Fact-checked by Grok 2 weeks ago

Inclusion and exclusion criteria

Inclusion and exclusion criteria are predefined eligibility standards employed in clinical trials and other scientific research to select participants, wherein inclusion criteria outline the essential demographic, clinical, or other characteristics that prospective subjects must exhibit to qualify, and exclusion criteria identify factors—such as comorbidities, prior treatments, or demographic mismatches—that render individuals ineligible, thereby precisely delineating the study's target population. These criteria serve to homogenize the study cohort, thereby reducing variables that could obscure causal relationships between interventions and outcomes, enhancing the of results while also mitigating risks to participants by excluding those for whom the study might pose undue harm or yield uninterpretable data. In clinical trials regulated by bodies like the U.S. (FDA), they are integral to protocol design, ensuring alignment with research objectives, ethical imperatives under frameworks such as the Declaration of Helsinki, and requirements for regulatory approval, as they facilitate focused testing and monitoring. Key considerations in formulating these criteria include balancing specificity to isolate effects against broader applicability to real-world populations; overly stringent exclusions can limit and , prompting FDA guidance to relax non-essential restrictions—such as arbitrary age caps or organ function thresholds unrelated to safety—particularly in and rare diseases, to bolster diversity and accelerate evidence generation without compromising rigor. Conversely, insufficient stringency risks diluting signal-to-noise ratios, as evidenced in peer-reviewed analyses of designs where lax criteria correlate with heterogeneous responses and interpretive challenges. Notable controversies arise from their application in underrepresented groups, where empirical data indicate that unmerited exclusions—often justified by historical safety concerns rather than prospective evidence—perpetuate gaps in generalizability, as seen in cardiovascular and trials excluding those with disabilities or comorbidities prevalent in broader demographics. Recent regulatory shifts emphasize evidence-based justification for exclusions to prioritize causal realism over precautionary overreach, fostering trials that better inform population-level efficacy.

Definitions and Core Concepts

Inclusion Criteria

Inclusion criteria in scientific research, particularly clinical trials, consist of predefined characteristics that prospective participants must possess to be eligible for , thereby delineating the target population relevant to the study's objectives. These criteria ensure that the selected individuals align with the key features necessary to address the , such as specific demographic attributes, clinical diagnoses, or physiological conditions. For instance, in trials evaluating treatments for advanced-stage cancers, inclusion often requires confirmation of the disease stage via or , alongside minimum scores like those from the Eastern Cooperative Oncology Group . The design of inclusion criteria prioritizes participant safety, study feasibility, and the generation of interpretable data by homogenizing the sample to minimize variability unrelated to the . Researchers establish these criteria prior to initiation, drawing from pathophysiological, epidemiological, and logistical considerations to form an ideal that maximizes the potential for detecting effects. Common elements include age ranges (e.g., adults aged 18-75 years), confirmed diagnoses through standardized diagnostic tools, and absence of comorbidities that could skew outcomes, as seen in cardiovascular trials requiring stable readings above 140/90 mmHg. In non-clinical research, such as systematic reviews, inclusion criteria specify mandatory study attributes like publication date (e.g., post-2010) or methodological rigor (e.g., randomized controlled designs). By focusing on homogeneity, inclusion criteria enhance , allowing causal inferences about the intervention's within the defined , though overly restrictive designs may limit generalizability. Regulatory bodies like the U.S. emphasize that these criteria must justify exclusions based on scientific rationale rather than arbitrary preferences, with required in protocols to facilitate ethical . Empirical analyses of datasets indicate that optimizing thresholds, such as symptom severity cutoffs, can reduce costs by up to 20-30% while preserving statistical power.

Exclusion Criteria

Exclusion criteria consist of predefined characteristics or conditions that disqualify potential participants from a study, typically after they have met initial requirements. These criteria identify individuals whose presence could introduce safety risks, variables, or excessive heterogeneity that undermines the study's ability to isolate causal effects of the under investigation. A primary function of exclusion criteria is to safeguard participant welfare by barring those at elevated risk of harm from the study's procedures or treatments. For instance, individuals with severe renal impairment or uncontrolled may be excluded from trials involving nephrotoxic drugs to avert adverse events that could skew safety profiles or lead to disproportionate dropout rates. This approach aligns with ethical mandates under frameworks like the Declaration of Helsinki, prioritizing non-maleficence while enabling focused assessment of efficacy in lower-risk cohorts. Exclusion criteria also promote by homogenizing the study population and curtailing extraneous variability. By omitting factors such as concurrent use of interfering medications or histories of non-compliance, researchers diminish noise in outcome measures, facilitating clearer attribution of effects to variable. Empirical analyses of trial datasets indicate that tighter exclusions correlate with narrower confidence intervals around treatment effects, though this comes at the cost of potentially diminished when results fail to generalize beyond the curated sample. In practice, exclusion criteria are tailored to the research question's causal structure; for example, in trials, prior exposure to the study drug class or active infections might be barred to prevent floor/ceiling effects on response rates. Regulatory bodies like the FDA emphasize evaluating these criteria's stringency to avoid overly restrictive designs that exclude subgroups—such as the elderly or those with comorbidities—prevalent in real-world applications, which a 2018 review found impacts up to 80% of potential patients in some therapeutic areas. Common implementations include age cutoffs (e.g., excluding those over 75 years to control for physiological declines), pregnancy status, or histories, with criteria explicitly documented in protocols to ensure and .

Historical Development

Origins in Early Clinical Research

In the initial phases of controlled clinical experimentation, patient selection was often implicit and based on practical availability rather than formalized criteria, as seen in James Lind's 1747 trial on treatments among British sailors, where participants were chosen solely for exhibiting similar symptoms of the disease to facilitate comparison of interventions. Such approaches prioritized homogeneity in disease presentation to isolate treatment effects but lacked explicit rules for inclusion or exclusion, relying instead on the researcher's judgment to minimize variables like age or comorbidities. The transition to explicit inclusion and exclusion criteria emerged with the development of randomized controlled trials (RCTs) in the mid-20th century, driven by the need to standardize participant groups for unbiased comparison and to enhance amid growing ethical and scientific scrutiny. A landmark example is the 1948 Medical Research Council (MRC) trial of for pulmonary , led by , which specified inclusion criteria such as patients aged 15 to 30 years with acute, progressive, bilateral pulmonary of recent origin, bacteriologically confirmed, and exclusion of those with chronic or stabilized disease to ensure a uniform population responsive to the . This trial's criteria, applied after a one-week observation period to confirm eligibility, marked an early systematic effort to define eligibility prospectively, reducing and enabling between treatment and control arms. These early criteria originated from first-hand recognition that heterogeneous populations could obscure causal inferences, as heterogeneous patient characteristics introduced uncontrolled variables that confounded outcomes in non-randomized studies. In the streptomycin trial, for instance, restricting to younger patients with fulminating disease aimed to capture rapid progression amenable to , while excluding older or stabilized cases avoided dilution of efficacy signals from less responsive subgroups. Such practices laid foundational principles for later regulatory frameworks, emphasizing —evident in pre-trial monitoring for adverse reactions—and scientific rigor, though they sometimes limited generalizability by prioritizing narrow cohorts over broader representation. By the , similar criteria appeared in trials for conditions like , where exclusions for concurrent therapies ensured attribution of effects to the tested agent.

Evolution Through Regulatory Standards

The Kefauver-Harris Amendments, enacted on October 10, 1962, established requirements for drugs to be proven effective through "adequate and well-controlled investigations," necessitating detailed protocols that incorporated explicit inclusion and exclusion criteria to define study populations, ensure participant safety, and support causal inferences about efficacy. These amendments, prompted by the disaster, shifted regulatory oversight toward rigorous subject selection to minimize risks, with the FDA codifying (IND) regulations under 21 CFR Part 312 by 1963, mandating protocols specify eligibility to protect vulnerable groups. In the ensuing decades, safety imperatives led to restrictive criteria; for instance, a FDA guideline recommended excluding women of childbearing potential from Phase I and early Phase II trials to avert fetal risks, reflecting thalidomide's legacy and prioritizing over population representativeness. This approach persisted until policy reversals in the 1990s, driven by evidence of underrepresentation compromising ; the NIH Revitalization of June 10, 1993, required of women and minorities in all NIH-funded unless scientifically justified, with mandates for outreach and subgroup analyses to detect differential effects. The FDA concurrently revised its policy in 1993, permitting women's participation in early phases with contraception requirements, marking a regulatory pivot toward broader eligibility justified by accumulating data on comparable across sexes. International harmonization advanced standardization through the International Council for Harmonisation (ICH), whose E6 Guideline for Good Clinical Practice, adopted in 1996 and revised as E6(R2) in 2016, explicitly required protocols to detail subject inclusion (6.5.1) and exclusion criteria (6.5.2), emphasizing ethical selection, risk-benefit assessment, and documentation of withdrawals to uphold across ICH regions (EU, , ). Similarly, ICH (1996) on clinical study reports mandated comprehensive descriptions of eligibility in sections 9.3.1 and 9.3.2, with rationales for exclusions and their implications for generalizability, fostering consistent reporting to regulators. Contemporary regulations prioritize balancing homogeneity for internal validity with inclusivity for real-world applicability; the (2016) and FDA Reauthorization Act (2017) spurred NIH and FDA initiatives from to address age-related barriers, requiring justifications for lifespan exclusions and promoting innovative designs like adaptive . FDA's draft guidance on modernizing eligibility, informed by a 2015-2017 ASCO-Friends of Cancer Research collaboration analyzing 290 INDs (revealing 77% exclusion of active CNS metastases and 84% of patients), recommended evidence-based broadening—e.g., including stable brain metastases post-treatment or cases with >350—absent heightened toxicity risks, to enhance enrollment without undermining safety. The ICH E6(R3), adopted January 6, 2025, further evolves this by integrating risk-proportionate subject selection, urging criteria tailored to and rather than historical precedents. These standards reflect empirical critiques of overly narrow criteria limiting feasibility—e.g., liver function exclusions barring over 60% of trials' potential enrollees despite chronic conditions affecting 60% of US adults—while demanding scientific substantiation to preserve causal reliability.

Purposes and Design Principles

Ensuring Internal Validity and Study Homogeneity

Inclusion and exclusion criteria serve as foundational mechanisms to enhance by minimizing variables that could obscure the causal effects of an under investigation. Internal validity refers to the degree to which a study's design and execution accurately reflect the true impact of the independent variable on the dependent variable, without extraneous influences distorting the relationship. By specifying characteristics such as age ranges, disease severity, comorbidities, or prior treatments, these criteria ensure that participants share baseline similarities, thereby isolating the experimental factor as the primary driver of observed outcomes. For instance, in randomized controlled trials (RCTs), excluding patients with uncontrolled prevents variability from drug efficacy assessments. Study homogeneity, achieved through stringent criteria, further bolsters by reducing heterogeneity in participant responses, which could otherwise lead to inconsistent results attributable to differences rather than the itself. Homogeneous groups facilitate precise estimation and statistical , as variability unrelated to the treatment is curtailed. Guidelines from regulatory bodies emphasize this: the U.S. (FDA) recommends criteria that standardize eligibility to control for factors like genetic polymorphisms or concurrent medications that might interact with the study drug, thereby preserving the integrity of causal inferences. A 2019 analysis of trials found that tighter exclusion for (e.g., Eastern Cooperative Oncology Group score ≤2) correlated with higher scores, as measured by reduced bias in hazard ratios. However, overly restrictive criteria can inadvertently introduce , undermining if the enrolled population deviates systematically from those experiencing the condition in practice. To mitigate this, researchers apply first-principles evaluation: criteria must be justified by linking excluded factors to outcome distortion, such as excluding pregnant participants in teratogenic drug studies due to ethical and physiological confounders. Empirical data from meta-epidemiological studies indicate that trials with well-defined, protocol-adherent criteria exhibit 20-30% lower risk of invalid causal claims compared to those with vague or absent specifications. In practice, operationalizing these criteria involves prospective screening protocols, often using validated scales (e.g., for exclusion thresholds), to enforce homogeneity without compromising feasibility. This approach aligns with guidelines, which advocate transparent reporting of criteria to allow replication and bias assessment. Ultimately, while enhancing , such homogeneity prioritizes causal clarity over broader applicability, a substantiated by decades of trial data showing cleaner signal-to-noise ratios in restricted cohorts.

Balancing Internal and External Validity

Strict inclusion and exclusion criteria enhance by reducing participant heterogeneity, minimizing variables, and enabling precise causal inferences about treatment effects within the controlled study environment. For example, excluding patients with comorbidities isolates the intervention's impact, thereby strengthening the reliability of results against biases such as selection or measurement errors. Conversely, these restrictions often undermine , as trial samples frequently fail to mirror real-world populations; meta-analyses indicate ineligibility rates surpass 50% in over 40% of trials, 67% of trials, and 89% of trials, primarily due to exclusions of elderly patients, those with multiple conditions, or underrepresented demographics. This discrepancy arises because criteria optimized for homogeneity prioritize methodological purity over representativeness, limiting the generalizability of findings to diverse clinical settings. Balancing these validities requires deliberate protocol design trade-offs, such as adopting pragmatic trials with broader eligibility to incorporate real-world variability while retaining and blinding for internal rigor. Effectiveness-implementation designs further aid this by testing interventions in heterogeneous settings, using limited exclusion criteria for patients but diverse provider samples to assess both and uptake. Regulatory guidance from the FDA emphasizes diversifying criteria to boost , with simulations demonstrating that relaxing restrictions can expand eligible pools by 78%, disproportionately aiding older adults, women, non-Latinx Black individuals, and lower-socioeconomic groups without proportionally eroding internal controls. Supplementary techniques include stratified subgroup analyses to evaluate effect heterogeneity, propensity score methods to gauge transportability to excluded populations, and integration of RCT data with observational studies for complementary evidence on generalizability. These approaches ensure criteria safeguard against internal threats like confounding while promoting applicability, though researchers must justify selections based on the intervention's target population to avoid overgeneralization.

Applications in Research

In Clinical Trials

Inclusion and exclusion criteria form the foundation of participant selection in clinical trials, delineating the characteristics that potential subjects must possess to enroll () and those that render them ineligible (exclusion). These criteria are explicitly defined in the trial protocol prior to initiation, ensuring standardized screening and reducing . According to Council for Harmonisation (ICH) guideline E6(R3), sponsors must predefined criteria for or exclusion from sets to maintain integrity. In clinical trials, these criteria primarily safeguard participant by excluding individuals at elevated of adverse events, such as those with contraindicating comorbidities or concomitant medications posing interactions. They also promote by creating a homogeneous study population, minimizing confounders that could obscure the intervention's causal effects on the primary . For example, in phase III trials for relapsing-remitting , often requires a confirmed , at least one documented relapse in the prior year, and an (EDSS) score between 1.0 and 5.5, while exclusions encompass progressive disease forms, recent steroid use, or severe psychiatric conditions to isolate treatment efficacy signals. Similarly, trials typically include patients with histologically confirmed stage III/IV cancer, Eastern Cooperative Oncology Group (ECOG) performance status of 0-2, and adequate hepatic/renal function (e.g., ≤1.5 times upper limit of normal), excluding those with active brain metastases or uncontrolled to control for baseline risks. Regulatory frameworks mandate rigorous justification for these criteria to balance trial rigor with applicability. The U.S. (FDA) requires protocols to scientifically rationalize exclusions, particularly age or organ function thresholds, as overly restrictive ones can undermine evidence for post-approval use. Institutional Review Boards (IRBs) review criteria for ethical compliance, ensuring they align with processes and do not unjustly bar subgroups without evidence-based rationale. In practice, deviations from criteria—such as minor laboratory value waivers—must be documented and minimized to preserve data reliability, though FDA draft guidance clarifies not all protocol variances constitute critical violations. Application varies by trial phase: early-phase studies (I/II) impose stricter exclusions for and dose-finding, prioritizing healthy volunteers or limited patient cohorts, whereas confirmatory phase III trials broaden inclusion to approximate real-world populations while retaining exclusions for ethical safety. This design isolates causal mechanisms—e.g., excluding pregnant individuals due to teratogenicity risks—yet empirical analyses reveal that applying phase III criteria to routine care cohorts often disqualifies 70-90% of eligible patients, highlighting trade-offs in homogeneity versus representativeness. Recent FDA initiatives emphasize streamlining criteria, such as elastic organ function limits or reduced concomitant therapy bans, to enhance enrollment efficiency without compromising evidentiary standards.

In Systematic Reviews and Meta-Analyses

In systematic reviews and meta-analyses, criteria delineate the predefined characteristics that primary studies must possess to be eligible for synthesis, typically framed around the framework—population, intervention, comparator, and outcomes—to ensure alignment with the review's objectives and facilitate comparable evidence pooling. These criteria are established a priori in the review protocol to minimize and enhance reproducibility, specifying elements such as designs, minimum sample sizes (e.g., at least 50 participants per arm), or interventions delivered within a defined timeframe like post-2010 to reflect contemporary practices. Exclusion criteria, conversely, identify disqualifying features, such as non-randomized observational studies, animal-only data, or outcomes lacking statistical reporting (e.g., absence of hazard ratios or odds ratios), thereby excluding heterogeneous or irrelevant evidence that could confound results. The application of these criteria in meta-analyses particularly emphasizes methodological and clinical homogeneity to enable valid statistical synthesis, as excessive variability among included studies can inflate heterogeneity metrics like I² statistics above 50%, undermining pooled effect estimates. For instance, criteria might require studies to intention-to-treat analyses or adjust for key confounders like age and comorbidities, ensuring that random- or fixed-effects models appropriately aggregate data without introducing systematic error. PRISMA guidelines mandate transparent of these criteria in the methods section, including rationale for thresholds, to allow of review rigor; deviations, such as post-hoc exclusions, are discouraged as they risk cherry-picking data inconsistent with the original protocol. Empirical evidence underscores their role in : a analysis of Cochrane reviews found that strict PICO-based criteria reduced between-study variance by up to 30% in meta-analyses of pharmacological interventions, correlating with narrower confidence intervals around summary effects. However, overly restrictive criteria can yield sparse datasets—e.g., fewer than five studies eligible for pooling—prompting sensitivity analyses or subgroup explorations to test robustness, while broader criteria necessitate tools like funnel plots to detect . In practice, dual independent screening by reviewers, with adjudication for borderline cases, operationalizes these criteria, achieving inter-rater agreement rates of 80-95% in high-quality reviews.

Examples and Practical Implementation

Hypothetical Example in a Cardiovascular Trial

In a hypothetical III investigating the efficacy of a novel lipid-lowering agent in reducing (MACE) among patients with established atherosclerotic (ASCVD), inclusion criteria would typically target a homogeneous to minimize variability in risk and treatment response. Eligible participants might include adults aged 40 to 80 years with a documented history of or ischemic stroke occurring at least 6 months but no more than 5 years prior, cholesterol (LDL-C) levels between 70 and 189 mg/dL despite maximally tolerated therapy, and adherence to guideline-directed medical therapy for secondary prevention. These criteria ensure by selecting individuals with similar disease severity and prognostic factors, thereby isolating the drug's effect on outcomes like cardiovascular death or nonfatal MI, as supported by trial designs in similar ASCVD studies where age and event history reduced . Exclusion criteria would further refine the by omitting factors that could introduce risks or assessments. Common exclusions might encompass severe renal impairment (estimated <30 mL/min/1.73 m²), active liver disease (alanine aminotransferase >3 times the upper limit of normal), uncontrolled hypertension (systolic >180 mmHg), or , and concomitant use of investigational drugs or certain high-risk comorbidities like advanced . Such exclusions protect participant and maintain study homogeneity; for instance, renal exclusions prevent disproportionate adverse events in subgroups with altered , a evidenced in cardiovascular trials where independently predicts poor outcomes and complicates attribution of causality to the intervention.
Criterion TypeExamplesRationale
InclusionAge 40-80 years; prior / 6 months-5 years ago; LDL-C 70-189 mg/dL on statinsDefines target population with modifiable risk; ensures ethical feasibility and statistical power by focusing on prevalent ASCVD phenotypes.
Exclusion <30 mL/min/1.73 m²; ALT >3x ULN; SBP >180 mmHgMitigates risks and confounders; avoids dilution of treatment effect from heterogeneous responses in high-comorbidity states.
This design balances internal rigor with feasible recruitment, though it may limit generalizability to younger patients or those with milder disease, highlighting trade-offs inherent in criterion selection for cardiovascular research.

Criteria in Observational Studies

In observational studies, such as , case-control, and cross-sectional designs, inclusion and exclusion criteria define the eligible population to address the , reduce , and facilitate control for confounders without the benefit of . These criteria specify characteristics like age, exposure status, disease presence, or comorbidities that participants must meet or lack, ensuring the study groups are comparable and relevant to the hypothesized associations. For instance, in a examining and risk, inclusion might require participants aged 40-70 with verified smoking history, while exclusion could eliminate those with prior cancer diagnoses to avoid reverse causation. Compared to randomized controlled trials, which impose narrow criteria for and homogeneity, observational criteria are typically broader to reflect real-world variability and enhance , though they must still mitigate like healthy user effects or immortal time . Retrospective observational designs, common in , rely on these criteria during data abstraction from records, prioritizing verifiable diagnoses via codes (e.g., ) to ensure diagnostic accuracy. The STROBE reporting guidelines emphasize detailing eligibility criteria, case ascertainment methods, and matching (if used) to enable evaluation, noting that explicit /exclusion is preferable but not always essential if selection processes are transparent.61602-X/fulltext) Practical implementation involves pre-specifying criteria in protocols to prevent post-hoc adjustments, with analyses testing robustness by varying thresholds (e.g., excluding mild vs. severe comorbidities). In case-control studies, criteria distinguish cases (e.g., incident confirmed by ) from controls (e.g., no cardiovascular events, frequency-matched by and ), excluding ambiguous cases to minimize misclassification. Challenges include incomplete in administrative databases, addressed by criteria mandating minimum follow-up duration, such as 12 months, to assess outcomes reliably. Adherence to these criteria correlates with higher study quality, as evidenced by meta-epidemiologic reviews showing reduced heterogeneity when selection is rigorous.

Controversies and Criticisms

Limitations on Generalizability

Strict inclusion and exclusion criteria in , designed to minimize heterogeneity and enhance , frequently restrict participant diversity, resulting in study populations that poorly represent real-world patient demographics and thereby undermine the of findings. For example, criteria often exclude individuals with common comorbidities, advanced age, or , which characterize a substantial portion of clinical populations; in , 60% of adults have at least one , and 42% have multiple. This selective enrollment can lead to inflated estimates of or when results are extrapolated beyond the trial cohort. Quantifiable highlights the scale of this issue across trial types. A of 305 randomized controlled trials (RCTs) for in musculoskeletal conditions reported a median exclusion rate of 77.1% (interquartile range: 55.5%–89.0%), with frequent criteria targeting , comorbidities, and concurrent medications, rendering results inapplicable to typical older or multimorbid patients encountered in practice. Similarly, in cancer trials analyzed from electronic health records of 235,234 patients across 22 types (2013–2022), strict criteria rendered only 48% of patients eligible, disproportionately excluding older adults ( 3.04 for 75+ versus 18–49), females, non-Latinx Black individuals, and lower groups; broadening criteria increased eligibility by 78%, underscoring how narrow standards limit relevance to diverse clinical realities. In chronic pain studies, such as those for low back pain, exclusion criteria eliminated 39.4% of potential participants (452 out of 1,148), who were characteristically older, less employed, more functionally limited, and opioid-dependent—groups that experienced modestly worse pain relief (mean difference 0.4/10 on visual analog scale) and functional gains compared to included patients. FDA analyses further reveal that approximately 27% of trials for prevalent diseases impose arbitrary upper age limits, underrepresenting older adults despite their higher disease burden, which creates evidentiary gaps for polypharmacy and frailty effects in routine care. These patterns persist because criteria prioritize statistical power and confounder control over inclusivity, yet they risk misguiding policy or practice when trial successes fail to replicate in heterogeneous populations. Pragmatic trial designs, which relax exclusions, have been proposed to bridge this divide, though they introduce challenges in isolating causal effects.

Underrepresentation of Diverse Populations

Strict inclusion and exclusion criteria in clinical trials, designed to enhance by minimizing variables, often disproportionately exclude members of racial, ethnic, , , and socioeconomic minorities, leading to underrepresentation relative to demographics. For instance, a 2024 population-based study of trials found that common exclusion criteria, such as limits over 80 or comorbidities like renal impairment, would exclude a higher proportion of women and patients compared to White men, potentially reducing their enrollment by up to 20-30% in real-world applicability. Similarly, analyses of trials indicate that broadening eligibility criteria could increase eligible patients from underrepresented groups—such as older adults, females, non-Latinx individuals, and those of lower —by 78% or more, highlighting how restrictive protocols limit access without always advancing scientific rigor. Racial and ethnic minorities remain notably underrepresented in U.S. clinical trials despite comprising significant portions of the population; for example, a review of over 32,000 participants in new drug trials approved in revealed only 8% and 6% Asian enrollment, compared to their 13.6% and 6% shares of the U.S. population, respectively. Reporting gaps exacerbate this issue, with more than half of trials registered on from 2000-2020 failing to disclose / data, and only 43% of U.S.-based trials with results providing any such breakdown. Historical patterns persist, as patients constituted just 5-8% of participants in many pivotal trials for conditions like vaccines and cancer therapies, often due to criteria excluding those with higher burdens prevalent in these groups. This underrepresentation undermines , as trial outcomes may not generalize to diverse patient populations, potentially resulting in therapies less effective or safe for excluded groups; for example, drugs tested predominantly on males have shown differential in minorities, as seen in cardiovascular and studies where pharmacogenomic variations by were overlooked. Critics argue that overly stringent criteria, justified by concerns but rarely evidence-based for broad exclusions like or mild disabilities, perpetuate inequities without proportional benefits to study homogeneity. Regulatory responses, such as FDA mandates for action plans in trials submitted after , aim to address this by requiring stratified enrollment targets, though implementation challenges persist due to barriers and the tension with maintaining integrity.

Debates Over Regulatory Mandates for Inclusion

Regulatory agencies, particularly the U.S. (FDA), have increasingly imposed mandates requiring sponsors to develop Diversity Action Plans (DAPs) for pivotal clinical trials, as stipulated by the Food and Drug Omnibus Reform Act (FDORA) enacted in December 2022. These plans mandate strategies to enhance enrollment of historically underrepresented groups, such as racial/ethnic minorities, older adults, and pregnant individuals, with requirements applying to Phase 3 drug trials and certain device studies starting in 2025. Proponents of these mandates, including FDA officials and advocates, assert that they address longstanding underrepresentation—such as Black patients comprising only 5% of participants despite higher burdens in some conditions—thereby improving post-approval safety monitoring and generalizability to real-world populations. For instance, the FDA's 2024 draft guidance emphasized DAPs as tools to mitigate approval delays for underserved groups, citing evidence that homogeneous trials have led to adverse events in diverse populations post-market, as seen in historical cases like the higher renal toxicity of certain drugs in patients. Supporters also highlight ethical imperatives, arguing that exclusion perpetuates inequities, with economic analyses estimating billions in lost productivity from non-generalizable results. Critics, including representatives and some clinical researchers, contend that these mandates impose undue administrative burdens and costs—potentially adding 10-20% to trial expenses through expanded efforts—without robust that demographic quotas enhance scientific validity. Stakeholder comments submitted to the FDA in revealed tensions over the plans' scope, with concerns that vague goals could function as quotas, complicating trial design and risking underpowered subgroup analyses that obscure treatment heterogeneity driven by genetic or physiological differences rather than . For example, mandating inclusion across diverse ancestries may dilute overall trial efficacy signals if pharmacokinetics vary significantly, as documented in pharmacogenomic studies showing ancestry-linked rates. The debate intensified in early 2025 following executive actions under the Trump administration targeting (DEI) initiatives, prompting the FDA to quietly remove its draft DAP guidance from its website on January 23, 2025, without formal announcement or replacement. This move created regulatory uncertainty for sponsors, with critics arguing it underscores the ideological overreach of prior mandates, while proponents warned of setbacks to equity goals, noting that international regulators like the continue emphasizing voluntary without binding plans. Empirical analyses suggest that while correlates with broader applicability, forced absent disease-specific relevance can compromise , as evidenced by trials where subgroup imbalances led to misleading primary endpoints.
AspectArguments For MandatesArguments Against Mandates
Scientific ImpactEnhances generalizability; reduces post-approval surprises (e.g., 30% of drugs show differences by ).Risks diluting statistical power; ignores biological heterogeneity not aligned with demographic proxies.
Practical EffectsPromotes ethical access; potential economic savings from better-targeted therapies.Increases costs and timelines; enforcement ambiguity leads to compliance burdens without clear benefits.
Regulatory StabilityStandardizes practices across trials; aligns with global trends.Vulnerable to political shifts, as seen in 2025 guidance withdrawal.

References

  1. [1]
    Inclusion and exclusion criteria in research studies - NIH
    Inclusion criteria are defined as the key features of the target population that the investigators will use to answer their research question.
  2. [2]
    [PDF] EVALUATING INCLUSION AND EXCLUSION CRITERIA IN ... - FDA
    Inclusion criteria specify characteristics for study entry, while exclusion criteria specify characteristics that disqualify patients from participation in  ...
  3. [3]
    Implementation of inclusion and exclusion criteria in clinical studies ...
    Dec 18, 2023 · The correct use of medical classification systems aiming at clearly defined inclusion and exclusion criteria in the studies is an important ...
  4. [4]
    ClinicalTrials.gov Glossary Terms
    May 21, 2025 · Eligibility criteria consist of both inclusion criteria (which are required for a person to participate in the study) and exclusion criteria ( ...
  5. [5]
    Eligibility criteria and clinical trials: An FDA perspective - PubMed
    Jul 27, 2021 · The objective of this study is to identify common trends in eligibility criteria and identify patterns of exclusion criteria among different diseases.Missing: studies | Show results with:studies
  6. [6]
    Scientific Rationale for the Inclusion and Exclusion Criteria for ...
    ... inclusion criteria for intravenous alteplase. This eligibility ... Similar to earlier reviews of intravenous alteplase exclusion criteria, the ...<|separator|>
  7. [7]
    Excluding People With Disabilities From Clinical Research
    Oct 3, 2022 · The exclusion of people with disabilities from clinical research without appropriate justification is discriminatory, is counter to federal regulations and ...
  8. [8]
    Inclusion Criteria - Clinical Research Explained - VIARES
    Aug 28, 2024 · The inclusion criteria are established by the research team before the study begins and are designed to ensure the safety of the participants ...
  9. [9]
    [PDF] Designing Inclusion and Exclusion Criteria
    Jul 29, 2020 · Another approach is to start with several inclusion/exclusion criteria, then remove exclu- sion or inclusion criteria if recruitment becomes ...Missing: peer- | Show results with:peer-
  10. [10]
  11. [11]
    Inclusion and Exclusion Criteria | Examples & Definition - Scribbr
    Sep 17, 2022 · Ethnographies and a few other types of qualitative research do not usually specify exclusion criteria. However, inclusion criteria help ...What are exclusion criteria? · Examples of inclusion and... · Why are inclusion and...
  12. [12]
    Choosing inclusion criteria that minimize the time and cost of clinical ...
    Patient numbers and total cost are strongly related to the choice of the cutoff for inclusion. Clear cost minimums exist between 5.6 and 6.1 on a 10-point ...
  13. [13]
    Encyclopedia of Research Design - Exclusion Criteria
    Exclusion criteria are a set of predefined definitions that is used to identify subjects who will not be included or who will have to withdraw from a ...
  14. [14]
    Methodology for research I - PMC - PubMed Central - NIH
    The exclusion criteria include factors or characteristics that make the recruited population ineligible for the study. These factors may be confounders for the ...
  15. [15]
    [PDF] Evaluating Inclusion and Exclusion Criteria in Clinical Trials
    Apr 16, 2018 · Discussion will encompass underrepresentation in clinical trials, how eligibility criteria impact patient access to investigational drugs and ...
  16. [16]
    A literature review of the impact of exclusion criteria on ... - NIH
    Nov 11, 2022 · We summarize and report new insights on published studies that report on how trial exclusions affect the generalizability of their results.
  17. [17]
    The Basics | National Institutes of Health (NIH)
    Apr 24, 2025 · Inclusion and exclusion criteria are not used to reject people personally. Instead, the criteria are used to identify appropriate participants ...
  18. [18]
    Evolution of Clinical Research - LWW
    The idea of randomization was introduced in 1923. However, the first randomized control trial of streptomycin in pulmonary tuberculosis was carried out in 1946 ...
  19. [19]
    Legumes, lemons and streptomycin: A short history of the clinical trial
    Jan 6, 2009 · The first widely publicized randomized clinical trial was a 1948 test of streptomycin for treating pulmonary tuberculosis (BMJ 1948;2:769-82).Missing: criteria | Show results with:criteria<|separator|>
  20. [20]
    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 ...
  21. [21]
    The MRC randomized trial of streptomycin and its legacy - NIH
    It was decided to limit the patients participating in the trial to those aged between 15 and 30 with 'acute progressive bilateral pulmonary tuberculosis of ...
  22. [22]
    UK Medical Research Council and multicentre clinical trials: from a ...
    Why the 1948 MRC trial of streptomycin used treatment allocation based on random numbers. ... If [the patient] satisfied the criteria for admission he was ...
  23. [23]
    rethinking eligibility and generalizability in clinical trials - The Lancet
    Jun 14, 2025 · ... 1948, evaluating streptomycin for the treatment of pulmonary tuberculosis, ... criteria aimed at reducing variability and insuring patient ...
  24. [24]
    STREPTOMYCIN TREATMENT OF PULMONARY TUBERCULOSIS
    Oct 31, 1998 · Condition on Admission​​ Each patient was under observation at a centre for at least one week before streptomycin treatment or observation proper ...
  25. [25]
    A BRIEF HISTORY OF THE RANDOMIZED CONTROLLED TRIAL
    In the late 1980s, for example, groups such as the Institute for Research on Women's Health documented the exclusion or artificial restriction of women from ...
  26. [26]
    The 50th Anniversary of the Kefauver‐Harris Amendments: Efficacy ...
    Oct 3, 2012 · The Kefauver‐Harris Amendments are a landmark driving force in the development of the RCT, including both scientific and ethical components.
  27. [27]
    Promoting Safe & Effective Drugs for 100 Years - FDA
    Apr 23, 2019 · "Also critically, the 1962 amendments required that the FDA specifically approve the marketing application before the drug could be marketed, ...Missing: impact protocols criteria
  28. [28]
    History of Women's Participation in Clinical Research
    Apr 24, 2024 · Policies that encourage the inclusion of women in research originated during the women's health movement, which emerged as part of the women's movement.
  29. [29]
    NIH Policy and Guidelines on the Inclusion of Women and Minorities ...
    Jul 21, 2025 · NIH policy requires inclusion of women and minorities in all clinical research, unless a compelling rationale justifies exclusion. Cost is not ...
  30. [30]
    Inclusion of Women in Clinical Trials: A Historical Overview of ...
    A historical overview of why women previously were excluded from clinical trials is presented, and the reasons for current policy changes are discussed.
  31. [31]
    [PDF] Integrated Addendum to ICH E6(R1): Guideline for Good Clinical ...
    Nov 9, 2016 · 6.5 Selection and Withdrawal of Subjects. 6.5.1 Subject inclusion criteria. 6.5.2 Subject exclusion criteria. 6.5.3 Subject withdrawal ...
  32. [32]
    [PDF] Structure And Content of Clinical Study Reports E3 - ICH
    The objective of this guideline is to allow the compilation of a single core clinical study report acceptable to all regulatory authorities of the ICH regions.
  33. [33]
    [PDF] Modernizing Eligibility Criteria for Clinical Trials | FDA
    Exclusions on basis of cardiac disease may decrease enrollment of older patients by ~5%. • Due to historical precedent patients must have EF of >45-50%. • ...
  34. [34]
    [PDF] GUIDELINE FOR GOOD CLINICAL PRACTICE E6(R3) - ICH
    Jan 6, 2025 · This Guideline has been developed by the appropriate ICH Expert Working Group and has been subject to consultation by the regulatory parties, in ...
  35. [35]
    Study Design, Precision, and Validity in Observational Studies - PMC
    Observational studies are evaluated in terms of both internal and external validity. Internal validity refers to the strength of the inferences from the study.
  36. [36]
    A literature review on the representativeness of randomized ...
    Nov 3, 2015 · In general, a thoughtful approach to RCT design is required in which the trade-offs between internal and external validity are considered in a ...<|separator|>
  37. [37]
    How to Assess the External Validity and Model Validity of ...
    This paper describe this model and offers an EV assessment tool (EVAT©) for weighing studies according to EV and MV in addition to IV.
  38. [38]
    Effectiveness-implementation Hybrid Designs - PubMed Central - NIH
    Review of hybrid designs requires at least an appreciation of the complexities balancing internal and external validity considerations in such trials, as well ...
  39. [39]
    Study designs for effectiveness and translation research - PubMed
    Consideration of the study design choices, trade-offs, and enhancements discussed here can guide the design, funding, completion, and publication of ...
  40. [40]
    Broadening Eligibility Criteria and Diversity among Patients for ...
    Mar 26, 2024 · Broadening criteria increased the number of eligible patients by 78%, with the strongest impact for older, female, non-Latinx Black, and lower-SES patients.
  41. [41]
    [PDF] E6(R3) Good Clinical Practice (GCP) | Guidance for Industry - FDA
    May 19, 2023 · (d) The sponsor should ensure that the criteria for inclusion or exclusion of trial participants from any analysis set is predefined (e.g. ...
  42. [42]
    Effect of applying inclusion and exclusion criteria of phase III clinical ...
    Jan 20, 2021 · 83% of MS patients in routine care would not have been eligible for phase III trials, with relapse being the most frequent exclusion criterion ...
  43. [43]
    FDA Draft Guidance Encourages Broader Inclusion Criteria in ...
    May 22, 2024 · However, the FDA argues that these criteria are sometimes more restrictive than necessary, potentially limiting patient access, slowing trial ...
  44. [44]
    Recruiting Study Subjects - FDA
    Sep 5, 2018 · FDA requires that an Institutional Review Board (IRB) review and have authority to approve, require modifications in, or disapprove all ...
  45. [45]
    Clinical Trial Protocol Deviations: A New FDA Draft Guidance to ...
    Jan 6, 2025 · The Draft Guidance highlights that FDA does not consider all potential violations of good clinical practice guidelines or requirements (“GCPs”) ...<|separator|>
  46. [46]
    Chapter 3: Defining the criteria for including studies and how they ...
    In particular, post-hoc decisions about inclusion or exclusion of studies should keep faith with the objectives of the review rather than with arbitrary rules.
  47. [47]
    Introduction to systematic review and meta-analysis - PMC
    Apr 2, 2018 · Defining inclusion and exclusion criteria. Information is included on the study design, patient characteristics, publication status (published ...
  48. [48]
    Systematic review and meta-analysis in clinical trials - ScienceDirect
    Develop inclusion and exclusion criteria – establish criteria to determine which studies are eligible for inclusion in the meta-analysis based on factors such ...
  49. [49]
    The PRISMA 2020 statement: an updated guideline for reporting ...
    Mar 29, 2021 · PRISMA 2020 is intended for use in systematic reviews that include synthesis (such as pairwise meta-analysis or other statistical synthesis ...
  50. [50]
    PRISMA statement
    PRISMA provides authors with guidance and examples of how to completely report why a systematic review was done, what methods were used, and what results were ...Prisma 2020 · PRISMA extensions · Flow diagram · PRISMA translations
  51. [51]
    Inclusion and Exclusion Criteria - Systematic Reviews - LibGuides
    Jul 9, 2025 · Inclusion criteria is everything a study must have to be included. Exclusion criteria are the factors that would make a study ineligible to be included.Missing: trials | Show results with:trials
  52. [52]
    Semaglutide in Patients with Heart Failure with Preserved Ejection ...
    Aug 25, 2023 · Persons 18 years of age or older were eligible to participate if they had a left ventricular ejection fraction of at least 45%; a body-mass ...
  53. [53]
    NCT03574597 | Semaglutide Effects on Heart Disease and Stroke in ...
    Eligibility Criteria. Description. Inclusion Criteria: Informed consent obtained before any trial-related activities. Trial-related activities are any ...Semaglutide Effects On Heart... · Contacts And Locations · Study Plan
  54. [54]
    Characteristics of Populations Excluded From Clinical Trials ...
    Mar 23, 2021 · All adults aged <40 years were excluded because of trial age requirements, and most younger adults met at least 1 additional exclusion criteria.
  55. [55]
    The Exclusion of Older People from Participation in Cardiovascular ...
    Poorly justified criteria included comorbidity described in a non-specific manner, use of medications that would not impact the study protocol, and visual and ...
  56. [56]
    Heart failure clinic inclusion and exclusion criteria - BMJ Open
    The most common exclusion criteria were that the patient's HF was secondary to congenital heart disease or pulmonary hypertension (16.1%), required intravenous ...
  57. [57]
    Randomized controlled trials vs. observational studies - NIH
    Oct 21, 2016 · The strengths of RCTs are obvious and include the development of a prospective study protocol with strict inclusion and exclusion criteria, a ...
  58. [58]
    Strengthening the Reporting of Observational Studies in ...
    Eligibility criteria may be presented as inclusion and exclusion criteria, although this distinction is not always necessary or useful.
  59. [59]
    The STROBE guidelines - PMC - PubMed Central - NIH
    The inclusion and exclusion criteria and methods to overcome any potential bias should be noted down as well. The method used to establish the study size ...
  60. [60]
    Exclusion rates in randomized controlled trials of treatments for ...
    Feb 26, 2020 · The most commonly applied exclusion criteria related to age, co-morbidity and co-prescribing, whereas more implicit criteria relating to life ...
  61. [61]
    rethinking eligibility and generalizability in clinical trials
    This exclusion commonly results from restrictive eligibility criteria aimed at reducing variability and insuring patient safety. However, this exclusion can ...
  62. [62]
    Clinical Trial Exclusion Criteria Affect Trial Inclusivity by Race and Sex
    Jun 21, 2024 · In our population‐based study, we found that common stroke clinical trial exclusion criteria have the potential to exclude more women and Black ...
  63. [63]
    Embracing Diversity: The Imperative for Inclusive Clinical Trials
    Jun 30, 2023 · If they only include participants from a narrow demographic group, the results may not accurately represent the broader population. This can ...
  64. [64]
    Race/ethnicity reporting and representation in US clinical trials
    Apr 10, 2022 · Among 20,692 US-based trials with reported results (representing ∼4·76 million enrollees), only 43% (8,871/20,692) reported any race/ethnicity ...
  65. [65]
    Why Diverse Clinical Trial Participation Matters
    Apr 1, 2023 · The goals of increasing diversity in clinical trial participation include earning and building trust, promoting fairness, and generating biomedical knowledge.<|separator|>
  66. [66]
    The Evolution of US FDA Diversity Requirements in Clinical Research
    Broadening eligibility criteria to include more diverse populations (e.g., sex, race, ethnicity, age, etc.) · Using study designs and mechanisms that reduce the ...
  67. [67]
    FDA Issues Overdue Guidance on Diversity Action Plans in Drug ...
    Jul 16, 2024 · This update provides key takeaways on the new draft guidance and Diversity Action Plan requirements, including when the new requirements will go ...
  68. [68]
    Why Diverse Representation in Clinical Research Matters ... - NCBI
    Lack of representation costs hundreds of billions of dollars. It is important to also quantify the potential economic benefits of greater inclusion in clinical ...
  69. [69]
    FDA officials clarify how diversity action plans intersect with ... - RAPS
    Jan 15, 2025 · FDA officials recently clarified how sponsors can achieve agency diversity action plan (DAP) goals within multiregional clinical trials (MRCTs).<|separator|>
  70. [70]
    Diversity Action Planning: 5 Myths About the FDA's Recent Guidance
    Oct 10, 2024 · Implementing diversity action plans significantly increases the cost and complexity of clinical trials. While there may be some additional costs ...
  71. [71]
    Stakeholder comments on FDA's Diversity Action Plan guidance ...
    Oct 10, 2024 · AgencyIQ reviewed the hundred-plus comments on this document, finding tension among stakeholder groups that the FDA may find difficult to reconcile.
  72. [72]
    The Role of Diverse Populations in US Clinical Trials - ScienceDirect
    Jan 15, 2021 · The potential of improving racial/minority enrollment in clinical trials has a direct influence on survival outcomes in these populations.
  73. [73]
    FDA purges clinical trial diversity pages - STAT News
    Jan 23, 2025 · FDA purges material on clinical trial diversity from its site, showing stakes of Trump DEI ban · The scrubbing could affect the ways researchers ...
  74. [74]
    FDA Quietly Removes Draft Guidance on Diversity in Clinical Trials ...
    Jan 31, 2025 · The FDA removed previously issued draft guidance on diversity in clinical trials from its website without public notice or explanation.
  75. [75]
    DEI in Clinical Research: A Pause, Not a Permanent Stop
    Outside the US, regulators such as the EMA continue to emphasize inclusion of diverse patients in clinical trials. International guidelines, including ICH E6 ( ...Missing: debates | Show results with:debates
  76. [76]
    Exploring the barriers to, and importance of, participant diversity in ...
    Mar 19, 2024 · A key reason for diversifying clinical trial populations is to improve fairness and to enable underserved groups to access novel treatments.