2.2 Context of use
With patient-informed endpoints giving your strategy a strong, empathetic core, the next step is to define exactly how those endpoints will operate within your trial—their specific role, scope, and relevance.
This is where the [Context of Use (COU)] comes in, serving as the blueprint that ensures your digital measures are precise, appropriate, and ready for regulatory scrutiny.
Think of COU as the “who, what, when, and why” that ensures your digital measure is fit-for-purpose.
FDA’s Jonas Santiago explains the importance of context of use
Digital Endpoints: Why Language Matters (Sept 13, 2023)
OVERVIEW
Understanding context of use
Every credible digital endpoint strategy begins with a clearly defined COU – a concise statement outlining how your endpoint will be applied in the trial: the population it serves, the clinical setting, the specific aspect of health it measures, and how it contributes to decision-making.
Anchoring the COU to both the Concept of Interest and the Meaningful Aspect of Health (see Section 2.1: Patient-informed endpoints) ensures the endpoint reflects patient priorities and regulatory expectations. A well-scoped COU also positions the team to estimate a minimal clinically important difference (MCID), the threshold of change that is both detectable and meaningful in clinical terms.

Adopters, this aligns the endpoint with your trial’s objectives and regulatory pathway, ensuring it integrates smoothly into protocols and analysis plans.

Developers, detail how your technology supports that use case, from data accuracy in real-world conditions to compatibility with the intended population.
Getting the COU right early prevents downstream issues, like mismatched data or rejected submissions, and builds confidence that your measure advances meaningful clinical insights.
A note on terminology – Context of use versus indications for use
Intended use / Indications for use:
- Describes what the medical device will be cleared/approved to do
- Example: “Intended to measure heart rate in adults”
- Relevant when seeking device marketing authorization
Context of use (COU):
- Describes how an endpoint or measure will be used in a specific trial
- Example: “Daily step count in COPD patients as secondary endpoint in Phase 3 trial”
- Relevant when using sDHTs to generate clinical trial endpoints
Both CDER and CDRH use “context of use” when discussing endpoints. CDRH additionally uses “intended use” or “indications for use” when discussing device marketing authorization.
In this roadmap: When we discuss establishing fitness-for-purpose for endpoints captured by sDHTs, we’re referring to context of use. Device marketing authorization (intended use) is a separate regulatory question.
in practice
For digital endpoints in clinical trials, the context of use typically includes:
What is being measured (e.g., walking speed, sleep efficiency, arrhythmias)
The population being studied (e.g., adults with early-stage Parkinson’s disease)
The purpose of the measure (e.g., as a primary efficacy endpoint, exploratory biomarker)
How and where the data is collected (e.g., continuous passive monitoring using a wrist-worn accelerometer in a home setting)
Duration and frequency of measurement
Example: A COU might specify using a specific wearable to track daily step count in adults with multiple sclerosis to assess mobility improvements over six months in a Phase II study.
Stakeholder considerations

Adopter considerations
For adopters, defining COU is the foundation of trial credibility. Adopters should:
- Begin with the clinical question. Clarify the patient experience or disease process the endpoint is meant to capture. This keeps the measure tied to an unmet need, not the capabilities of any single technology. The work around patient-prioritized aspects of health and concepts of interest outlined in Section 2.1: Patient-informed endpoints are a critical orientation. If you already have a tool and want to understand which clinical questions it may be most appropriate for, this work will be essential.
- Classify the endpoint appropriately. Distinguish whether the measure is a clinical outcome assessment (COA) or a biomarker (discussed below), as this shapes evidentiary standards and validation expectations (see Section 4: Your validation strategy for more on regulatory implications).
- Engage patients and clinicians. Input from patient communities, advocacy groups, and clinical experts grounds the endpoint in outcomes that are both relevant and interpretable. This input should inform MCID estimation, ensuring the trial is designed to detect a change that is meaningful.
- Look to precedent. Reviewing existing validation evidence and published endpoints helps avoid duplication and strengthens the rationale for advancing a novel measure.

Developer contributions
Developers play an equally important role in bringing clarity and feasibility to this stage:
- Translate concepts into measures. Map the pathway from the intended endpoint to the measures their sDHT captures, showing how technical measures align with clinical intent.
- Clarify technical assumptions. Share what is known about operating conditions, and data pipelines, developers help adopters judge whether the measure is feasible in the proposed COU.
- Support MCID estimation. Provide evidence that anchors what degree of change is technically detectable, complementing clinician and patient insights about what degree of change is meaningful.
Due Diligence
Before finalizing your COU, pause for a landscape review.
Before initiating new evidence generation, it is essential to first survey the existing landscape. Early review of public data sources reveals precedent and maturity, and can identify gaps that justify novel digital endpoints. This ensures you are not duplicating efforts and helps assess whether there is an unmet measurement need.
This step helps you avoid duplicating existing efforts, strengthen endpoint rationale, and support regulatory conversations with evidence of unmet need or validation precedent.
Sources to consider as you review the existing landscape:
- Peer-reviewed scientific literature
- FDA databases (e.g., 510(k), PreMarket Approval), summary letters, and Voice of the Patient/PFDD meetings
- Core digital measures developed in precompetitive working groups like DATAcc by DiMe (see the Industry spotlight below)
FDA SPOTLIGHT
“FDA encourages collaboration among multiple stakeholders and the use of methods to combine and leverage existing data (e.g., national registry data, archival databases, published literature) to fit the specific needs of the research questions and study goals.”
– Section II.F.2 (Leveraging Existing Data), p. 19
Patient-Focused Drug Development: Collecting Comprehensive and Representative Input
Patient experience data encompass a range of inputs, including symptom burdens, treatment impacts, patient preferences, and views on unmet medical needs.
These data inform all stages of medical product development, from discovery to post-market use.
Quantitative methods (e.g., surveys) provide numerical insights, while qualitative methods (e.g., interviews) offer in-depth understanding. Mixed methods combine both for a fuller perspective.Social media and verified patient communities present novel data collection opportunities but require consideration of verification and representativeness challenges.
Probability sampling (e.g., stratified random sampling) is emphasized for generalizability, while non-probability methods (e.g., convenience sampling) are useful for exploratory research. Representativeness ensures that patient input reflects the diversity and heterogeneity of the target population.
Data collection should adhere to good clinical practices and regulatory standards.
Research protocols should address missing data, quality assurance, and confidentiality.
Early collaboration with the FDA is recommended to align on study designs and regulatory requirements.
Recommendations
Define clear research objectives and determine specific research questions before selecting data collection methods.
Use probability sampling methods whenever feasible to ensure representativeness of the target population.
Address data quality through rigorous planning, data management, and adherence to FDA-supported standards.
Incorporate diverse perspectives by including underrepresented patient populations, tailoring methods to specific subgroups as needed.
Leverage existing data sources, such as patient registries and literature, to complement primary data collection efforts.
Regulatory Considerations
Data submitted to FDA should include clear documentation of the study protocol, intended use, and data collection methodologies.
Researchers must comply with human subject protection regulations (e.g., 21 CFR Parts 50 and 56) and good clinical practice guidelines.
For data intended to support regulatory submissions, adherence to FDA-supported data standards (e.g., CDISC) is strongly encouraged.
Missing data should be addressed through pre-planned strategies and summarized in the study report.
Patient experience data must meet methodological rigor to ensure their reliability and relevance for regulatory decision-making.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.
what’s at stake?
Biomarkers vs. Clinical Outcome Assessments
A key part of shaping your COU involves deciding whether your endpoint functions as a [biomarker] or a [clinical outcome assessment (COA)]. Why does this distinction matter? Because there are different evidentiary expectations.
Biomarker
clinical outcome
assessment
Biomarkers are objective indicators of biological processes, disease states, or responses to treatment. In the context of sDHTs, these may include physiologic or behavioral signals captured by sensors that are intended to reflect underlying biology rather than direct patient experience.
Clinical outcome assessments (COAs) measure how a patient feels, functions, or survives and are intended to directly reflect treatment benefit. When derived from sDHTs, COAs translate digital signals into measures that represent patient-relevant aspects of health within a defined context of use.
Evidentiary expectations
A biomarker requires analytical validation to demonstrate that the digital signal and processing pipeline accurately and reliably measure the biological process of interest, along with clinical validation appropriate to its intended role in decision-making. Additional justification is needed if the biomarker is proposed as a surrogate for a patient-meaningful outcome, consistent with FDA’s biomarker-related guidance and terminology.
Evidentiary expectations
A COA also requires clinical validation to demonstrate that the endpoint meaningfully reflects change in the concept of interest within the defined context of use. Patient input is essential to establish relevance, and when COAs are derived from sDHTs, analytical validation of signal acquisition and processing is necessary to ensure the measure is reliable and interpretable.
Diving deeper
Validation and regulatory implications
This determination influences your validation strategy and regulatory interactions, so weigh it carefully against your overall aims.
These categories are straightforward until they aren’t. Increasingly, sDHTs capture outcomes that are just below the threshold of human perception. Measurements may not be directly noticeable to patients but could still reflect meaningful aspects of their daily functioning or disease experience.
If you’re unsure whether your measure leans toward a digital biomarker or a COA, start by examining its primary focus. Ask:
Does it prioritize biological signals independent of patient perception, or does it center on how the patient experiences their health?
For borderline cases, like a sleep tracker that combines physiological data with self-reported quality, consult frameworks from the FDA or collaborative groups to clarify the classification. Engaging cross-functional experts early resolves ambiguities and guides your evidence-gathering. This clarity ensures your COU remains sharp and defensible.
Researchers and regulators tackle definitions and implications of COAs and biomarkers
Digital Endpoints: Why Language Matters (Sept 13, 2023)
Endpoint classification: Key questions to consider
Core classification questions
1. Directness of clinical benefit
Does the endpoint aim to indicate a biological process/pathophysiology or a response to exposure/intervention?
→ This is a biomarker
Does the endpoint aim to describe how a person feels, functions, or survives?
→ This is a COA
2. Trial role
Is the endpoint for diagnostic support, safety signals, pharmacodynamic/response, monitoring/prognostic/predictive use?
→ This supports biomarker
Is the endpoint for evaluating treatment benefit meaningful to patients (e.g., symptom relief, functional improvement)?
→ This supports COA
3. COA “fit” check
Can you specify a valid respondent/source (patient, clinician, caregiver)?
Does the measure involve a standardized task with clear instructions and scoring?
→ ‘No’ to either suggests reconsidering a biomarker
→ ‘Yes’ to either supports COA
4. Final step
If these indicators conflict, return to your primary intent statement and select the class that best aligns with your intended decision and claims in the specific context of use.
COA vs. biomarker examples
PRO (Patient-reported outcome)
The patient reports directly
Example: A patient uses a daily smartphone-based questionnaire or interactive voice prompt to report their pain severity.
ObsRO (Observer-reported outcome)
Non-clinician caregiver/observer reports observable signs/behaviors
Example: A caregiver uses a tablet app to document the frequency and duration of a child’s observable agitation episodes.
ClinRO (Clinician-reported outcome)
Clinician observation + clinical judgment
Example: A clinician conducts an assessment (e.g., motor function) during a telehealth visit, recording their scoring and judgment directly into a digital electronic data capture (EDC) system.
PerfO (Performance Outcome)
Patient completes standardized task(s) per instructions; an sDHT may measure/score the task
Example: A patient performs a prescribed finger-tapping task using a smartphone’s sensor, which automatically scores the speed and consistency.
Gait and mobility
Measurements like gait variability, stride velocity, or freezing of gait episodes detected during daily life
sDHT: Wearable sensor (e.g., wrist-worn accelerometer, patch-worn sensor) or smartphone motion sensors. Used to indicate disease progression (e.g., in Parkinson’s or Huntington’s Disease) or risk of falls.
Speech and voice
Acoustic features (e.g., jitter, pause ratios, pitch consistency) or specific vocal characteristics
sDHT: Smartphone microphone/app or a digital recorder. Used as indicators of pathophysiology (e.g., Parkinson’s disease, mental fitness, or suicidality) or a pharmacodynamic response.
Heart rhythm and rate variability
Irregular pulse rate, episodes of atrial fibrillation (AFib), or Heart Rate Variability (HRV)
sDHT: Smartwatch or wearable ECG patch. Used to diagnose or monitor cardiovascular conditions, or to assess physiological/stress responses.
Keystroke dynamics/tapping
Tapping, swiping, or keystroke latency variability
sDHT: Smartphone or tablet application capturing active user interaction. Used to indicate neural processes or cognitive processing speed.
What FDA guidance tell us
FDA SPOTLIGHT
The Patient-Focused Drug Development (PFDD) Guidance Series are primarily directed at the development of Clinical Outcome Assessments (COAs)—patient-reported, observer-reported, clinician-reported, and performance outcome measures. These assessments are used to capture aspects of “how a patient feels, functions, or survives as a result of treatment”, as described in Guidance 3. Because COAs provide direct or indirect evidence of clinical benefit that is meaningful to patients, they require robust evidence of patient input during their development, including insight into what outcomes are important to patients.
What About Biomarkers?
Biomarkers—such as laboratory values, imaging findings, or physiological signals—are not the primary focus of the PFDD guidance. Unlike COAs, biomarkers typically reflect biological processes or disease states and are often used as indirect or surrogate measures of clinical benefit. As such, they are generally assessed through analytical and clinical validation processes, rather than through direct input from patients regarding their meaningfulness.
However, when a biomarker is proposed as a surrogate endpoint to support regulatory approval—such as in the Accelerated Approval pathway—the FDA looks for scientific evidence that it can reliably predict a clinical benefit that is meaningful to patients (e.g., improved survival or symptom relief). In these cases, the requirement centers on scientific justification, rather than direct patient-reported evidence of meaningfulness.
While the PFDD guidances don’t mandate patient involvement in biomarker qualification or selection, patient input may still be valuable in early development. Patients can help identify which disease-related changes are most impactful, which may in turn influence the relevance and selection of certain biomarkers. FDA increasingly encourages integration of the patient voice across all drug development stages, even when not required by specific guidances.
Next steps
If your endpoint is classified as a biomarker:
Anchor your approach in the FDA–NIH BEST glossary’s biomarker definitions and categories.
For biomarkers, evidentiary expectations focus on two pillars: analytical validation and clinical validation (showing that the biomarker meaningfully reflects disease biology, progression, or treatment response). Some teams may choose to pursue FDA’s Biomarker Qualification Program (see below), but this is not the default path for most digital biomarkers. Even outside the formal program, these evidentiary domains remain essential to ensure regulators view the biomarker as fit-for-purpose in a clinical trial context.
Biomarker Qualification Program
The traditional process of evaluating biomarkers within the context of a single drug development program is inefficient and creates uncertainty for sponsors. This case-by-case approach leads to redundant efforts, slows down the development of novel therapies, and hinders the broad adoption of promising scientific tools. There is a clear need for a centralized, collaborative pathway to formally validate biomarkers, which can de-risk drug development, encourage innovation, and make the process more predictable and cost-effective for all stakeholders.
Recommendations
Drug developers, academic researchers, and other stakeholders should proactively engage with the FDA through the formal Biomarker Qualification Program to validate biomarkers for specific contexts of use. It is recommended to form public-private partnerships and other collaborations to pool resources and data, which strengthens the evidence package for a biomarker’s utility. Developers should use the qualification process to establish a biomarker’s value early, making it a publicly available and reliable tool that can accelerate the development of multiple drug products.
Regulatory Considerations
The Biomarker Qualification Program provides a distinct regulatory pathway for establishing a biomarker’s validity for a specific Context of Use (COU), separate from an individual Investigational New Drug (IND) or New Drug Application (NDA). The process involves a three-stage submission and review cycle: the Letter of Intent, the Qualification Plan, and the Full Qualification Package. Once qualified, a biomarker is publicly listed and can be incorporated into multiple drug development programs without the need for sponsors to re-submit and re-justify the validation data for that specific COU, streamlining subsequent regulatory reviews.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.
If your endpoint is classified as a Clinical Outcome Assessment (COA):
Look to FDA’s Patient-Focused Drug Development Guidances 3 and 4 (see below), which describe expectations for selecting, developing, and incorporating COAs into regulatory decision-making.
For COAs, evidence must demonstrate a clear line of sight from patient input to the outcome assessed. This includes documenting how meaningful aspects of health (MAH) informed the endpoint, defining a minimal clinically important difference (MCID) that reflects meaningful change (discussed below!), and ensuring traceability throughout the process. These inputs should then feed into your V3+ plans, especially clinical validation and usability validation, to show that the measure is both scientifically rigorous and meaningful to patients.
Patient-Focused Drug Development: Selecting, Developing, or Modifying Fit-for-Purpose Clinical Outcome Assessments
The guidance applies to four types of Clinical Outcome Assessments (COAs): Patient-Reported Outcomes (PROs), Observer-Reported Outcomes (ObsROs), Clinician-Reported Outcomes (ClinROs), and Performance Outcomes (PerfOs). A COA is considered fit-for-purpose when the validation evidence is sufficient to support its context of use (COU). To determine if a COA is fit-for-purpose, sponsors must clearly describe the Concept of Interest (COI) and the COU, and present sufficient evidence to support a clear rationale for the COA’s proposed interpretation and use. The rationale for using a COA should include up to eight components, such as justification for the COA type, capturing the important parts of the COI, appropriate administration and scoring, minimal influence from irrelevant factors or measurement error, and correspondence with the Meaningful Aspect of Health (MAH). The most direct assessment of how a patient feels or functions (MAH) should be used as the COI whenever possible.
Recommendations
Sponsors should use the Roadmap to Patient-Focused Outcome Measurement to guide the selection, modification, or development of a COA. The process begins with understanding the disease/condition (including patient perspectives) and conceptualizing clinical benefits and risks (defining the MAH, COI, and COU). When feasible, existing COAs are generally preferred, especially for well-established COIs, as this approach is often the least burdensome. If an existing COA is modified or used in a different context, additional evidence (e.g., cognitive interviews, psychometric studies) must be collected to justify its fitness for the new context of use. For new COA development, sponsors should involve patients, document all steps, and generally avoid using the new COA for the first time in a registration (pivotal) trial; a standalone observational study or early phase trial is recommended for evaluation.
Regulatory Considerations
Sponsors are encouraged to interact early and throughout medical product development with the relevant FDA review division to ensure COAs are appropriate for the intended COU. Sponsors should communicate their proposed COA-based endpoint approach, including the MAH, COI, COA type/name/score, and the final COA-based endpoint, ideally using the suggested format. The type and amount of evidence required to support the rationale for a COA’s use is weighed against the degree of uncertainty regarding that part of the rationale. For ClinROs, it is recommended to use an assessor masked to treatment assignment and study visit for primary endpoints, if feasible. FDA strongly discourages proxy-reported measures for concepts known only to the patient (e.g., pain) and recommends using an ObsRO to measure observable behaviors instead when the patient cannot self-report.
Recommendations
Clearly define the concept of interest and its context of use to ensure COAs align with trial objectives.
Use conceptual and measurement frameworks to communicate how COAs measure patient experiences and generate interpretable scores.
Leverage existing COAs where possible, modifying them only when justified, and document all modifications rigorously.
Ensure COAs are accessible and inclusive, incorporating features like large fonts, touch interfaces, or audio assistance for diverse populations.
Conduct early engagement with FDA to discuss COA selection, development, and validation plans.
Regulatory Considerations
Fit-for-purpose validation requires evidence of conceptual alignment, scoring reliability, and sensitivity to clinically meaningful changes.
Digital health technologies used for COAs must comply with FDA’s guidance on data integrity, usability, and technical performance.
COAs intended for regulatory submissions must be developed and validated before pivotal trials to avoid jeopardizing trial outcomes.
Modifications to COAs or scoring methods during trials necessitate justification and revalidation.
Sponsors should submit comprehensive documentation on COA development, including scoring algorithms and item tracking matrices.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.
Patient-Focused Drug Development: Incorporating Clinical Outcome Assessments Into Endpoints for Regulatory Decision-Making
COA-based endpoints should reflect meaningful patient health aspects and support clear treatment effect inferences.
Selection of endpoints requires a well-supported rationale, including evidence of their importance to patients.
Use of MSD and MSR approaches enhances the interpretation of treatment effects by linking COA scores to meaningful patient experiences. Proper anchors (e.g., global impression of severity) are essential for validating these approaches.
Frequency and timing of COA data collection must align with disease characteristics and study objectives.
Adjustments for potential practice effects and assistive device use are critical for robust outcome measurement.
Proper handling of missing data and sensitivity analyses ensure valid conclusions from COA-based endpoints.
Continuous, ordinal, and dichotomized endpoints require tailored statistical methods for analysis.
Early engagement with the FDA is crucial for aligning study designs and COA approaches with regulatory expectations.
Recommendations
Engage patients and caregivers early to identify meaningful endpoints and assess potential barriers to COA use.
Use anchor-based methods to validate COA scores and define meaningful thresholds for interpretation.
Develop and pilot test study protocols to ensure COA reliability, usability, and alignment with regulatory requirements.
Implement strategies to reduce participant burden, such as concise COA instruments and patient-friendly data collection methods.
Submit comprehensive documentation, including endpoint justification and scoring rationale, to FDA for feedback before trial initiation.
Regulatory Considerations
Endpoints must be supported by evidence of their fit-for-purpose status and alignment with the trial’s objectives.
COAs used in digital or adaptive formats must meet FDA’s standards for usability and data integrity.
Trials with nonrandomized designs require robust measures to mitigate bias in COA-based endpoint analysis.
Thresholds for MSD or MSR must be prespecified and justified with empirical evidence.
Sponsors must follow FDA guidance for submitting COA-based data, ensuring compliance with electronic data standards.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.
Clinical meaningfulness
Clinical meaningfulness ensures the changes your endpoint detects actually matter to patients and the study.
A digital endpoint isn’t fit-for-purpose unless it reflects a change that’s meaningful to patients. Estimating an MCID helps ensure your endpoint has clinical relevance and statistical power — and is a key requirement for both adopters and regulators
Additionally, endpoints need to be interpretable by clinicians. Aligning clinical expert input with patient-derived priorities ensures that the selected endpoint is both patient-relevant and medically credible. Clinicians can offer insight into disease mechanisms, symptom trajectories, and feasible interventions—contextualizing what change is not just meaningful, but also measurable and clinically interpretable. This alignment strengthens the justification for endpoint selection and helps define a realistic MCID.
FDA’s Jeffrey Seigel discusses clinical meaningfulness
Digital Measurement of Nocturnal Scratch: New Developments (June 18, 2024)
As you engage clinicians during stakeholder alignment (see Section 2.3: Stakeholder alignment), be sure to gather their input on what constitutes a meaningful change. Their insights will be essential for defining a clinically credible MCID and building the endpoint rationale.
Methods for estimating a minimal clinically important difference (MCID)
| Approach | What it does | When to use |
|---|---|---|
| Anchor-based* | Interprets change in the measure relative to an external, interpretable reference (e.g., patient or clinician global ratings) | Preferred when anchor data are available (see Regulatory spotlight below) |
| Distribution-based | Uses statistical benchmarks (e.g., 0.5 SD, standard error, effect size) | Useful in early development or when anchors are not feasible |
| Hybrid | Combines both methods to triangulate a stronger estimate | Recommended where possible for regulatory robustness |
* An anchor is an independent external measure of change that is already understood and interpretable, and that reflects a meaningful change from the patient or clinician perspective. In anchor-based approaches, changes in a digital or clinical measure are interpreted relative to this reference point (e.g., a patient global impression of change, a clinician rating, or another established outcome). Anchors help translate numerical change into evidence of meaningful benefit. To learn more about anchors, see Devji, Tahira, et al. “Evaluating the credibility of anchor based estimates of minimal important differences for patient reported outcomes: instrument development and reliability study.” Bmj 369 (2020).
When to engage biostatisticians
Engaging biostatisticians (and, if relevant, COA specialists) at key touchpoints ensures that assumptions about MCID are scientifically credible, statistically defensible, and operationally feasible. Key collaboration points include:
- During endpoint selection: to align MCID assumptions with trial objectives and effect size estimates.
- Before or during pilot studies: to design strategies for estimating MCID, set data anchors, and assess missingness tolerability. Biostatisticians can also advise on design features to proactively reduce missingness. Data missingness should be proactively mitigated through best practices for patient engagement, sDHT selection, and trial design.
- When drafting the SAP: to ensure MCID justifications align with power calculations and the planned analytic role of the endpoint.
According to FDA’s PFDD Guidance 4, Draft, 2023 (FDA) Section II.A.1, sponsors should include “support for the importance of the endpoint to patients and/or caregivers from literature review and/or primary data collection,” and, per Section III.B.1, may consider using anchor-based methods to justify meaningful score differences and interpret the treatment benefit.
Patient-Focused Drug Development: Incorporating Clinical Outcome Assessments Into Endpoints for Regulatory Decision-Making
COA-based endpoints should reflect meaningful patient health aspects and support clear treatment effect inferences.
Selection of endpoints requires a well-supported rationale, including evidence of their importance to patients.
Use of MSD and MSR approaches enhances the interpretation of treatment effects by linking COA scores to meaningful patient experiences. Proper anchors (e.g., global impression of severity) are essential for validating these approaches.
Frequency and timing of COA data collection must align with disease characteristics and study objectives.
Adjustments for potential practice effects and assistive device use are critical for robust outcome measurement.
Proper handling of missing data and sensitivity analyses ensure valid conclusions from COA-based endpoints.
Continuous, ordinal, and dichotomized endpoints require tailored statistical methods for analysis.
Early engagement with the FDA is crucial for aligning study designs and COA approaches with regulatory expectations.
Recommendations
Engage patients and caregivers early to identify meaningful endpoints and assess potential barriers to COA use.
Use anchor-based methods to validate COA scores and define meaningful thresholds for interpretation.
Develop and pilot test study protocols to ensure COA reliability, usability, and alignment with regulatory requirements.
Implement strategies to reduce participant burden, such as concise COA instruments and patient-friendly data collection methods.
Submit comprehensive documentation, including endpoint justification and scoring rationale, to FDA for feedback before trial initiation.
Regulatory Considerations
Endpoints must be supported by evidence of their fit-for-purpose status and alignment with the trial’s objectives.
COAs used in digital or adaptive formats must meet FDA’s standards for usability and data integrity.
Trials with nonrandomized designs require robust measures to mitigate bias in COA-based endpoint analysis.
Thresholds for MSD or MSR must be prespecified and justified with empirical evidence.
Sponsors must follow FDA guidance for submitting COA-based data, ensuring compliance with electronic data standards.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.
exampleS
MCID, sDHTS, and Alzheimer’s disease: Regulatory and implementation perspectives
Assessing clinical meaningfulness in clinical trials for Alzheimer’s disease: A U.S. regulatory perspective
In a progressive neurodegenerative illness like Alzheimer’s disease, slowing the rate of disease progression is considered a clinically meaningful outcome for patients and their caregivers.
The assessment of what constitutes a clinical benefit is highly dependent on the specific stage of AD being studied, the drug’s mechanism of action, and the symptoms present in that patient population.
Direct input from patients and caregivers is critical for understanding disease burden and defining treatment benefits that are truly meaningful from their perspective.
The interpretation of score changes on Clinical Outcome Assessments (COAs) requires full context; an absolute point difference must be considered relative to the study’s duration, the expected placebo decline, and the specific disease stage.
Evidence from biomarkers that show an effect on underlying disease pathology provides additional support and increases the persuasiveness of the changes observed on clinical endpoints.
Recommendations
Drug developers should implement multiple “fit-for-purpose” COAs that use different reporters (e.g., clinicians, observers) and methods to generate broad and diverse evidence of a drug’s clinical benefit.
Sponsors should utilize both qualitative and quantitative methodologies to explore clinical meaningfulness, including assessing “meaningful within-patient change” throughout the development process.
Developers are encouraged to create and validate new COAs and leverage innovative approaches, such as digital health technologies, to better capture concepts that are relevant to patients, especially in the earliest stages of AD.
Throughout the drug development lifecycle, stakeholders should systematically collect and incorporate patient experience data to ensure that the perspectives, needs, and priorities of patients are meaningfully captured.
Regulatory Considerations
For a drug to gain approval, it must meet the regulatory standard of “substantial evidence of effectiveness,” which is typically derived from adequate and well-controlled investigations designed to minimize bias.
The FDA defines clinical benefit as a clinically meaningful effect of a drug on how an individual feels, functions, or survives.
An assessment of clinical benefit is not limited to the primary endpoint; the consistency of findings across multiple endpoints (primary and secondary) is a key consideration during regulatory review.
The accelerated approval pathway may be used for serious conditions with unmet needs based on a surrogate endpoint, but traditional approval requires verification of clinical benefit in confirmatory trials.
The FDA’s evaluation includes a benefit-risk analysis, which considers the severity of the disease and the availability of alternative therapies, recognizing that patients and physicians may accept greater risks for life-threatening illnesses.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.
FDA neurosciences reviewers outline practical ways sponsors can demonstrate that trial endpoints truly reflect benefits that matter to patients and caregivers for individuals with Alzheimer’s disease. They emphasize:
- Tying within-patient meaningful change thresholds to patient experience data.
- Selecting stage-appropriate COAs and combining cognition + function to show benefit.
- Leveraging innovative COAs and biomarker or surrogate endpoints when they are “reasonably likely” to predict benefit.
DiMe Digital Measures projects
Defining a Context of Use and identifying a Meaningful Aspect of Health is a rigorous process that requires months of stakeholder alignment. If your research involves Sleep, Physical Activity, Nocturnal Scratch, or Alzheimer’s and Related Dementias (ADRD), much of this foundational work has already been done for you.
The DiMe Digital Core Measures projects provide pre-competitive, expert-vetted blueprints that allow you to skip the “blank page” stage. These expert-led core digital measures projects (worked examples for MAH to COI to digital endpoint) include ontologies, conceptual models, best practices, and other resources.

Core Digital Measures of Alzheimer’s disease and related dementias
Digital health measures for ADRD must align with patient and care partner priorities, including functional daily activities such as remembering object locations and maintaining speech fluency.
Sensor placement, data collection modalities, and algorithmic interpretation significantly impact the accuracy and reliability of digital measures.
While digital cognitive and behavioral assessments have strong potential as clinical endpoints, standardization is needed to ensure regulatory acceptance.
Sleep and mobility disruptions in ADRD can be measured with actigraphy, EEG, and ambient sensor-based approaches, but usability considerations are crucial.
Metadata, including environmental conditions and patient comorbidities, must be accounted for to ensure valid interpretations of digital measures in both research and clinical practice.
Recommendations
Researchers and technology developers should adopt standardized ontologies for digital measures to improve consistency across studies and regulatory submissions.
Digital biomarkers should be selected and validated with reference to patient and care partner needs, ensuring they reflect meaningful aspects of health.
Considerations such as sensor placement, data processing methods, and cultural neutrality of cognitive assessments must be accounted for in study designs.
Clinical trials should incorporate digital health technologies as both exploratory endpoints and potential screening tools for ADRD progression.
Further research is needed to refine algorithms for sleep, mobility, and speech-based digital biomarkers to enhance their predictive power for cognitive decline.
Regulatory Considerations
Digital measures of sleep and mobility have been recognized as potential clinical trial endpoints by regulatory agencies such as the FDA.
Standardized reporting and frameworks should be followed to ensure interoperability and data integrity in digital health studies.
Developers must document and validate scoring algorithms used for cognitive and behavioral assessments to meet regulatory expectations.
Data privacy and security regulations must be adhered to, particularly when collecting real-world behavioral and biometric data.
Ongoing validation and real-world evidence generation are necessary to establish digital measures as reliable clinical and regulatory endpoints in ADRD research.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.

Core Digital Measures of Nocturnal Scratch
Nocturnal scratch is a clinically relevant behavior that impacts sleep quality, skin integrity, and overall disease burden in conditions like AD.
Traditional clinical outcome assessments (COAs) often fail to adequately measure scratching behavior, making digital measurement an important complement.
Digital health technologies, including wearables and sensor-based monitoring, enable passive and objective measurement of scratch behavior without relying on patient recall.
Regulatory agencies emphasize the importance of validation, ensuring digital measures are fit-for-purpose and aligned with patient needs.
Privacy, security, and compliance considerations must be prioritized, particularly in decentralized clinical trials using real-world data collection methods.
Recommendations
Digital measurement of nocturnal scratch should be integrated as an endpoint in clinical trials to capture patient-relevant outcomes objectively.
Sensor-based tools must undergo validation processes, including analytical and clinical validation, to ensure accuracy and reliability in different populations.
Stakeholders should align terminology and measurement definitions to support consistency across studies and regulatory submissions.
Usability testing with patients is critical to ensuring that wearable devices are practical and minimally burdensome.
Clinical trials should incorporate data privacy protections and clear informed consent processes to safeguard patient information.
Regulatory Considerations
FDA encourages early engagement to discuss digital endpoints, particularly through the Critical Path Innovation Meeting (CPIM) process.
Digital tools used for clinical investigations should align with 21 CFR Part 11 compliance for electronic records and data integrity.
Sponsors should ensure that digital health technologies used in trials meet validation criteria, including fit-for-purpose assessment and clinical relevance.
Privacy regulations, including GDPR and HIPAA, must be considered when handling patient data collected via wearable sensors.
Post-market monitoring and long-term validation studies are recommended to ensure continued accuracy and reliability of nocturnal scratch measurements.
Open source: Core Digital Measures of Nocturnal Scratch
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.

Core Digital Measures of Sleep
Sleep disturbances are common across multiple therapeutic areas, making standardized digital measures essential for cross-condition research.
Measurement accuracy varies depending on sensor placement, algorithms, and contextual factors such as sleep environment.
While home-based digital sleep tracking improves accessibility, challenges remain in ensuring consistency with clinical polysomnography.
Digital measures of sleep provide new opportunities for continuous and longitudinal monitoring, but standardization in data collection and interpretation is needed.
Stakeholders, including regulatory agencies, increasingly recognize digital sleep biomarkers, but additional validation is required to ensure widespread adoption.
Recommendations
Researchers and clinicians should integrate core digital sleep measures into study designs to improve data comparability across trials and clinical contexts.
Algorithm transparency and validation protocols should be established to enhance the accuracy of digital sleep monitoring tools.
Regulatory engagement should be prioritized early in the development process to ensure that digital sleep measures meet evidentiary standards.
Multi-stakeholder collaboration, including patient and care partner input, is essential to ensure sleep measures reflect meaningful aspects of health.
Further research is needed to refine wearable and sensor-based technologies to improve real-world applicability and clinical utility of digital sleep biomarkers.
Regulatory Considerations
The FDA and other regulatory bodies increasingly acknowledge sleep measures as potential clinical endpoints, but clear validation frameworks are necessary.
Digital sleep measures should align with industry standards such as HL7 to ensure interoperability and data integrity.
Data privacy and security regulations must be followed, particularly for continuous sleep monitoring in real-world settings.
Post-market validation and real-world evidence generation are critical to support regulatory acceptance of digital sleep biomarkers.
Developers must document the derivation of sleep measures, including algorithmic processing and sensor accuracy, to meet regulatory review requirements.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.

Core Digital Measures of Physical Activity
Measurement variability arises from different wearable sensor placements, algorithms, and environmental contexts.
Standardized ontologies are needed to ensure consistency in physical activity measurement across digital health studies.
Regulatory agencies, including the FDA, have endorsed specific digital measures such as MVPA as clinical trial endpoints.
Advances in sensor technology and data analysis have improved the feasibility of measuring real-world physical activity with high accuracy.
Additional validation efforts are required for postural sway measures, as current technologies primarily rely on force plates and laboratory-based assessments.
Recommendations
Researchers and developers should adopt standardized ontologies to enhance the comparability of digital measures in clinical research.
Sensor placement and algorithm transparency must be considered to minimize measurement variability in digital endpoints.
Stakeholders should engage with regulatory bodies early to ensure that digital biomarkers meet evidentiary requirements for clinical trials.
Digital health technology developers should prioritize usability and patient-centered design to increase adoption and adherence.
Further research is needed to expand real-world applicability and validation of postural sway measures for clinical and therapeutic use.
Regulatory Considerations
FDA has recognized certain digital measures, such as time spent in MVPA, as valid clinical trial endpoints.
Digital measures used in clinical research should align with HL7 and industry standards for interoperability and data integrity.
Transparency in data processing, including raw data versus processed metrics, is essential for regulatory acceptance.
Developers must ensure compliance with data privacy regulations when collecting real-world physical activity data.
Post-market monitoring of digital endpoints is recommended to ensure continued accuracy and reliability in diverse patient populations.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.

Digital Measures: De-risking Cytokine Release Syndrome (CRS)
Cytokine Release Syndrome (CRS) is a common and potentially life-threatening adverse event of immunotherapies, particularly in Oncology, complicating patient care and increasing healthcare costs. Standard-of-care inpatient monitoring for CRS is manual, intermittent, costly, and restrictive, providing an incomplete view of the syndrome’s development and progression. The use of Digital Health Technologies (DHTs) for continuous, remote monitoring of vital signs (like heart rate, respiratory rate, skin temperature, SpO2, and activity) can capture early indicators of CRS up to two hours earlier than standard episodic monitoring. This ability to collect multivariate continuous data is valuable for informing robust model development for CRS risk prediction.
Recommendations
Investigators should deploy DHTs available today to monitor vital signs and symptoms currently observed in the hospital setting, but in an outpatient or home environment. The goal is to develop Early Warning Products that assess the probability of developing CRS, providing clinical decision support. Product developers should follow a strategic roadmap that outlines milestones for building products that are clinically relevant and commercially viable. Researchers should use a common set of digital clinical measures to gather high-quality datasets and ensure comparability across studies to build more robust predictive models. Predictive algorithms should be built on a robust reference measure for analytical validation and be clinically validated with sufficient data.
Regulatory Considerations
The resources are designed to help developers build products that are clinically appropriate, regulatory-acceptable, and commercially viable. Future regulatory submissions for CRS de-risking products will benefit from aligning with this industry-wide dialogue that is being built in collaboration with the FDA. Developing a robust CRS safety biomarker could enhance the safety profile of clinical trials, increase trial access, and streamline regulatory decision-making, possibly through a qualification pathway. Products that aim for a higher level of autonomy, such as a Diagnostic that redefines current CRS grading classes, may require very high clinical evidence and likely stringent regulatory review.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.

Advancing the use of sensor-based digital health technologies (sDHTs) for mental health research and clinical practice
The most promising aspects of mental health for digital measurement are sleep, physical activity, stress, and social behavior, which have the strongest scientific evidence. Core barriers to adoption include high cost and limited access, data privacy concerns, poor technological literacy, and a lack of technology adaptation for specific mental health needs. Essential technology characteristics for “fit-for-purpose” sDHTs include usability, reliable performance, strong data privacy and security, and long battery life.
Recommendations
Research and development should prioritize moving promising measures (sleep, activity, stress, social behavior) to large-scale clinical trials. Algorithms must be refined and clinically validated for mental health indications, and new sensor modalities should be explored. Infrastructure must be developed by creating standards and ontologies for mental health sensor data to ensure interoperability and scalability. To improve access and equity, financial support mechanisms and inclusive, culturally tailored design are critical.
Regulatory Considerations
The report does not provide a separate section for “Regulatory Considerations” but emphasizes that future development and funding should prioritize clinical validation across diverse populations. It notes the importance of a clear understanding of the intended measurement claims and the need for rigorous validation studies to move beyond pilot and feasibility stages to demonstrate real-world clinical utility.
Some summaries are generated with the help of a large language model; always view the linked primary source of a resource you are interested in.
KEY TAKEAWAYS
If there is lingering ambiguity about the COU or how an endpoint should be classified, scheduling a short early touchpoint with FDA can provide practical guidance on plausibility and evidentiary priorities. This step can be especially valuable before committing resources to study design or contracting (see Section 3: Engage regulators). Your endpoint’s COU will shape your validation needs across all four domains of V3+: verification, analytical, clinical, and usability (see Section 4: Your validation strategy).
Define a clear context of use for your particular use case.
Prepare an MCID justification memo with clinician input.
Draft a working conceptual model, which is a visual map that links patient-meaningful aspects of health to concepts of interest and clearly defined digital endpoints.
Draft the endpoint rationale for any candidate digital endpoints for inclusion in briefing documents, statistical analysis plan, etc.
2.2 Context of use
Library resources to guide you
The sDHT roadmap library gathers 200+ external resources to support the adoption of sensor-based digital health technologies. To help you apply the concepts in this section, we’ve curated specific spotlights that provide direct access to critical guidance and real-world examples, helping you move from strategy to implementation.
Features essential guidance, publications, and communications from regulatory bodies relevant to this section. Use these resources to inform your regulatory strategy and ensure compliance.
Gathers real-world examples, case studies, best practices, and lessons learned from peers and leaders in the field relevant to this section. Use these insights to accelerate your work and avoid common pitfalls.