resulting in significant start-up delays. These delays stem from factors including regulatory submissions, contract negotiations, and insurance approvals, with global randomized trials experiencing median activation times exceeding 12 months in some regions. For instance, discrepancies in FDA and EMA guidelines for therapies like cell and gene treatments necessitate tailored strategies, complicating multi-country studies and increasing administrative burdens. The financial toll of these regulatory hurdles is substantial, with each day of delay in clinical trials estimated to cost sponsors between $40,000 and $500,000 on average, depending on therapeutic area and trial scale. FDA user fees for applications can exceed $500,000, while inefficiencies in site activation amplify overall timelines, contributing to a clinical trial surpassing 90% often linked to regulatory non-compliance alongside and shortfalls. Exponential growth in bureaucratic mandates has further slowed progress, deterring despite intentions to safeguard participants. From a perspective, clinical trials face challenges in ensuring generalizable and beyond controlled settings, particularly in diverse populations where trial homogeneity limits real-world applicability. Post-market systems, reliant on passive reporting mechanisms like the FDA's Adverse Event Reporting System, suffer from profound underreporting—estimated at less than 1% for serious events—hindering timely detection of rare adverse effects after approval. This gap has prompted calls for active using electronic health records, though implementation remains incomplete, as evidenced by GAO reports on FDA's ongoing efforts without full operationalization by 2024. Trial failures frequently arise from concerns, insufficient , or issues, underscoring systemic inefficiencies that delay benefits while exposing participants to risks without proportional safeguards.

Societal Impact

Empirical Successes in Medical Advances

Clinical trials have provided robust evidence for the efficacy of interventions that have substantially reduced mortality and morbidity from infectious diseases, cardiovascular conditions, and cancers. The randomized controlled trial (RCT) design, exemplified by early landmark studies, has enabled causal attribution of benefits by minimizing biases such as selection and confounding. For instance, the UK Medical Research Council's 1948 streptomycin trial for pulmonary tuberculosis, involving 107 patients randomized to streptomycin plus bed rest versus bed rest alone, demonstrated a significant reduction in mortality (7% versus 27% at six months) and radiographic improvement, establishing streptomycin's life-saving role and marking the advent of modern RCTs. Similarly, the 1954 Salk polio vaccine field trial, the largest ever conducted with over 1.8 million U.S. children, showed the inactivated vaccine prevented 60-90% of paralytic poliomyelitis cases compared to controls, leading to its rapid licensure and contributing to the near-eradication of polio in vaccinated populations. In cardiovascular medicine, the ISIS-2 trial (1988), randomizing 17,187 patients with suspected acute to aspirin, , both, or neither, found that aspirin alone reduced 35-day vascular mortality by 23% (from % to 10%), by 25%, and the combination by %, with benefits persisting long-term and informing global guidelines for immediate antiplatelet and thrombolytic therapy.92833-4/fulltext) The Scandinavian Simvastatin Survival Study (4S, 1994), a double-blind RCT of 4,444 high-risk patients with coronary heart disease, showed simvastatin reduced total by 25% and by 35%, yielding a 30% decrease in all-cause mortality (from 12% to 8%) and % in coronary mortality over 5.4 years, overturning prior skepticism about lipid-lowering therapies and establishing statins as a for secondary prevention.90866-5/fulltext) For , adjuvant therapy in receptor-positive has been validated through multiple RCTs, including early Scottish and NATO trials in the 1970s-1980s showing 20-30% reductions in recurrence, with meta-analyses confirming that 5-10 years of treatment lowers 15-year mortality by 30% (e.g., from 25% to 17.5% in node-positive cases), enabling targeted endocrine therapy that has saved millions of lives without the biases plaguing observational data. These examples underscore how rigorous has driven causal, population-level advances, though successes are tempered by the need for ongoing of long-term effects and .

Causal Contributions to Public Health

The of for pulmonary , conducted by the British Medical Research Council in 1948, demonstrated a causal reduction in mortality from approximately 50% to 7% at six months among treated patients compared to controls receiving only . This trial, involving 107 participants, established streptomycin's despite emerging bacterial , prompting its integration into combination therapies that further lowered death rates globally; by the 1950s, such regimens contributed to a decline in U.S. mortality from 39.9 per 100,000 in 1945 to 14.1 per 100,000 by 1955. The causal link from trial evidence to gains is evident in the shift from supportive care to , averting deaths that prior observational data could not reliably attribute to the drug. The 1954 Salk field , the largest in history with over 1.8 million U.S. children randomized or observed, provided causal evidence of 80-90% efficacy against paralytic upon announcement in 1955. This double-blind study, coordinated by the National Foundation for Infantile Paralysis, directly informed mass programs, resulting in U.S. paralytic polio cases falling from 28,000 in 1955 to under 5,600 by 1957 and near elimination by the 1960s. Globally, polio campaigns, grounded in such trial data, reduced incidence by over 99% since 1988, preventing an estimated 20 million cases of ; the trial's overcame skepticism about vaccine safety and efficacy, enabling scalable interventions that disrupted transmission chains. Beyond isolated examples, randomized trials have causally advanced by validating interventions like beta-blockers post-myocardial , which trials in the 1970s-1980s showed reduced five-year mortality by 20-30%, informing guidelines that lowered cardiovascular death rates. Empirical estimates attribute new pharmaceuticals, vetted through trials, to substantial life-year gains; for instance, U.S. drug launches from 1998-2008 correlated with 0.2-0.4 additional life-years by reducing disease-specific mortality. These contributions hinge on trials' ability to isolate effects from confounders, though population-level impacts require effective dissemination and adherence, underscoring that trial causality does not guarantee unmediated real-world outcomes.

Critiques of Over-Reliance and Alternatives

Critics argue that regulatory and evidentiary over-reliance on randomized controlled trials (RCTs) can hinder medical progress by prioritizing over practical applicability, as RCTs often occur in controlled environments that fail to capture real-world patient heterogeneity, comorbidities, and adherence patterns, limiting their . For instance, RCTs may underestimate long-term effects or rare adverse events due to finite sample sizes, which are typically underpowered for detecting harms occurring in fewer than 1% of cases, potentially leading to approvals followed by post-market withdrawals. This emphasis also imposes substantial costs—Phase III trials can exceed $200 million and span several years—delaying access to treatments where preliminary evidence from other sources suggests benefit, particularly in fields like where blinding is infeasible or ethical concerns preclude . Such dependence overlooks confounding factors and generalizability issues inherent to RCTs, including from exclusion criteria that render trial populations unrepresentative of broader clinical practice, as evidenced by studies showing treatment effects diminish outside trial settings. Over-reliance can foster a false , dismissing robust observational data that better reflects population-level outcomes, especially for rare diseases or interventions with historical controls, thereby stifling innovation in resource-limited contexts. Economists like have contended that RCTs, while useful for specific causal questions, cannot substitute for broader theoretical and empirical frameworks, as their narrow focus risks missing systemic interactions and requires supplementation with quasi-experimental methods. Alternatives include pragmatic trials, which embed randomization in routine care to enhance real-world relevance while retaining causal inference strengths, and real-world evidence (RWE) from registries or electronic health records, which facilitates large-scale safety monitoring and effectiveness assessments post-approval. Single-patient (n-of-1) trials offer individualized causal evaluation through repeated measures and crossover designs, proving effective for chronic conditions where population averages obscure personal responses. Regulatory pathways like expanded access or equivalence trials, which compare new agents to standards without placebos, provide expedited options when RCTs are impractical, as seen in FDA approvals leveraging foreign data or historical comparators. Computational modeling and Bayesian approaches further complement by integrating prior mechanistic knowledge with sparse data, enabling predictions without exhaustive randomization, though these require validation against empirical outcomes to mitigate simulation biases.

References

  1. [1]
    NIH's Definition of a Clinical Trial | Grants & Funding
    Sep 18, 2024 · A research study in which one or more human subjects are prospectively assigned to one or more interventions (which may include placebo or other control)
  2. [2]
    Conducting Clinical Trials - FDA
    Jun 30, 2020 · Clinical trials, also known as clinical studies, test potential treatments in human volunteers to see whether they should be approved for wider use in the ...
  3. [3]
    Importance of clinical trials and contributions to contemporary ... - NIH
    Jan 9, 2025 · Firstly, clinical trials help ensure the safety and effectiveness of new interventions. For example, before any drug is approved for use, it ...
  4. [4]
    Evolution of Clinical Research: A History Before and Beyond James ...
    The recorded history of clinical trials goes back to the biblical descriptions in 500 BC. The journey moves from dietary therapy – legumes and lemons – to drugs ...
  5. [5]
    Ethical issues in clinical research - PMC - PubMed Central - NIH
    The ethical issues in clinical research primarily involves protection of rights, safety, and well being of the research participants.
  6. [6]
    Controversies in Clinical Research Ethics | Journal of Law, Medicine ...
    Jan 1, 2021 · Even today, the question of who can consent to participate as a research subject remains controversial, especially in consideration of ...
  7. [7]
    Why 90% of clinical drug development fails and how to improve it?
    Therefore, the success/failure of clinical drug development depends on a delicate balance among clinical dose, efficacy in disease-targeted organs, and toxicity ...
  8. [8]
    The Basics | National Institutes of Health (NIH)
    Apr 24, 2025 · Clinical trials look at new ways to prevent, detect, or treat disease. Clinical trials can study: New drugs or new combinations of drugs; New ...<|separator|>
  9. [9]
    Clinical Trials and Clinical Research: A Comprehensive Review - PMC
    Feb 16, 2023 · The clinical trials aim to evaluate the pharmacodynamic, and pharmacokinetic properties including ADME, efficacy, and safety of new drugs.
  10. [10]
    Objectives and outcomes of a clinical trial - PMC - NIH
    Apr 10, 2023 · A clinical trial aims to find an answer to the main research question or hypothesis that the study plans to answer.Missing: core | Show results with:core
  11. [11]
    [PDF] Considerations for the Design of Early-Phase Clinical Trials of ... - FDA
    This guidance provides OCTGT's current recommendations regarding clinical trials in which the primary objectives are the initial ... to assess safety or efficacy ...
  12. [12]
    Fundamentals of clinical trial design - PMC - PubMed Central
    The objective of clinical trials is to establish the effect of an intervention. Treatment effects are efficiently isolated by controlling for bias and ...Missing: core | Show results with:core
  13. [13]
    Randomized Controlled Trials - PMC - NIH
    Jul 2, 2020 · Randomized controlled trials (RCTs) are considered the highest level of evidence to establish causal associations in clinical research.
  14. [14]
    Randomized Controlled Trials: Part 17 of a Series on Evaluation of ...
    In clinical research, randomized controlled trials (RCTs) are the best way to study the safety and efficacy of new treatments. RCTs are used to answer ...
  15. [15]
    Principles of Good Clinical Trial Design - PMC - NIH
    Clinical trials are a fundamental component of medical research and serve as the main route to obtain evidence of the safety and efficacy of a treatment ...Missing: core | Show results with:core
  16. [16]
    [PDF] ICH guideline E8 (R1) on general considerations for clinical studies
    Oct 14, 2021 · Exploratory and confirmatory safety and efficacy studies ... expectations (E10 Choice of Control Group in Clinical Trials). The ...
  17. [17]
    Clinical Trial Designs - PMC - NIH
    The purpose of the clinical trial is assessment of efficacy, safety, or risk benefit ratio. Goal may be superiority, non-inferiority, or equivalence.Missing: core | Show results with:core
  18. [18]
    [PDF] Guidance for Industry - FDA
    Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing ... are conducted to further evaluate both the safety and efficacy of the drug.
  19. [19]
    Learn About Studies | ClinicalTrials.gov
    Jun 10, 2024 · This page will help you understand clinical research, in general. Choosing to participate in a study is an important personal decision.Missing: applications | Show results with:applications
  20. [20]
    New Drug, Antibiotic, and Biologic Drug Product Regulations - FDA
    Sep 22, 2015 · The Food and Drug Administration (FDA) is revising its regulations governing the submission and review of investigational new drug applications (IND's).
  21. [21]
    Frequently Asked Questions About Therapeutic Biological Products
    May 16, 2024 · What Center has the regulatory responsibility for therapeutic biological products? Both the FDA's Center for Drug Evaluation and Research (CDER) ...<|separator|>
  22. [22]
    Drugs, Devices, and the FDA: Part 2 - PubMed Central
    Class I and II devices are subject to less stringent regulatory processes than Class III devices are; Class I or II device approvals are focused on registration ...
  23. [23]
    Characteristics of Randomized Clinical Trials in Surgery From 2008 ...
    Jun 30, 2021 · In this systematic review of 388 randomized clinical trials, the sizes of surgical trials were small and the focus was on minor clinical events.
  24. [24]
    The challenges faced in the design, conduct and analysis of surgical ...
    This article will consider the design, conduct and analysis of an RCT of a surgical intervention. The issues will be reviewed under three headings.
  25. [25]
    Standards for randomized controlled trials of efficacy of ... - NIH
    May 10, 2024 · The randomized controlled trial (RCT) is the gold standard of evidence for health interventions, including psychological treatments such as psychotherapies.
  26. [26]
    CBT: Review of Randomized Trials - Beck Institute
    Jun 8, 2021 · This systematic review shows that there has been a steady dissemination and adoption of the cognitive-behavioral therapies in practitioner's clinical work.<|control11|><|separator|>
  27. [27]
    [PDF] Evolution of clinical trials throughout history - Marshall University
    The arrival of the placebo marked an important breakthrough in the history of clinical research. In 1863, Austin Flint conducted a trial involving patients ...Missing: key | Show results with:key
  28. [28]
    Lind and scurvy: 1747 to 1795 - PMC - NIH
    During a 10-week absence from shore, 80 out of 350 sailors were struck down by scurvy, and Lind's prospective controlled experiment—in which he compared the ...
  29. [29]
    Lind J (1753) - The James Lind Library
    In 1747, James Lind, a Scottish naval surgeon faced with uncertainty about which of many proposed treatments for scurvy to use, compared six of them in a ...
  30. [30]
    Edward Jenner and the history of smallpox and vaccination - NIH
    In May 1796, Edward Jenner found a young dairymaid, Sarah Nelms, who had fresh cowpox lesions on her hands and arms (Figure 3).
  31. [31]
    Edward Jenner's Case Series on Smallpox Vaccination
    Dec 1, 2020 · Edward Jenner was aiming for when he published his observations of 23 case-patients in 1798, all of whom received inoculations with cowpox in an attempt to ...
  32. [32]
    Nuremberg Code: Directives for Human Experimentation | ORI
    No experiment should be conducted where there is an a priori reason to believe that death or disabling injury will occur. The degree of risk to be taken should ...
  33. [33]
    The MRC randomized trial of streptomycin and its legacy - NIH
    Austin Bradford Hill, Professor of Medical Statistics at the London School of Hygiene and Tropical Medicine, was one of the members of the committee. The ...
  34. [34]
    “A calculated risk”: the Salk polio vaccine field trials of 1954 - NIH
    The results, announced in 1955, showed good statistical evidence that Jonas Salk's killed virus preparation was 80-90% effective in preventing paralytic ...
  35. [35]
    Promoting Safe & Effective Drugs for 100 Years - FDA
    Apr 23, 2019 · In October 1962, Congress passed the Kefauver-Harris Drug Amendments to the Federal FD&C Act. Before marketing a drug, firms now had to ...
  36. [36]
    WMA Declaration of Helsinki – Ethical Principles for Medical ...
    The World Medical Association (WMA) has developed the Declaration of Helsinki as a statement of ethical principles for medical research involving human ...
  37. [37]
    Effects on Research | The U.S. Public Health Service ... - CDC
    Sep 4, 2024 · In 1974, the National Research Act was signed into law, creating the ... IRBs decide whether research protocols meet ethical standards.
  38. [38]
    E6(R2) Good Clinical Practice: Integrated Addendum to ICH E6(R1)
    Nov 9, 2023 · Good Clinical Practice (GCP) is an international ethical and scientific quality standard for designing, conducting, recording and reporting trials
  39. [39]
    [PDF] Integrated Addendum to ICH E6(R1): Guideline for Good Clinical ...
    Nov 9, 2016 · A systematic and independent examination of trial related activities and documents to determine whether the evaluated trial related activities ...
  40. [40]
    Critical Path Initiative - FDA
    Apr 11, 2025 · The Critical Path Initiative (CPI) is FDA's strategy to transform how FDA-regulated medical products are developed, evaluated, and manufactured.
  41. [41]
    FDAAA 801 and the Final Rule | ClinicalTrials.gov
    Sep 10, 2025 · A summary of the regulatory requirements for clinical trial registration and results reporting described in FDAAA 801 and 42 CFR Part 11.
  42. [42]
    Update on the clinical trial landscape: analysis of ClinicalTrials.gov ...
    Oct 6, 2022 · The clinical trial landscape has evolved over the last two decades, shaped by advances in therapeutics and drug development and innovation ...
  43. [43]
    Step 2: Preclinical Research - FDA
    Jan 4, 2018 · FDA requires researchers to use good laboratory practices (GLP), defined in medical product development regulations, for preclinical laboratory ...
  44. [44]
    Drug Development and Review Definitions - FDA
    Aug 20, 2015 · At the preclinical stage, the FDA will generally ask, at a minimum, that sponsors: (1) develop a pharmacological profile of the drug; (2) ...
  45. [45]
    In Vitro vs In Vivo Preclinical Studies - News-Medical
    Feb 23, 2021 · In preclinical studies, only animals are used for in vivo tests as this stage comes prior to human testing to ensure its safety. tissue ...
  46. [46]
    Preclinical Testing Techniques: Paving the Way for New Oncology ...
    During the preclinical stage, the screening of multiple compounds is performed with in vitro and in vivo platforms, including animal testing. Two ...
  47. [47]
    Preclinical Regulatory Requirements | Social Science Research ...
    Preclinical studies are also subject to the FDA regulations known as Good Laboratory Practices (GLP) for Nonclinical Laboratory Studies, 21 CFR 58. The GLP ...
  48. [48]
    Preclinical Studies in Drug Development | PPD
    Preclinical studies must adhere to strict regulatory guidelines and ethical considerations. Regulatory bodies such as the FDA and the European Medicines ...
  49. [49]
    How long do clinical trials take? - Antidote.me
    Apr 4, 2025 · This phase is referred to as preclinical research, which can take anywhere from one to six years. During this phase, in vitro and in vivo ...
  50. [50]
    [PDF] Guidance for Industry - S6 Preclinical Safety Evaluation of ... - FDA
    The objectives of the preclinical safety studies are to define pharmacological and toxicological effects not only prior to initiation of human studies but ...
  51. [51]
    [PDF] Safety Considerations in Clinical Drug Development - FDA
    Dec 6, 2023 · Guidance for Industry: Estimating the Maximum Safe Starting Dose in Initial Clinical Trials ... • Overview of safety in Phase 1 trials.
  52. [52]
    Trial Phases 1, 2 & 3 Defined | Clinical Research Management (CRM)
    Phase I trials are concerned primarily with establishing a new drug's safety and dose range in about 20-100 healthy volunteers. How a drug is absorbed, ...
  53. [53]
    Dose Escalation Methods in Phase I Cancer Clinical Trials - NIH
    Here we review dose escalation methods for phase I trials, including the rule-based and model-based dose escalation methods that have been developed to ...
  54. [54]
    [PDF] Guidance for Industry - FDA
    Jul 6, 2005 · Toxicity should be avoided at the initial clinical dose. However, doses should be chosen that allow reasonably rapid attainment of the phase 1 ...
  55. [55]
    [PDF] Phase I Clinical Trial Design - Biometric Research Program
    The dose escalation rules (Table 1) proceed as follows, escalating in cohorts of 3-6 patients per dose level 35. Three patients are treated at the current dose ...
  56. [56]
    Designing phase I oncology dose escalation using dose–exposure ...
    Jul 19, 2022 · Dose-escalation designs for phase I clinical trials in oncology begin by assigning the first patient (or cohort of patients) to a starting dose ...
  57. [57]
    [PDF] Expansion Cohorts: Use in First-in-Human Clinical Trials to Expedite ...
    toxicities have been observed in the dose escalation phase that may be fatal or result in serious morbidity, FDA strongly recommends that expansion be ...<|separator|>
  58. [58]
    Mastering Dose Escalation Studies Part 1: Optimizing Efficiency in ...
    Apr 8, 2024 · This methodology is particularly beneficial and increasingly common in phase 1 clinical trials, which focus primarily on safety, tolerability, ...
  59. [59]
    Step 3: Clinical Research - FDA
    Jan 4, 2018 · Researchers design clinical trials to answer specific research questions related to a medical product. These trials follow a specific study plan ...
  60. [60]
    An Overview of Phase II Clinical Trial Designs - PMC - NIH
    Phase II trials are designed to determine if the new treatment has sufficiently promising efficacy to warrant further investigation in a large-scale randomized ...
  61. [61]
    Randomized Phase II Designs - PMC - NIH
    We review the principal statistical designs for historically controlled and randomized phase II trials, along with their advantages, disadvantages, and ...
  62. [62]
    [PDF] Clinical Trial Design TF (CTD-TF) Recommendations for Phase 2
    Jul 24, 2009 · (2) For randomized trials, blinded designs are encouraged where feasible. While placebo controlled trials are challenging, they are encouraged.
  63. [63]
    The Design of Phase II Clinical Trials Testing Cancer Therapeutics ...
    Mar 14, 2010 · Randomization is usually essential for a phase II trial in which progression-free survival is the most appropriate endpoint. Nonetheless, single ...
  64. [64]
    Phase II Trials in Drug Development and Adaptive Trial Design - JACC
    Jun 24, 2019 · Following phases II and III, 30.7% and 58.1% of drugs fail, respectively (4). The picture is even worse for cardiovascular (CV) agents; 6.6% of ...
  65. [65]
    Phase II Clinical Trials: Goals, Examples, and Real-Life Insights
    May 28, 2025 · Randomization and Blinding Approaches. Randomization and blinding reduce bias and strengthen the validity of Phase II trial results. Single- ...
  66. [66]
    [PDF] 22 Case Studies Where Phase 2 and Phase 3 Trials Had ... - FDA
    These unexpected results could occur even when the phase 2 study was relatively large and even when the phase 2 trials assessed clinical outcomes. In two cases ...
  67. [67]
    Phase III Trial – Medical School Office of Research
    Phase III studies are randomized controlled multicenter trials on large patient groups (300–3,000 or more depending upon the disease/medical condition studied) ...
  68. [68]
    The Difference between Phase 2 and Phase 3 Clinical Trials
    Jan 10, 2022 · Length of study: Phase 2 clinical trials last on average about 2 years, whereas Phase 3 clinical trials are often much longer – from 2.5 years ...What are Phase 1 clinical trials? · What are Phase 3 clinical trials?
  69. [69]
    Phase III Clinical Trials: Ultimate Guide & Real-World Case Studies
    Jun 8, 2025 · The Role of Phase III in the Clinical Trial Lifecycle. Phase III trials are the final and most critical evaluative stage before a drug or ...
  70. [70]
    Phases of Clinical Trials | American Cancer Society
    Jun 3, 2025 · Phase III clinical trials compare the safety and effectiveness of the new treatment against the current standard treatment. Side effects of the ...
  71. [71]
    22 Case Studies Where Phase 2 and Phase 3 Trials Had Divergent ...
    Dec 21, 2017 · FDA studied 22 recent cases in which promising phase 2 clinical trial results were not confirmed in phase 3 clinical testing.
  72. [72]
    Phase 3 Trial | NIH - Clinical Info .HIV.gov
    Phase 3 trials are conducted to confirm and expand on safety and effectiveness results from Phase 1 and 2 trials, to compare the drug to standard therapies.
  73. [73]
    Maximizing the value of phase III trials in immuno-oncology - NIH
    Sep 29, 2022 · Examples of agents that have been approved in ... phase III IMpower110 study that led to regulatory approval for atezolizumab monotherapy.
  74. [74]
    Phase IV of Drug Development - PMC - NIH
    Phase IV includes post-marketing surveillance (PMS) studies, evaluating real-world effectiveness and safety, and is considered the "real test" of a drug.Missing: EMA | Show results with:EMA
  75. [75]
    Understanding Clinical Phase 4: Post-Market Surveillance and Its ...
    Clinical Phase 4, or post-market surveillance, occurs after drug approval, monitoring safety and effectiveness in real-world settings.
  76. [76]
    Cardiovascular Events Associated with Rofecoxib in a Colorectal ...
    The Adenomatous Polyp Prevention on Vioxx (APPROVe) Trial was designed to evaluate the hypothesis that three years of treatment with rofecoxib would reduce the ...
  77. [77]
  78. [78]
    Drug safety on trial - PMC - NIH
    The APPROVe (Adenomatous Polyp Prevention on Vioxx) study aimed to determine if rofecoxib prevented the malignant transformation of colon polyps and excluded ...
  79. [79]
    FDA in Brief: FDA modernizes clinical trial designs and approaches ...
    Sep 28, 2018 · FDA modernizes clinical trial designs and approaches for drug development, proposing new guidance on the use of adaptive designs and master protocols.
  80. [80]
    Adaptive Designs for Clinical Trials of Drugs and Biologics Guidance
    Nov 29, 2019 · The guidance describes important principles for designing, conducting, and reporting the results from an adaptive clinical trial. The guidance ...
  81. [81]
    [PDF] Adaptive Designs for Clinical Trials of Drugs and Biologics - FDA
    ADAPTIVE DESIGNS BASED ON NON-COMPARATIVE DATA. This section addresses adaptive clinical trial designs in which adaptations are based entirely on analyses of ...
  82. [82]
    Adaptive clinical trial designs in oncology - PMC - PubMed Central
    Adaptive designs provide a flexible and effective way to conduct clinical trials. The designs have potential advantages of improving the study power, reducing ...
  83. [83]
    Recent innovations in adaptive trial designs: A review of ... - NIH
    There are three general types: basket, umbrella, and platform trials. Basket trials evaluate single therapies across multiple diseases with common molecular ...
  84. [84]
    Basket and Umbrella Clinical Trials – What Are They and How Can ...
    Oct 18, 2024 · Basket trials, also called bucket trials, are designed to test a single drug or combination therapy across multiple cancer types that share a common genetic ...
  85. [85]
    Clinical trial design in the era of precision medicine - PMC
    Aug 31, 2022 · This review focuses on the design, advantages, limitations, and challenges of a spectrum of clinical trial designs in the era of precision oncology.
  86. [86]
    E20 Adaptive Designs for Clinical Trials September 2025 - FDA
    Sep 30, 2025 · The draft guidance focuses on principles for the planning, conduct, analysis, and interpretation of clinical trials with an adaptive design that ...
  87. [87]
    Different Types of Clinical Trial Designs - Alimentiv
    This guide provides a look at clinical trial designs, suggests the best designs for research objectives and highlights their key features.
  88. [88]
    Alternative clinical trial designs - PMC - NIH
    Feb 4, 2020 · These include pragmatic trials, cluster randomization, cluster randomized stepped wedge designs, factorial trials, and adaptive designs.
  89. [89]
    The Promising Future for Complex Innovative Trial Design in Clinical ...
    Complex Innovative Trial Design (CID) is an emerging approach that integrates novel statistical and methodological techniques to address these difficult ...
  90. [90]
    Interacting with the FDA on Complex Innovative Trial Designs for ...
    Apr 8, 2024 · This document provides guidance to sponsors and applicants on interacting with the FDA on complex innovative trial design (CID) proposals ...
  91. [91]
    The importance of randomization in clinical research - PMC - NIH
    Aug 8, 2022 · Randomization attempts to reduce the systemic error introduced by observational studies by ensuring equal distribution of prognostic factors ...
  92. [92]
    A roadmap to using randomization in clinical trials
    Aug 16, 2021 · Randomization is the foundation of any clinical trial involving treatment comparison. It helps mitigate selection bias, promotes similarity ...
  93. [93]
    Understanding the role of randomization in clinical research and ...
    Randomization is essential for statistical validity, learning from experience, and optimizing care in uncertainty, playing crucial scientific and ethical roles.
  94. [94]
    The Magic of Randomization versus the Myth of Real-World Evidence
    Feb 12, 2020 · Randomization Provides Evidence about Treatment Effects That Can Be Trusted · Obstacles to Randomized Trials Should be Removed to Protect ...<|separator|>
  95. [95]
    [PDF] ich harmonised tripartite guideline statistical principles for clinical ...
    Blinding or masking is intended to limit the occurrence of conscious and unconscious bias in the conduct and interpretation of a clinical trial arising from the ...
  96. [96]
    Blinding in Clinical Trials: Seeing the Big Picture - PMC
    The terms single-blind, double-blind, and triple-blind are often used to describe studies in which one, two, or three parties, respectively, are blinded to ...
  97. [97]
    [PDF] Guidance for Industry - FDA
    Assay sensitivity is a property of a clinical trial defined as the ability to distinguish an effective ... means of a trial showing non-inferiority of the ...
  98. [98]
    Use and misuse of blinding in randomized controlled trials
    The use of blinding strengthens the credibility of randomized controlled trials (RCTs) by minimizing bias.
  99. [99]
    Double-Blind Study - StatPearls - NCBI Bookshelf - NIH
    Jul 17, 2023 · Single-, double-, and triple-blinding are commonly used blinding strategies in clinical research. A single-blind study masks the subjects ...
  100. [100]
    [PDF] Basics of Clinical Trial Design - FDA
    Control groups allow discrimination of patient outcomes caused by the test treatment from outcomes caused by other factors. • What would have happened to ...
  101. [101]
    [PDF] ICH Topic E 10 Choice of Control Group in Clinical Trials Step 5
    ... active control trials can include a placebo group, multiple-dose groups of the test drug, and/or other dose groups of the active control. Comparative dose ...
  102. [102]
    [PDF] Choice of Control Group and Related Issues in Clinical Trials E10
    Randomization and blinding are the two techniques usually used to minimize the chance of such bias and to ensure that the test treatment and control groups are.
  103. [103]
    [PDF] Multiple Endpoints in Clinical Trials - Guidance for Industry - FDA
    This FDA guidance document for industry covers multiple endpoints in clinical trials, including general principles and nonbinding recommendations.
  104. [104]
    Multiple Endpoints in Clinical Trials | FDA
    Apr 16, 2024 · The purpose of this guidance is to describe various strategies for grouping and ordering endpoints for analysis and applying some well- ...Missing: surrogate | Show results with:surrogate
  105. [105]
    Table of Surrogate Endpoints That Were the Basis of Drug Approval ...
    Aug 29, 2025 · This surrogate endpoint table includes surrogate endpoints that sponsors have used as primary efficacy clinical trial endpoints for approval of new drug ...
  106. [106]
    Sample size estimation and power analysis for clinical research ...
    A related quantity is the statistical power; this is the probability of identifying an exact difference between 2 groups in the study samples when one genuinely ...
  107. [107]
    Statistical Power and Why It Matters | A Simple Introduction - Scribbr
    Feb 16, 2021 · Statistical power, or sensitivity, is the likelihood of a significance test detecting an effect when there actually is one.
  108. [108]
    Power Analysis and Sample Size, When and Why? - PMC - NIH
    The power analysis is performed by some specific tests and they aim to find the exact number of population for a clinical or experimental study.
  109. [109]
    [PDF] Estimating Power and Sample Size - Stanford Medicine
    Power = 1 – β (typically 1 – 0.2 = 0.8). • Sample size planning aims to select a sufficient number of subjects to keep α and β low without making the study too ...
  110. [110]
    Sample size, power and effect size revisited: simplified and practical ...
    Although increasing the sample size is suggested to decrease the Type II errors, it will increase the cost of the project and delay the completion of the ...
  111. [111]
    Type I and Type II Errors and Statistical Power - StatPearls - NCBI
    By limiting type I and type II errors, healthcare providers can ensure patients' decisions based on research outputs are safe.[8] Additionally, while power ...
  112. [112]
    [PDF] GUIDELINE FOR GOOD CLINICAL PRACTICE E6(R3) - ICH
    Jan 6, 2025 · CLINICAL TRIAL PROTOCOL AND PROTOCOL AMENDMENT(S). Clinical trials should be described in a clear, concise and operationally feasible protocol.
  113. [113]
    IND Applications for Clinical Investigations: Clinical Protocols - FDA
    Oct 9, 2015 · Clinical trial protocols should include a clear description of trial design and patient selection criteria as well as description of clinical ...
  114. [114]
    Protocol and Conduct - FDA
    Apr 7, 2015 · The protocol shall contain but shall not necessarily be limited to the following information: A descriptive title and statement of the purpose ...
  115. [115]
    Protocol Deviations Explained: Understanding the FDA's Draft ...
    Sep 8, 2025 · In December 2024, the FDA released a draft guidance titled “Protocol Deviations for Clinical Investigations of Drugs, Biological Products, and ...
  116. [116]
    The Ethics of Placebo-controlled Trials: Methodological Justifications
    Sep 12, 2013 · Most people accept the use of placebo controls in trials for conditions with no effective treatment. However, PCTs raise ethical concerns when a ...
  117. [117]
    The ethics of placebo-controlled trials: Methodological justifications
    This article sets out key considerations relevant to considering whether methodological reasons for a placebo control are compelling.
  118. [118]
    The Declaration of Helsinki and Clinical Trials: A Focus on Placebo ...
    Thus, the advantages associated with placebo use include both reducing the number of subjects at risk and preventing false negative and false positive results.
  119. [119]
    Ethical Use of Placebo Controls in Research | AMA-Code
    Used appropriately, a placebo control can provide valuable data, particularly when there is no accepted therapy for the condition under study.
  120. [120]
    The ethics of placebo-controlled trials: The case of asthma
    Some have argued that placebo-controlled trials (PCTs) are unnecessary and unethical in conditions such as asthma for which there is standard therapy.
  121. [121]
    Is It Always Unethical to Use a Placebo in a Clinical Trial? - PMC - NIH
    Mar 29, 2005 · Placebo-controlled trials are controversial. Critics of such trials argue that if a proven effective therapy exists, a placebo should not be used.
  122. [122]
    Use of Active Comparator Trials for Topical Medications in ...
    Apr 7, 2021 · Active comparator trials designs, in which the new medication is compared with an established treatment, provide data that can guide clinical decision-making.
  123. [123]
    [PDF] Non-Inferiority Clinical Trials to Establish Effectiveness - FDA
    It must be estimated based on the past performance of the active control, preferably in placebo-controlled trials, and then assumed to hold for the NI study ...
  124. [124]
    [PDF] E 9 Statistical Principles for Clinical Trials Step 5
    This document is a note for guidance on statistical principles for clinical trials, a harmonised tripartite guideline.
  125. [125]
    Transitioning to active-controlled trials to evaluate cardiovascular ...
    Aug 24, 2022 · Using an active comparator addresses the clinical and ethical concerns of a placebo-controlled trial as it provides high-risk participants in ...
  126. [126]
    Comparison of the FDA and EMA guidance on drug development in ...
    Jul 22, 2025 · Therefore, the FDA and EMA both support double-blind trial designs with parallel arms, including active comparator and/or placebo, and require ...
  127. [127]
    Ethics of Placebo Use in Pediatric Clinical Trials | Hypertension
    Placebo use could also be ethical if placebos were used as an “add-on” to current treatment or if currently available treatments were themselves risky to the ...
  128. [128]
    In 2021 When Is It Unethical to Use a Placebo in a Clinical Trial?
    Use of a placebo as a comparator is only ethically justified when there is no proven effective intervention, or there are no harms from delaying or forgoing ...
  129. [129]
    Nuremberg Code - UNC Research
    The Nuremberg Code is a ten-point statement delimiting permissible medical experimentation, requiring voluntary consent, and that it should benefit society.Missing: Helsinki | Show results with:Helsinki
  130. [130]
    [PDF] Informed Consent Guidance for IRBs, Clinical Investigators ... - FDA
    This guidance represents the current thinking of the Food and Drug Administration (FDA or Agency) on this topic. It does not create any rights for any ...
  131. [131]
    [PDF] E6(R3) Good Clinical Practice (GCP) | Guidance for Industry - FDA
    May 19, 2023 · 9.4 Clinical trials should incorporate efficient and robust processes for managing records (including data) to help ensure that record integrity ...<|separator|>
  132. [132]
    Read the Belmont Report | HHS.gov
    Jul 15, 2025 · It is a statement of basic ethical principles and guidelines that should assist in resolving the ethical problems that surround the conduct of research with ...
  133. [133]
    Therapeutic Misconception in Research Subjects: Development and ...
    The characteristics of TM suggest that it may undercut the process of obtaining meaningful consent to clinical research participation by distorting participants ...
  134. [134]
    Therapeutic misconception and the appreciation of risks in clinical ...
    Failure to distinguish the consequences of research participation from receiving ordinary treatment may seriously undermine the informed consent of research ...
  135. [135]
    Readability Standards for Informed-Consent Forms as Compared ...
    When documents are incomprehensible, health care providers may risk liability. Obtaining informed consent for participation in medical research is particularly ...
  136. [136]
    The reality of informed consent: empirical studies on patient ...
    Jan 14, 2021 · Research on patients' comprehension of an informed consent's basic components shows that their level of understanding is limited.
  137. [137]
    Informed Consent for Clinical Trials: a Comparative Study of ...
    Factors associated with decreased comprehension of informed consent include limited education, increasing age of the patient, and the readability of the consent ...
  138. [138]
    45 CFR 46 - HHS.gov
    Feb 11, 2025 · 45 CFR 46 is HHS regulations for protecting human subjects in research, including the Common Rule (subpart A) and additional protections for ...45 CFR 46 FAQs · Subpart B · Subpart D · OHRP Expedited Review
  139. [139]
    Vulnerable and Other Populations Requiring Additional Protections
    Sep 9, 2024 · The populations requiring additional protections are pregnant women, human fetuses, neonates, children, and prisoners.
  140. [140]
    Vulnerable Subjects | CHOP Research Institute
    The CIOMS Guidelines also state that Special justification is required for inviting vulnerable individuals to serve as research subjects and, if they are ...<|separator|>
  141. [141]
    Clinical research: Protection of the “vulnerable”?
    Current ethical guidelines prohibit or severely limit what types of research can be performed involving these “vulnerable” populations. Although this might ...
  142. [142]
    World Medical Association announces adoption of the 2024 ...
    Nov 6, 2024 · The Declaration promotes fair and responsible inclusion of vulnerable populations, balancing the harms of inclusion and exclusion while ...<|separator|>
  143. [143]
    Investigator experiences with financial conflicts of interest in clinical ...
    Financial conflicts of interest (fCOI) can introduce actions that bias clinical trial results and reduce their objectivity.
  144. [144]
    Industry Involvement and Transparency in the Most Cited Clinical ...
    Nov 14, 2023 · This cross-sectional study of 600 of the most-cited clinical trials published after 2018 found that industry involvement was very common for funding, ...
  145. [145]
    Financial conflicts of interest in clinical research
    Aug 20, 2018 · Conflicts of interest do not necessarily cause biased trial results, but create the risk thereof. Unfortunately, our knowledge of what factors ...<|separator|>
  146. [146]
    2 Do Sponsoring Organizations Influence Research?
    Industry-sponsored studies were about 30 times more likely than non-industry sponsored studies to report statistically significant efficacy estimates.
  147. [147]
    Sponsorship bias in clinical trials: growing menace or dawning ... - NIH
    Sponsorship bias is the distortion of design and reporting of clinical experiments to favour the sponsor's aims.
  148. [148]
    Industry sponsorship bias in research findings: a network meta ...
    Oct 3, 2014 · Our analysis shows that the findings obtained from industry sponsored statin trials seem similar in magnitude as those in non-industry sources.
  149. [149]
    Conflict of interest disclosure in biomedical research
    May 3, 2016 · These biases, when hidden, can affect clinical decision-making by making interventions appear safer or more effective than they really are.
  150. [150]
    Do Conflict of Interests Disclosures Work? Evidence from Citations ...
    Feb 2, 2023 · Financial ties between drug companies and medical researchers are thought to bias studies published in medical journals.
  151. [151]
    Food and Drug Administration Amendments Act (FDAAA) of 2007
    Mar 29, 2018 · Complete or check registration for a product or facility ... Advice/information about a medical device, drug, food, or other FDA-regulated product ...
  152. [152]
    Clinical Trial Reporting Requirements | ClinicalTrials.gov
    Aug 7, 2025 · Guidance for sponsors and investigators about the legal and policy requirements for registering clinical trials and submitting results to ...
  153. [153]
    The Importance of Clinical Trial Transparency and FDA Oversight
    Dec 4, 2023 · The clinical trial is the most reliable method to determine the benefits and risks of interventions. The formation of ClinicalTrials.gov in ...Missing: definition | Show results with:definition
  154. [154]
    Clinical Trials Regulation | European Medicines Agency (EMA)
    Feb 10, 2023 · Under the Regulation, clinical trial sponsors must submit all new clinical trial applications in the Clinical Trials Information System (CTIS) ...
  155. [155]
    [PDF] Guidance for Industry - FDA
    Adverse Reactions From Clinical Trials ... of reporting, (2) whether the adverse event rate for the drug exceeds the placebo rate, (3).
  156. [156]
    What is a Serious Adverse Event? - FDA
    May 18, 2023 · An adverse event is any undesirable experience associated with the use of a medical product in a patient. The event is serious and should be reported to FDA ...
  157. [157]
    [PDF] Safety Reporting Requirements for INDs and BA/BE Studies | FDA
    An adverse reaction means any adverse event caused by a drug. Adverse ... frequency for reporting suspected adverse reactions from clinical trials once a study or ...
  158. [158]
    [PDF] Definitions and Standards for Expedited Reporting E2A - ICH
    Expedited reporting is also inappropriate for serious events from clinical investigations that are considered not related to study product, whether the event is ...
  159. [159]
    Adverse Event Reporting to IRBs — Improving Human Subject ... - FDA
    May 6, 2020 · This guidance is intended to assist the research community in interpreting requirements for submitting reports of unanticipated problems, including certain ...
  160. [160]
    The role of data and safety monitoring boards in implementation trials
    The DSMB's role in conducting periodic benefit–risk assessments and their authority to recommend trial termination distinguishes them from other research ...
  161. [161]
    Data Safety Monitoring Boards Facilitate Ethical Research - Advarra
    Jun 5, 2023 · The board specifically seeks to address any unexpected change in adverse events arising during study conduct, or early indications of ...
  162. [162]
    Adverse Event Detection, Processing, and Reporting - NCBI - NIH
    The FDA maintains MedWatch, a Web-based reporting system that allows consumers and health professionals to voluntarily report serious adverse events and other ...
  163. [163]
    Institutional Review Boards (IRBs) and Protection of Human ... - FDA
    Sep 11, 2019 · An Institutional Review Board is group that has been formally designated to review and monitor biomedical research involving human subjects.
  164. [164]
    Institutional Review Boards: Purpose and Challenges - PMC
    IRBs are charged with providing an independent evaluation that proposed research is ethically acceptable, checking clinical investigators' potential biases, and ...
  165. [165]
    Institutional Review Boards Frequently Asked Questions - FDA
    The regulations do outline the criteria for IRB approval of research. 21 CFR 56.111(a)(1) requires the IRB to assure that risks to the subjects are minimized.
  166. [166]
    Institutional Review Boards: Actions Needed to Improve Federal ...
    Jan 17, 2023 · Institutional review boards (IRBs) assess the ethics and safety of research studies involving human subjects, such as behavioral studies or clinical trials.
  167. [167]
    Institutional Review Board (IRB) Written Procedures: Guidance
    The IRB Written Procedures Checklist is designed to prompt a thorough evaluation of procedures essential for ensuring the protection of human research subjects.
  168. [168]
    Regulations: Good Clinical Practice and Clinical Trials - FDA
    Here are links to FDA regulations governing human subject protection and the conduct of clinical trials.
  169. [169]
    Development & Approval Process | Drugs - FDA
    Aug 8, 2022 · Priority Review means that FDA aims to take action on an application within six months, compared to 10 months under standard review. A Priority ...How Drugs are Developed · Conducting Clinical Trials · Guidance Documents for...
  170. [170]
    Clinical Trials Regulation becomes fully applicable
    Jan 31, 2025 · The European Commission oversees the implementation of the Clinical Trials Regulation. Throughout 2025, the performance and the user experience ...
  171. [171]
    An Overview of FDA's Regulatory Review and Research Activities
    Nov 20, 2024 · FDA 101 offers a broad overview of FDA's regulatory review and research activities as well as its product quality and safety responsibilities.<|separator|>
  172. [172]
    Ethical Guidelines and the Institutional Review Board – An Introduction
    The IRB's primary responsibility is to protect research participants' rights, safety, and welfare, especially society's most vulnerable groups (i.e., prisoners, ...<|separator|>
  173. [173]
    FDA Regulatory Review - Assuring Data Quality and Validity ... - NCBI
    The Food and Drug Administration (FDA) clinical reviewer audit focuses on whether a study report and related documents are an accurate reflection of the ...Paper Auditing · Clinical Site Review and... · Sanctions · Assessment of Drugs
  174. [174]
    Mining Patterns of Adverse Events Using Aggregated Clinical Trial ...
    Adverse event reports contain the most important metrics for evaluating the hazards, harms, and risks of a clinical intervention. In this paper, we present ...
  175. [175]
    New FDA guidance lays out clinical trial sponsor safety reporting ...
    The new draft guidance offers greater flexibility for the requirement that clinical trial sponsors conduct aggregate data analyses.Ind Safety Reporting... · Aggregate Data Analysis · Unblinding Of Safety Data<|separator|>
  176. [176]
    Aggregate Safety Assessment Planning for the Drug Development ...
    Mar 23, 2021 · The ASAP evolves over a product's life-cycle and promotes interdisciplinary, systematic safety planning as well as ongoing data review and characterization.
  177. [177]
    Understanding the Aggregate Safety Assessment Plan in Pharma ...
    The proposed Aggregate Safety Assessment Plan (ASAP) has several components including identification of the safety topics of interest, how the safety data will ...A Valuable Tool For Clinical... · Why Should Company... · How Does The Asap Facilitate...
  178. [178]
    The importance of long-term follow up of participants in clinical trials
    Dec 1, 2022 · Long-term follow up is an important part of evaluating any medical condition or treatment. This is well appreciated in epidemiologic studies.
  179. [179]
    Practical and Statistical Considerations for the Long Term Follow ...
    Oct 27, 2023 · This manuscript addresses how to plan, conduct, and monitor complex multimodal data from long term follow-up gene therapy trials.
  180. [180]
    Overcoming the Challenges of Long-Term Follow-Up Studies for ...
    Oct 31, 2023 · Long-term follow-up studies for cell and gene therapy are required by regulators to ensure the safety and efficacy of these therapies.
  181. [181]
    The importance of long-term follow-up in clinical trials - The Lancet
    5-year follow-up takes more than 5 years to observe, and trials are already expensive to run. The protection against bias of randomisation tends to diminish as ...
  182. [182]
    Medable Introduces Long-Term Follow-Up Model for Cell & Gene ...
    May 28, 2025 · “Long-term follow-up is essential to ensure the durability and safety of advanced therapies, but the current system isn't working for patients ...
  183. [183]
    Gene Therapy Products, Protocols and Potential Issues
    Long-term follow‑up observations are extended assessments that continue some of the scheduled observations of a clinical trial past the active follow-up period.
  184. [184]
    Subpart D—Responsibilities of Sponsors and Investigators - eCFR
    Sponsors are responsible for selecting qualified investigators, providing them with the information they need to conduct an investigation properly.
  185. [185]
    Protecting the Rights, Safety, and Welfare of Study Subjects - FDA
    Apr 12, 2019 · This guidance provides an overview of the responsibilities of a person who conducts a clinical investigation of a drug, biological product, or medical device.
  186. [186]
    [PDF] Guidance for Industry - FDA
    This guidance discusses the general investigator responsibilities that are applicable to clinical trials of drugs, biologics, and medical devices. An ...
  187. [187]
  188. [188]
  189. [189]
    [PDF] ICH: E 6 (R2): Guideline for good clinical practice - Step 5
    Good Clinical Practice (GCP) is an international ethical and scientific quality standard for designing, conducting, recording and reporting ...
  190. [190]
    Comparison of paper-based and electronic data collection process ...
    For the sample clinical trial considered in our simulation study and our parameters estimations the EDC process decreased data collection costs for 55%. For ...
  191. [191]
    Comparing the efficiency of paper-based and electronic data ... - NIH
    Mar 8, 2021 · The perceived benefits of EDC over PDC include increased efficiency and the provision of more timely data, cost-effectiveness, accuracy, in ...
  192. [192]
    Quality Control and Assurance in Clinical Research
    The QC plan includes comparison of the study's CRF to the objectives set forth in the protocol to ensure that it is designed to collect all necessary data. A ...Missing: guidance | Show results with:guidance
  193. [193]
    Source Data Verification (SDV) quality in clinical research
    Source data verification (SDV): the comparison of study data to their original recording to ensure that, “the reported trial data are accurate, complete, and ...
  194. [194]
    [PDF] Oversight of Clinical Investigations — A Risk-Based Approach ... - FDA
    This guidance focuses principally on monitoring, which is one aspect of the processes and procedures needed to ensure clinical trial quality and subject safety.
  195. [195]
    Source Data Verification Types, Trends, and Tips
    Feb 5, 2025 · Source data verification (SDV) plays a pivotal role in safeguarding the accuracy and reliability of clinical trial data.
  196. [196]
    [PDF] Integration Quality into Clinical Trials - FDA
    Sep 28, 2022 · FDA oversees clinical trials to ensure they are designed, conducted, analyzed and reported according to federal law and good clinical practice ...
  197. [197]
    Data Validation in Clinical Data Management - Quanticate
    Jul 26, 2024 · The data validation process is a structured approach designed to verify the accuracy, completeness, and consistency of collected data.
  198. [198]
    [PDF] Guidance for Industry: Electronic Source data in Clinical Investigations
    For this reason the EHR is the source, and the pertinent data for the subjects in the clinical study should be made available for review during an FDA ...
  199. [199]
    The importance of Good Clinical Practice guidelines and its role in ...
    The ICH-GCP guidelines are therefore considered the 'bible' of clinical trials, and have become a global law which safeguards humanity as we know it today.
  200. [200]
    Electronic Data Capture in Clinical Trials
    Today nearly half of all new clinical trials are initiated using EDC. Industry analysis predicts that investments in EDC solutions will increase at a 14.7% ...
  201. [201]
    Considerations for the Use of Artificial Intelligence - FDA
    Jan 6, 2025 · This guidance provides recommendations to sponsors and other interested parties on the use of artificial intelligence (AI) to produce information or data
  202. [202]
    Bridging the Gap: From AI Success in Clinical Trials to Real-World ...
    Mar 22, 2025 · This narrative review critically examines the discrepancy between AI's robust performance in clinical trials and its inconsistent real-world implementation.
  203. [203]
    A Permissioned Blockchain-Based Clinical Trial Service Platform to ...
    Immutable clinical trial data recorded using a blockchain may inspire greater confidence in its integrity, resulting in better science, safer medicines, and ...
  204. [204]
    Blockchain as a Prime Guardian: Securing Clinical Trial Data Integrity
    Jan 23, 2025 · The present study focuses on the possible use of the emerging technology of blockchain in ensuring data management security in clinical ...
  205. [205]
    Interoperability Challenges in Medium Healthcare Organizations
    Apr 10, 2025 · Up to 26.9% of hospital data errors occur due to interoperability issues. · Healthcare organizations can lose up to $20 million annually from ...
  206. [206]
    Clinical Interoperability: Everything You Need to Know - Medidata
    Nov 25, 2024 · There are three core challenges to address: ambiguity and flexibility in data standards, challenges related to point-to-point connections or ...
  207. [207]
    How to Calculate Sample Size and Why - PMC - NIH
    ... power for a trial is widely accepted as 0.8 (80%). The higher the power (power = 1 - beta) for a trial, the larger the sample size that is required. The ...
  208. [208]
    E9 Statistical Principles for Clinical Trials September 1998 - FDA
    Aug 24, 2018 · This guidance is written primarily to attempt to harmonize the principles of statistical methodology applied to clinical trials for marketing applications.
  209. [209]
    Intention to treat and per protocol analysis in clinical trials - PubMed
    The data analysis of an RCT can be performed by using two complementary strategies, that is according to the intention to treat (ITT) principle and the per ...
  210. [210]
    Per‐Protocol Versus Intention‐to‐Treat in Clinical Trials
    May 16, 2022 · The most important result is that per‐protocol analysis confirms the results of the intention‐to‐treat. After censoring data from protocol ...
  211. [211]
    The clinician's guide to p values, confidence intervals, and ... - Nature
    Nov 26, 2021 · Confidence intervals are estimates that provide a lower and upper threshold to the estimate of the magnitude of effect. By convention, 95% ...
  212. [212]
    BEYOND STATISTICAL SIGNIFICANCE: CLINICAL ... - NIH
    It is important for clinicians to be able to interpret confidence intervals using effect sizes, minimal clinically important differences, and magnitude‐based ...
  213. [213]
    why statistical significance is not enough in clinical practice - NIH
    Feb 4, 2025 · P-values have traditionally guided clinical research, but over-reliance on them can lead to misinterpretation and poor decision-making.
  214. [214]
    The roles, challenges, and merits of the p value - ScienceDirect
    Dec 8, 2023 · First, a significant p value may not equate to clinical relevance. When employing hypothesis tests in clinical studies, a p value that is ...
  215. [215]
    E9(R1) Statistical Principles for Clinical Trials: Addendum: Estimands
    May 11, 2021 · This addendum presents a structured framework to strengthen the dialogue between disciplines involved in the formulation of clinical trial objectives.
  216. [216]
    Current use of effect size or confidence interval analyses in clinical ...
    Sep 18, 2021 · Reporting and interpretation of results from clinical trials that did not claim a treatment difference: Survey of four general medical journals.
  217. [217]
    Publication bias in clinical trials due to statistical significance or ...
    Trials with positive findings were more likely to be published than trials with negative or null findings (odds ratio 3.90; 95% confidence interval 2.68 to 5.68) ...
  218. [218]
    Ending publication bias: A values-based approach to surface null ...
    Sep 24, 2025 · Null studies are often perceived as less valuable, leading to harsher peer reviews [26–28] and lower citation rates [29,30]. Null studies are ...
  219. [219]
    Reducing publication bias with Registered Reports - Nature
    Sep 5, 2024 · This publishing format is designed to counteract positive publication bias and shift the focus from the results to the research question. A ...
  220. [220]
    Why Most Published Research Findings Are False | PLOS Medicine
    Aug 30, 2005 · Published research findings are sometimes refuted by subsequent evidence, with ensuing confusion and disappointment. Refutation and controversy ...
  221. [221]
    Reporting bias in clinical trials: Progress toward transparency and ...
    Jan 19, 2022 · Study publication bias occurs when trials showing negative or no effect are not published; outcome reporting bias occurs when authors fail to ...
  222. [222]
    Biases in randomized trials: a conversation between trialists and ...
    Biases in randomized trials: a conversation between trialists and epidemiologists · Selection bias · Performance bias · Detection bias · Attrition bias · Reporting ...
  223. [223]
    Risk of bias: why measure it, and how? | Eye - Nature
    Sep 30, 2021 · There are five main forms of bias that are important to consider for clinical trials: Selection Bias, Performance Bias, Detection Bias, ...
  224. [224]
    Reducing risk of bias in interventional studies during their design ...
    Apr 1, 2025 · While bias is difficult to quantify, risk of bias can be assessed, and studies with low risk of bias are more likely to produce results closer ...
  225. [225]
    Trends of Publication of Negative Trials Over Time - Laviolle
    Jan 5, 2025 · Our study shows that the proportion of published negative trials increased between 2000 and 2020 in high-impact medical journals and that ...Abstract · Study Highlights · Results
  226. [226]
  227. [227]
    EU Clinical Trials Register - Update
    The EU Clinical Trials Register provides a free and accurate search of clinical trials in European Union member states and the European Economic Area.Home & Search · Joining a trial · About · First
  228. [228]
    Trial registration - World Health Organization (WHO)
    The Declaration of Helsinki states that "Every clinical trial must be registered in a publicly accessible database before recruitment of the first subject".
  229. [229]
    Frequently Asked Questions | ClinicalTrials.gov
    Sep 10, 2025 · Clinical trial registration information must include information applicable to the entire trial, as is the case with all multi-site trials with ...
  230. [230]
    Clinical trials - Regulation EU No 536/2014 - Public Health
    The Regulation ensures a greater level of harmonisation of the rules for conducting clinical trials throughout the EU.Eu Clinical Trial Portal And... · Transparency · Safety Reporting
  231. [231]
    Clinical Trials Information System designated as WHO primary registry
    Apr 3, 2025 · Becoming a primary registry means CTIS adheres to specific criteria for content, data quality and validity, accessibility, unique identification ...
  232. [232]
    Publication bias: the case for an international registry of clinical trials
    In this report, a model is proposed for reviewing clinical trial results which is free from publication bias based on the selection of trials registered in ...Missing: effectiveness | Show results with:effectiveness
  233. [233]
    Discrepancies in reported results between trial registries and journal ...
    Jan 20, 2025 · There were more results in trial registries than in articles, but researchers showed a clear preference for rapid dissemination of results ...
  234. [234]
    Compliance with legal requirement to report clinical trial results on ...
    Jan 17, 2020 · The Food and Drug Administration Amendments Act (FDAAA) of 2007 now requires sponsors of applicable trials to report their results directly onto ClinicalTrials ...<|control11|><|separator|>
  235. [235]
    Clinical trial registration was associated with lower risk of bias ...
    Review. Clinical trial registration was associated with lower risk of bias compared with non-registered trials among trials included in systematic reviews.
  236. [236]
    Publication bias continues despite clinical-trial registration | Nature
    Sep 11, 2009 · Fewer than half of published trials are adequately registered, and, on the other hand, fewer than half of registered trials are ever published ...Missing: effectiveness | Show results with:effectiveness<|separator|>
  237. [237]
    CONSORT 2025 Statement: updated guideline for reporting ...
    Apr 18, 2025 · The CONSORT 2025 Statement is an updated guideline for reporting randomized trials, published in 5 journals.Abstracts · Pilot and feasibility trials · Search for reporting guidelines
  238. [238]
    CONSORT 2025 statement: updated guideline for reporting ... - Nature
    Apr 15, 2025 · The CONSORT 2025 statement consists of a 30-item checklist of essential items that should be included when reporting the results of a randomized trial.
  239. [239]
    About ICMJE | Clinical Trials Registration
    Because it is critical that trial registries are independent of for-profit interests, the ICMJE policy requires registration in a WHO primary registry rather ...
  240. [240]
    consort-spirit.org: Home
    The SPIRIT–CONSORT guidelines offer a standard way to report trial protocols and findings for randomized trials. This helps authors to report their trials ...
  241. [241]
    The Consolidated Standards of Reporting Trials (CONSORT ... - NIH
    CONSORT is a set of reporting principles for RCTs, using a 25-item checklist and flow diagram to ensure proper reporting and identify pitfalls.
  242. [242]
    The CONSORT statement - PMC - NIH
    The CONSORT statement, or 'Consolidated Standards of Reporting Trails,' is a 25-item checklist to help standardize and aid in the reporting of RCTs.
  243. [243]
    How Much Does a Clinical Trial Cost? - Sofpromed
    The average cost of phase 1, 2, and 3 clinical trials across therapeutic areas is around $4, 13, and 20 million respectively. Pivotal (phase 3) studies for new ...
  244. [244]
    Clinical Trial Costing: Phase-By-Phase Budget Guide | Abacum
    Jun 25, 2025 · Clinical trials cost between $4 million for Phase I to over $100 million for Phase III, with per-patient costs ranging from $113,000 to $136,000 ...
  245. [245]
    How to Budget for a Clinical Trial - CCRPS
    Apr 4, 2025 · Recruiting and retaining participants is one of the most expensive aspects of clinical trials, often consuming 30-40% of the total budget.Missing: categories | Show results with:categories
  246. [246]
    Phase-by-Phase Clinical Trial Costs Guide for Sponsors.
    May 5, 2025 · The clinical trial cost can range from $1 million for early-phase trials to over $100 million for large Phase 3 studies, depending on the phase, ...
  247. [247]
    Clinical cancer research in the U.S. is increasingly dominated by ...
    Sep 27, 2024 · The study showed that between 2018 and 2022, industry sponsored trials enrolled over eight times more patients than federally sponsored trials.
  248. [248]
    Costs of Drug Development and Research and ... - JAMA Network
    Jun 28, 2024 · In this study, the mean cost of the clinical phase per drug candidate was estimated at $117.4 million, lower than the majority of the reported ...
  249. [249]
    Spending on Phased Clinical Development of Approved Drugs ... - NIH
    Jul 14, 2023 · Spending by NIH on phased development represented 9.8% to 10.7% of estimated industry spending, including 24.6% to 25.3% of estimated phase 1, ...
  250. [250]
  251. [251]
    Costs of Drug Development and Research and Development ...
    Jun 28, 2024 · Studies have estimated that the R&D cost for a new drug ranges from $314 million to $4.46 billion, depending on the therapeutic area, data, and ...
  252. [252]
    How Much Does a Day of Delay in a Clinical Trial Really Cost?
    Jun 6, 2024 · The results indicate that, across all therapeutic areas, the mean direct cost to conduct a clinical trial per day is approximately $40,000—about ...
  253. [253]
    Examination of Clinical Trial Costs and Barriers for Drug Development
    Jul 24, 2014 · Priority Review/Priority Review vouchers; Improvements in FDA review process efficiency and more frequent and timely interactions with FDA.
  254. [254]
    Clinical Trials – Supplementing Your Income and “Doing Good”
    Oct 18, 2023 · Certain testing organizations have been reimbursed $2,000 per patient ... In addition to financial benefits, participation in clinical trials can ...
  255. [255]
    [PDF] Investigator Compensation: Understanding the Basics, Avoiding the ...
    Investigator compensation includes clinical services, professional fees for procedures, and time/effort tasks like study meetings and safety reports.
  256. [256]
    Financial Disclosures by Clinical Investigators - FDA
    Payments that are greater for one outcome than another or that are in the form of stock options or royalties are examples of such payment arrangements and ...
  257. [257]
    How Are PIs Paid All The Options For Clinical Research Investigator ...
    May 16, 2024 · For study sponsors, not adhering to FMV can result in allegations of improper financial relationships or conflicts of interest, which can damage ...
  258. [258]
    The Financing of Drug Trials by Pharmaceutical Companies and Its ...
    A number of studies in recent years have shown that clinical drug trials financed by pharmaceutical companies yield favorable results for the company's product ...Missing: costs revenues
  259. [259]
    Conflict of interest and its importance - PMC - NIH
    Compensation affected by the outcome of clinical studies means compensation that could be higher for a favorable outcome than for an unfavorable outcome, such ...
  260. [260]
    Conflict-of-Interest Policies for Investigators in Clinical Trials
    Nov 30, 2000 · Federal regulations require investigators applying for funds from the U.S. Public Health Service to disclose “significant” financial interests ...
  261. [261]
    Payment and Reimbursement to Research Subjects - FDA
    Jul 12, 2018 · Payment for participation in research should be just and fair. The amount and schedule of all payments should be presented to the IRB at the ...
  262. [262]
    Determining Optimal Compensation for Clinical Trial Participants
    Apr 12, 2021 · In practice, up to about 25% of the amount that the participant would be paid can generally be held over as a bonus. But anything more than 25% ...<|control11|><|separator|>
  263. [263]
    How much do healthy volunteers actually earn from clinical trial ...
    May 2, 2021 · Overall, the median clinical trial compensation was US$3070 (range = US$150-US$13,000). Participants seeking new healthy volunteer trials tended ...
  264. [264]
    How Much do Clinical Trials Pay? Everything You Need to Know ...
    Dec 20, 2023 · On the higher end, some clinical trials may offer payment in the tens of thousands of dollars for participation, particularly if the trial ...
  265. [265]
    Walking the Fine Line Between Fairness and Inducement ... - ACRP
    Apr 10, 2025 · The dilemma is whether to offer upfront stipends or reward after study completion. Concerns exist about inducement and how much is too much, ...
  266. [266]
    [PDF] Ethical Considerations in Decentralized Clinical Trials | MRCT Center
    Mar 1, 2024 · The burden of travel and trial out- of-pocket expenses have shown a differential impact on the participation of lower-income compared with.
  267. [267]
    Breaking Financial Barriers: Making the Shift to Reasonable ...
    Participant payments typically fall into three categories: reimbursement for study-related expenses, compensation for time and effort, and incentive payments.<|separator|>
  268. [268]
    MONEY, COERCION, AND UNDUE INDUCEMENT: A SURVEY OF ...
    Oct 30, 2014 · ... undue inducement may unnecessarily interfere with the recruitment of research participants and impede valuable research. Policy guidance and ...
  269. [269]
    Medical research participants deserve to be paid well - STAT News
    Jul 17, 2025 · Thus, the collapse of lower payments as an undue inducement: Either we stop paying anything above, perhaps a few hundred dollars, because that ...
  270. [270]
    When Does the Amount We Pay Research Participants Become ...
    According to Ruth Macklin, an inducement is undue if it encourages participants to lie or conceal information in order to participate or prompts participants ...
  271. [271]
    Effectiveness of Recruitment in Clinical Trials: An Analysis of ... - NIH
    This paper reports on the effectiveness of standard recruitment methods such as physician referral, newspaper advertisements, fliers, audio and video media
  272. [272]
    Successes and Challenges in Clinical Trial Recruitment - NIH
    Aug 14, 2024 · Strategies include increasing potential participants' awareness of the health problem being studied [7,8], using advertising and social media ...
  273. [273]
    Improving Participant Recruitment in Clinical Trials
    Dec 18, 2024 · This study aims to investigate the use of novel digital platforms and their features, such as e-recruitment, e-consent, and matching, aiming to optimize and ...
  274. [274]
    Finding trials for participants: an ethnographic study of successful ...
    Aug 8, 2025 · Specific methods for making trials “patient-centric” include involving patient and public groups in drafting consent forms, using technology to ...
  275. [275]
    Why Diverse Representation in Clinical Research Matters ... - NCBI
    Ensuring diverse participation in scientific studies allow for exploration of all of these factors and their interactions and is critical to the interpretations ...
  276. [276]
    Why Diverse Clinical Trial Participation Matters
    Apr 1, 2023 · Gains in biomedical knowledge associated with increased trial diversity generally depend on the existence of heterogeneity in treatment effects ...
  277. [277]
    Minority Health Month: Why Diversity is Necessary in Clinical Trials
    Apr 18, 2023 · According to the Food & Drug Administration (FDA), in 2020 75% of trial participants were white, 11% were Hispanic, 8% were Black, and 6% were ...<|separator|>
  278. [278]
    Plans and Enrollment Often Fell Short for Underrepresented Groups ...
    May 28, 2024 · Two-thirds of the clinical trials in our sample had inclusive enrollment plans, but one-third did not plan to include all racial and ethnic ...
  279. [279]
    Barriers to Representation of Underrepresented and Excluded ...
    Individual and community factors are often cited as reasons for lack of inclusion of underrepresented and excluded populations in clinical trials.
  280. [280]
    Race/ethnicity reporting and representation in US clinical trials
    Apr 10, 2022 · The majority of US trials in ClinicalTrials.gov do not report race/ethnicity enrollment data, and minorities are underrepresented in trials with modest ...
  281. [281]
    NIH-developed AI algorithm matches potential volunteers to clinical ...
    Nov 18, 2024 · The AI algorithm, called TrialGPT, could successfully identify relevant clinical trials for which a person is eligible and provide a summary that clearly ...
  282. [282]
    Enhancing Patient-Trial Matching With Large Language Models - NIH
    Jun 9, 2025 · Patient recruitment remains a major bottleneck in clinical trial execution, with inefficient patient-trial matching often causing delays and ...
  283. [283]
    Patients' selection and trial matching in early-phase oncology ...
    Trial matching could increase patients' recruitment to trials by up to 20%. Eligibility prediction could bring early discontinuation to less than 20%. Those ...<|separator|>
  284. [284]
    [PDF] Clinical Trial Participation Among US Adults
    In 2020, 41% of Americans reported not knowing anything about clinical trials. • Reducing barriers to trial participation would ensure more generalizable and ...
  285. [285]
    Low Income Is a Barrier to Clinical Trial Enrollment, Study Suggests
    Nov 16, 2015 · A new study has found that patients with annual household incomes below $50000 were less likely to participate in a cancer clinical trial ...Missing: statistics | Show results with:statistics
  286. [286]
    [PDF] ISSUE BRIEF - https: // aspe . hhs . gov.
    Jul 1, 2025 · Barriers such as patient and provider awareness, proximity and access to trial sites, trial exclusion criteria, and financial stress can all ...
  287. [287]
    Promoting Factors and Barriers to Participation in Early Phase ... - NIH
    Practical barriers, such as lack of transportation, lack of health insurance, financial constraints, lack of family support, trial duration, and high frequency ...
  288. [288]
    Barriers for cancer clinical trial enrollment: A qualitative study of the ...
    These factors include distance to clinical trial location, financial aspects, limited beliefs/trust in the clinical research, low diversity of staff, and ...Missing: statistics | Show results with:statistics
  289. [289]
    Four Trends Shaping Clinical Trials in 2024 - ACRP
    Jan 3, 2024 · Eighty-five percent of trials fail to recruit enough patients, while 80% are delayed due to other factors like participant dropouts.
  290. [290]
    Unique Considerations for Patient Retention in Decentralized ...
    Feb 22, 2023 · According to Forte Research, the average dropout rate across all clinical trials is 30% and the average cost to recruit just one participant is ...
  291. [291]
    Differential dropout and bias in randomised controlled trials - NIH
    Dropout in randomised controlled trials is common and threatens the validity of results, as completers may differ from people who drop out.
  292. [292]
    Dropout Prevalence and Associated Factors in Randomized Clinical ...
    The adverse effects most associated with dropout were attempted suicide followed by mania, skin rash, and headache. Problems relating to clinical trials and ...Dropout Prevalence And... · Abstract · Implications<|separator|>
  293. [293]
    Strategies for participant retention in long term clinical trials - NIH
    [4] Other retention challenges include inconvenient location, schedule conflicts, perception that the disease condition is not improving, lack of appreciation ...Study Participants · Involving Families · Retention ToolsMissing: 2020-2025 | Show results with:2020-2025
  294. [294]
    Exploring non-retention in clinical trials - BMJ Open
    Being too well to engage further with trial processes · Lack of compatibility with personal sense of self · Being 'too ill' to be able to engage appropriately ...Results · Discussion · Principal Findings
  295. [295]
    Optimizing Recruitment and Retention in Cancer Clinical Trials in ...
    Feb 13, 2025 · We found skill-related gaps, poor attitude of clinical staff, lack of incentives, and high clinical workload as provider-related barriers, ...
  296. [296]
    Health and study dropout: health aspects differentially predict attrition
    Jan 30, 2022 · Participation attrition/Survey Dropout may lead to biased inferences and thus false conclusions thereby threatening the validity of longitudinal ...
  297. [297]
    [PDF] Conducting Clinical Trials With Decentralized Elements - FDA
    In this guidance, a decentralized clinical trial. (DCT) refers to a clinical trial that includes decentralized elements where trial-related activities occur ...
  298. [298]
    Conducting Clinical Trials With Decentralized Elements - FDA
    Oct 16, 2025 · In this guidance, a decentralized clinical trial refers to a clinical trial that includes decentralized elements where trial-related ...Missing: history | Show results with:history
  299. [299]
    Digitally enabled decentralised research: opportunities to improve ...
    The US Food and Drugs Administration (FDA) defines decentralised clinical trials as trials executed through telemedicine and mobile or local healthcare ...<|separator|>
  300. [300]
    Everything You Wanted To Know About Decentralized Clinical Trials
    Jun 22, 2022 · The first decentralized clinical trial was conducted by Pfizer in 2011, but its foundations had been laid in 2007 itself. It enrolled 600 ...Missing: definition guidance
  301. [301]
    Decentralized Clinical Trials: FDA Guidance Analyzed - Medidata
    Jan 13, 2025 · A DCT is a clinical trial with activities at non-traditional sites, including participant locations, local facilities, and remote data ...
  302. [302]
    A look ahead decentralized clinical trials in 2023 - ObvioHealth
    2022 marked the rapidly growing adoption of DCTs, with 40 to 45% of studies incorporating decentralized methods, up from 20 to 25% of studies in 2021, ...Missing: statistics | Show results with:statistics
  303. [303]
  304. [304]
    Decentralized Clinical Trials (DCTs) Market Size & Forecast [2033]
    Sep 29, 2025 · Decentralized trials reported 19% drop-out rates, versus 28% in traditional formats.Missing: statistics | Show results with:statistics
  305. [305]
    The Digital Platform and Its Emerging Role in Decentralized Clinical ...
    Sep 3, 2024 · Digital platforms allow participants to be monitored remotely, which is associated with improved self-management of conditions and better ...
  306. [306]
    Opportunities and Challenges for Decentralized Clinical Trials
    Key challenges recognized by regulators for DCTs include concerns regarding investigator oversight and participants' safety when physical examinations and face ...
  307. [307]
    Using digital technologies in clinical trials: current and future ... - NIH
    Some of the main challenges include ethical and privacy issues as well as issues related to data quality. A list of the advantages and challenges are summarized ...
  308. [308]
    Decentralized trial data quality strong but adoption still slow - Healio
    Jul 29, 2025 · Use of decentralized elements in cancer clinical trials did not have long-term negative effects on data quality. Regulatory and ...
  309. [309]
    A Scoping Review of Artificial Intelligence Applications in Clinical ...
    Jan 22, 2025 · This scoping review analyzes 142 studies published between 2013 and 2024, focusing on safety (n=55), efficacy (n=46), and operational (n=45) risk prediction.
  310. [310]
    Artificial intelligence for optimizing recruitment and retention in ...
    Sep 11, 2024 · Applying AI to recruitment in clinical trials has demonstrated several positive outcomes, such as increasing efficiency, cost savings, improving recruitment, ...<|separator|>
  311. [311]
    Artificial intelligence and clinical trials: a framework for effective ...
    Jul 23, 2025 · Artificial intelligence (AI) could offer an unprecedented opportunity to streamline the clinical trial lifecycle—from design to reporting—and ...
  312. [312]
    Case Studies and Real-World Applications of Wearables in Clinical ...
    Jun 20, 2025 · 1. Cardiovascular Trials Using Smartwatches · 3. Remote Monitoring in Oncology Trials · 4. Neurological Trials with Motion Sensors · 5. Sleep and ...
  313. [313]
    How Can Wearable Devices Help in Clinical Trials? - Thryve
    Examples of wearable use in clinical studies ... Roche has found considerable utility in the use of wearables for research on Parkinson's disease. “[…] We found ...<|separator|>
  314. [314]
    Recent Advances in the Wearable Devices for Monitoring and ...
    Commercial ECG patches used in clinical trials and in the monitoring of heart rate in HF management. (a) VitalPatch wearable sensor used in the continuous ...
  315. [315]
    New clinical trial design in precision medicine - Nature
    Mar 4, 2024 · Umbrella trials provide a highly efficient method to evaluate multiple interventions simultaneously in a single trial.
  316. [316]
    Clinical trial design in the era of precision medicine
    Aug 31, 2022 · Overall, clinical trials have evolved, shifting from tumor type-centered to gene-directed and histology-agnostic trials, with innovative ...
  317. [317]
    Advancing Precision Medicine: A Review of Innovative In Silico ...
    This review explores the interplay of these multiple in silico approaches in advancing precision medicine and fostering individual healthcare. Despite intrinsic ...2. Omics In Advancing... · 3. Biomarkers And Molecular... · 6.1. Examples Of Ml...<|control11|><|separator|>
  318. [318]
    Precision Medicine Clinical Trials: Defining New Treatment Strategies
    Precision medicine is an approach to deliver optimal patient outcomes by integrating clinical and molecular patient data to understand the biological basis of ...
  319. [319]
    Real-World Evidence - FDA
    Sep 22, 2025 · Real-world evidence is the clinical evidence about the usage and potential benefits or risks of a medical product derived from analysis of RWD.Center for Biologics Evaluation ...Guidance document
  320. [320]
    [PDF] Framework for FDA's Real-World Evidence Program
    FDA's RWE Program will cover clinical trials that generate RWE in some capacity (i.e., sources other than traditional clinical trials) and observational studies ...
  321. [321]
    Real-world Evidence versus Randomized Controlled Trial - NIH
    Jun 26, 2018 · Table 2. Advantages and limitations of real-world evidence compared with RCTs. ; Detection of less frequent side effects ; Rapid access/easier ...
  322. [322]
    Rationale, Strengths, and Limitations of Real-World Evidence in ...
    Jun 28, 2022 · Limitation of RCTs to Define the Safety of an Intervention ... RCTs are time-limited and often not designed or powered to assess safety outcomes.
  323. [323]
    Adaptive design clinical trials: Methodology, challenges and prospect
    Types of Adaptive Design Trials. Based on adaptations employed, commonly considered adaptive design methods in clinical trials include an adaptive randomization ...
  324. [324]
    Adaptive Designs for Clinical Trials | New England Journal of Medicine
    Jul 7, 2016 · Adaptive trial design has been proposed as a means to increase the efficiency of randomized clinical trials, potentially benefiting trial participants and ...
  325. [325]
    Adaptive designs in clinical trials: a systematic review-part I
    Oct 4, 2024 · Adaptive designs (ADs) are intended to make clinical trials more flexible, offering efficiency and potentially cost-saving benefits.<|control11|><|separator|>
  326. [326]
    Trial designs using real‐world data - PubMed Central - NIH
    This review explores how hybrid study designs that include features of RCTs and studies with real‐world data (RWD) can combine the advantages of both.
  327. [327]
    The Evidence REVEAL Study: Exploring the Use of Real‐World ...
    Nov 20, 2020 · Our study explored how two emerging approaches, the use of real-world evidence (RWE) and complex clinical trial (CCT) design, are currently being used by the ...Real World Evidence · Results · Use Of Rwe By Pharmaceutical...<|separator|>
  328. [328]
    Adaptive design clinical trials: current status by disease and ... - NIH
    An adaptive design is a clinical trial design that allows for modification of a structured plan in a clinical trial based on data accumulated during ...
  329. [329]
    Beyond Nazi War Crimes Experiments: The Voluntary Consent ... - NIH
    The Nuremberg Code consists of 10 principles that constitute basic legal and ethical rules for research with human subjects. The first principle, the consent ...
  330. [330]
    The Nuremberg Code | Holocaust Encyclopedia
    Medical professionals were put on trial after the war for their participation in war crimes and crimes against humanity during the Holocaust.Missing: clinical | Show results with:clinical
  331. [331]
    The US Sexually Transmitted Disease Experiments in Guatemala
    More than 5000 uninformed and unconsenting Guatemalan people were intentionally infected with bacteria that cause sexually transmitted diseases.
  332. [332]
    U.S. Officials Apologize for 'Appalling' 1940s Syphilis Study - Science
    A US-funded scientist ran an experiment that infected his Guatemalan patients with syphilis, the US government today issued a formal apology to the Central ...
  333. [333]
    The Untreated Syphilis Study at Tuskegee Timeline - CDC
    Sep 4, 2024 · The study initially involved 600 Black men – 399 with syphilis, 201 who did not have the disease. Participants' informed consent was not collected.
  334. [334]
    Tuskegee Experiment: The Infamous Syphilis Study - History.com
    May 16, 2017 · The Tuskegee experiment began in 1932, at a time when there was no known cure for syphilis, a contagious venereal disease.
  335. [335]
    The Discovery of Hepatitis Viruses: Agents and Disease - PMC - NIH
    Human experiments at Willowbrook identified two forms of hepatitis namely infectious hepatitis and serum hepatitis. The discovery of Australia antigen in ...
  336. [336]
    Hepatitis Studies at the Willowbrook State School for Children
    Dec 8, 2020 · From 1956 through 1971, residents at the Willowbrook State School for Children with Mental Retardation were infected with live hepatitis in order to develop a ...
  337. [337]
    How the thalidomide scandal led to safer drugs - MedicalNewsToday
    Dec 15, 2020 · The thalidomide tragedy demonstrated the need for strictly enforced regulation of drug testing, marketing, dispensing, and use. Clinical Trials ...
  338. [338]
    Clinical Trials in Pregnancy and the “Shadows of Thalidomide” - NIH
    Thalidomide “created an aversion” to involving pregnant women or women of reproductive age in research on drugs [28]. In addition, the effect of ...
  339. [339]
    Medicine is plagued by untrustworthy clinical trials. How ... - Nature
    Jul 18, 2023 · Investigations suggest that, in some fields, at least one-quarter of clinical trials might be problematic or even entirely made up, warn some researchers.
  340. [340]
    Pharmaceutical industry sponsorship and research outcome ... - NIH
    Studies sponsored by pharmaceutical companies were more likely to have outcomes favouring the sponsor than were studies with other sponsors.
  341. [341]
    Industry sponsorship and research outcome | Cochrane
    Feb 16, 2017 · Sponsorship of drug and device studies by the manufacturing company leads to more favorable efficacy results and conclusions than sponsorship by other sources.Missing: influence | Show results with:influence
  342. [342]
    Drug Company Used Ghostwriters to Write Work Bylined by ...
    Dec 2, 2010 · Newly released documents show how medical ghostwriters--paid for by a UK drug company--penned material published in medical journals and even a textbook.
  343. [343]
    Exposing Drug Company-Hired Ghostwriters in Medical Journals
    Several drug companies, such as Merck and Pfizer, were caught using ghostwriters to promote their drugs and hide their negative side effects.
  344. [344]
    Merck Manipulated the Science about the Drug Vioxx
    Oct 12, 2017 · Scientists from the pharmaceutical giant Merck skewed the results of clinical trials in favor of the arthritis drug, Vioxx, to hide evidence that the drug ...<|separator|>
  345. [345]
    Novartis's Zolgensma: exploring the problem of manipulated data
    Mar 23, 2022 · The FDA accused Novartis of submitting a drug application with manipulated data and then not informing regulators of the issue until a month after approval.
  346. [346]
    Data fraud in clinical trials - PMC - PubMed Central
    Highly publicized cases of fabrication or falsification of data in clinical trials have occurred in recent years and it is likely that there are additional ...
  347. [347]
    Shifts in the clinical trial landscape - Nature
    Mar 8, 2024 · The report also pegs the composite drug development success rate across all therapy areas at 10.8% in 2023, up from a low-point of 5.9% in 2022.
  348. [348]
    Benchmarking R&D success rates of leading pharmaceutical ...
    ... clinical development phases was found to be 10.4%. The success rates per phase were as follows: Phase I to Phase II at 64%; Phase II to Phase III at 32%; Phase ...
  349. [349]
    90% of drugs fail clinical trials
    Mar 12, 2022 · Lastly, 10% of failures were attributed to lack of commercial interest and poor strategic planning. This high failure rate raises the question ...
  350. [350]
    The High Failure Rate of Clinical Trials in the U.S - LinkedIn
    Oct 19, 2024 · Approximately 70-80% of drugs fail during this phase. Lack of efficacy is the most common cause of failure at this stage, as many drugs that ...<|separator|>
  351. [351]
    Why 90% of clinical drug development fails and how to improve it?
    Analyses of clinical trial data from 2010 to 2017 show four possible reasons attributed to the 90% clinical failures of drug development: lack of clinical ...
  352. [352]
    Factors associated with clinical trials that fail and opportunities ... - NIH
    Aug 7, 2018 · Clinical trials also fail with respect to safety. Hwang et al. [58] found that 17% of the failed phase 3 trials examined were due to safety.
  353. [353]
    The Cost of Drug Development: How Much Does It Take to Bring a ...
    Sep 28, 2025 · The average cost to develop a new drug is about $2.6 billion, and the process takes 10 to 15 years from discovery to market approval.
  354. [354]
    Clinical development times for innovative drugs - PMC - NIH
    Controlling for other development factors, the clinical development time of a typical innovative drug is 9.1 years (95% confidence interval (CI) = 8.2–10.0 ...
  355. [355]
    Research and Development in the Pharmaceutical Industry
    Apr 8, 2021 · In recent studies, estimates of the average R&D cost per new drug range from less than $1 billion to more than $2 billion per drug.
  356. [356]
    Overcoming inefficiencies of clinical trials with technology
    Feb 27, 2020 · Research by ZS found that approximately 80% of clinical trials fail to meet enrolment deadlines, while 30% of Phase III studies last longer than ...
  357. [357]
    A crisis in clinical research - PMC - PubMed Central - NIH
    Jul 10, 2024 · Unfortunately, progress has slowed since the COVID-19 pandemic due to uncovered, systemic inefficiencies across critical processes.
  358. [358]
    The unsustainability of clinical trials and need for change
    Jul 11, 2025 · Mounting costs, workforce shortages, and poor enrollment rates are having a significant impact on clinical trials. Industry leaders say it's time for reform.
  359. [359]
    Drivers of Start-Up Delays in Global Randomized Clinical Trials - PMC
    Sep 21, 2020 · We identified key drivers for start-up delays in the following categories: regulatory, contracts and budgets, insurance, clinical supplies, site identification ...
  360. [360]
    FDA vs. EMA: Navigating Divergent Regulatory Expectations for Cell ...
    Apr 3, 2025 · This article explores key distinctions between the FDA and EMA in CGT regulation and provides strategies for biopharma companies to optimize their regulatory ...
  361. [361]
    The True Cost of Delay in Clinical Trials: Why Innovation is Needed
    Dec 11, 2024 · According to a May 2024 report in Therapeutic Innovation and Regulatory Science, each day of delay in drug development costs approximately $500 ...
  362. [362]
    Navigating Regulatory Hurdles in Drug Development - DrugBank Blog
    Jun 18, 2025 · The failure rate in clinical trials exceeds 90%, often due to insufficient safety data, efficacy concerns, or regulatory non-compliance. Even ...
  363. [363]
    Reducing Bureaucracy in Clinical Research: A Call for Action - PMC
    This article discusses the issues resulting from the exponential growth of regulatory and administrative requirements for the conduct of clinical studies.
  364. [364]
    Clinical Trials: Assessing Safety and Efficacy for a Diverse Population
    This educational workshop aims to describe the significance of population diversity in evaluating medical products and to advance discussion on methodological ...
  365. [365]
    Enhancing Postmarket Safety Monitoring - Challenges for the FDA
    While this passive surveillance system may be capable of detecting rare serious adverse events, it has several limitations, including profound underreporting, ...
  366. [366]
    [PDF] FDA Has Begun Building an Active Postmarket Surveillance System
    Jul 24, 2024 · Active postmarket surveillance involves the ongoing review of evidence—derived from the analysis of data sources such as electronic health ...
  367. [367]
    Factors associated with clinical trials that fail and opportunities for ...
    Failures can arise from a lack of efficacy, issues with safety, or a lack of funding to complete a trial, as well as other factors such as failing to maintain ...
  368. [368]
    Streptomycin Treatment of Pulmonary Tuberculosis - The BMJ
    Streptomycin treatment of pulmonary tuberculosis. Br Med J 1948; 2 doi: https://doi.org/10.1136/bmj.2.4582.769 (Published 30 October 1948)
  369. [369]
    1955 Polio Vaccine Trial Announcement
    The Salk vaccine has been proved to be up to 80-90 percent effective in preventing paralytic polio.
  370. [370]
    People and Discoveries: Salk produces polio vaccine - PBS
    A nationwide testing of the vaccine was launched in April 1954 with the mass inoculation of school children. The results were amazing -- 60-70 percent ...
  371. [371]
    Long-Term Data from 20 Trials Confirm Tamoxifen's Long-Lasting ...
    Nov 23, 2011 · The decrease in recurrence risk for ER-positive breast cancer after tamoxifen treatment was accompanied by reductions in breast cancer deaths ...Missing: success | Show results with:success
  372. [372]
    Treatment of Tuberculosis. A Historical Perspective | Annals of the ...
    Sep 23, 2015 · (A) As shown in the table, streptomycin alone decreased deaths and brought about radiographic improvement, but many patients remained culture ...
  373. [373]
    History of polio vaccination - World Health Organization (WHO)
    The first successful vaccine was created by US physician Jonas Salk. Salk tested his experimental killed-virus vaccine on himself and his family in 1953.
  374. [374]
    [PDF] How many life-years have new drugs saved? A three-way fixed ...
    Mar 27, 2024 · The models control for the average decline in the YLL rate in each country and from each disease. Results: One additional drug launch 0–11 y ...
  375. [375]
    Causal Inference in Public Health - PMC - PubMed Central
    Causal inference has a central role in public health; the determination that an association is causal indicates the possibility for intervention.
  376. [376]
    Evidence for Health Decision Making — Beyond Randomized ...
    Aug 3, 2017 · Randomized, controlled trials are key in advancing medical understanding, but some questions are not amenable to this approach.Missing: rationale | Show results with:rationale
  377. [377]
    Why the 'gold standard' of medical research is no longer enough
    Aug 2, 2017 · Despite their strengths, RCTs have substantial limitations. They can be very expensive to run. They can take many years to complete, and even ...
  378. [378]
    Strengths and Limitations of RCTs - NCBI - NIH
    RCTs bear some important limitations. First, RCTs may be underpowered to detect differences between comparators in harms.
  379. [379]
    Randomized controlled trials – The what, when, how and why
    Randomized controlled trials (RCTs) are at the top of the pyramid of evidence as they offer the best answer on the efficacy of a new treatment.
  380. [380]
    Limitations of Randomized Clinical Trials - PubMed
    Aug 15, 2020 · The types of limitations discussed in this review may be grouped in 11 categories including incorrect statistical inference, low internal or external validity.
  381. [381]
    Rethinking the pros and cons of randomized controlled trials and ...
    Jan 18, 2024 · The purpose of this manuscript, aims to raise awareness of the pros and cons of RCTs and observational studies in order to help guide clinicians, researchers, ...
  382. [382]
    Angus Deaton's Critique of Randomized Controlled Trials - SIOE
    Oct 16, 2015 · When RCTs are not possible, this movement advocates quasi-randomization through instrumental variable (IV) techniques or natural experiments. I ...
  383. [383]
    The pitfalls of randomized controlled trials
    Sep 1, 2010 · Randomized clinical trials have their place, but critics argue that researchers would get better results if they also embraced other methodologies.
  384. [384]
    Alternatives to placebo-controlled trials - PubMed
    In equivalence trials, the aim is to show that the new and standard agents have similar degrees of effectiveness or adverse events.
  385. [385]
    Expanded Access - FDA
    Sep 8, 2025 · Expanded access is a potential pathway for a patient with a serious or immediately life-threatening disease or condition to gain access to an investigational ...
  386. [386]
    Understanding and misunderstanding randomized controlled trials
    RCTs can play a role in building scientific knowledge and useful predictions but they can only do so as part of a cumulative program.