90 Canal St., 4th Floor, Boston, MA 02214 | +1-765-234-3463 | www.dimesociety.org
March 19, 2022
Dockets Management Staff (HFA-305)
Food and Drug Administration
5630 Fishers Lane, Rm. 1061
Rockville, MD 20852
To Whom it May Concern:
This letter represents comments submitted on behalf of the Digital Medicine Society
(DiMe) for consideration by the U.S. Food and Drug Administration (FDA) regarding Docket
No. FDA-2021-D-1128 for “Digital Health Technologies for Remote Data Acquisition in
Clinical Investigations Draft Guidance for Industry, Investigators, and Other Stakeholders.”
DiMe is a 501(c)(3) non-profit organization dedicated to advancing digital medicine to
optimize human health. We do this by serving professionals at the intersection of the global
healthcare and technology communities, supporting them in developing digital medicine
through interdisciplinary collaboration, research, teaching, and the promotion of best
practices.
Founded in 2019, DiMe is the first professional organization for experts from all disciplines
comprising the diverse field of digital medicine. Together, we drive scientific progress and
broad acceptance of digital medicine to enhance public health.
Many of our DiMe members conduct clinical trials of new medical products or develop digital
health technologies for use in these trials. The following comment leverages the combined
expertise of our members regarding the use of digital health technologies (DHTs) in clinical
trials. We appreciate the opportunity to offer our comments on this draft guidance
document.
We appreciate that this guidance was issued by multiple centers: Oncology Center of
Excellence (OCE), The Center for Devices and Radiological Health (CDRH), the Center for
Drug Evaluation and Research (CDER), and the Center for Biologics Evaluation and Research
(CBER). In particular, we are pleased to see there is a continuing trend in harmonizing
guidance and hope this continues. Reduction in gray areas for those developing digital
health technologies and those using them in clinical investigations will allow regulations to
be more approachable and followable, lessening the burden for all involved.
We applaud FDAs human-centered approach to this guidance. Recognizing that patients
should be the beneficiary of the work done in drug, tool, and measure development, digital
or otherwise, is critical to identifying problems and developing solutions that matter.
We note that the scope of this guidance allows for generalized use of best practices across
technologies and appreciate FDA providing that flexibility.
Table of contents
Comment Structure: Feedback and Resources From the Digital Medicine Community 3
Definitions of and Surrounding Digital Health Technology 3
Comments on Regulatory Considerations and Engagement with the Agency 4
Comments on Considerations When Using Digital Health Technologies in Clinical
Investigations 4
Conclusion 10
Comment Structure: Feedback and Resources
From the Digital Medicine Community
Our comment is structured around feedback and questions that we have received from the
digital medicine community. We do not presume to provide answers, that is the sole domain
of the Agency. Rather, following each question we pose in this comment, we highlight
resources that 1) the digital medicine community is currently referencing, and 2) that we
believe are of high quality.
Definitions of and Surrounding Digital Health
Technology
Lines 15-18 and 813-818 of the Glossary define digital health technology (DHT) as a “system
that uses computing platforms, connectivity, software, and/or sensors, for healthcare and
related uses.” We recognize the complexity of DHTs and appreciate the broad definition and
2
recommend the further inclusion of language that references a modular stack of hardware
and software components
1
in this definition.
Line 23 references hardware and/or software that may comprise a DHT, pointing to a
footnote defining the two terms, as well as firmware. There is opportunity to clarify these
definitions to remove ambiguity.
FDA might consider aligning to definitions provided by ISO/IEEE:
Hardware - an electronic or mechanical element, including its interface and
documentation
2
Software - computer programs, procedures and possibly associated documentation
and data pertaining to the operation of a computer system
3
Firmware - combination of a hardware device and computer instructions or computer
data that reside as read-only software on the hardware device
4
It is worth noting that ISO/IEEE considers the firmware to be a part of the software, as
opposed to the guidance, which includes the firmware in the hardware. Aligning to ISO/IEEE
standards may reduce the burden on manufacturers of DHTs as they work to comply with
regulations.
FDA might also consider the addition of language regarding the core operation of the
hardware and the specific role of the firmware, including the appropriate regulations for
each.
Comments on Regulatory Considerations and
Engagement with the Agency
The inclusion of language regarding the optional qualification regulatory path is welcome
guidance. It provides clarity for the device manufacturer and sponsor. Additional information
provided in the FDAs webinar covering this draft guidance made a clarifying note that
qualification is advantageous when using the same DHT in multiple clinical trials. We
recommend adding this additional language to the guidance as it creates clarity for
following the qualification path.
4
ISO/IEC 12207:2008 Systems and software engineering — Software life cycle processes, 4.14
3
IEEE 828-2012 IEEE Standard for Configuration Management in Systems and Software Engineering, 2.1
2
IEEE 1012-2012 IEEE Standard for System and Software Verification and Validation, 3.2
1
Coravos A, Doerr M, Goldsack J, Manta C, Shervey M, Woods B, William A. Wood WA. Modernizing and designing evaluation
frameworks for connected sensor technologies in medicine. npj Digital Medicine (2020) 3:37 ;
https://www.nature.com/articles/s41746-020-0237-3
3
Lines 110-112 describe devices used in clinical investigations as “exempt from most
requirements applicable to devices.” We request additional clarification on this
terminology, who is able to make this distinction, and through what mechanism this
distinction should be pursued. Specifically, whether a medical device clearance is preferred
or irrelevant to the agency would assist our community.
There is an opportunity to define the boundaries between exempt DHTs, DDT/MDDT,
SaMD, and other qualification programs, including specifics across different centers.
Clarifying these boundaries could allow for harmony across guidances and centers to
produce a guidance library that supports digital health innovation through a shared unifying
language and understanding of processes.
Comments on Considerations When Using Digital
Health Technologies in Clinical Investigations
We recognize that it is challenging to create one guidance that meets the needs of a
diverse audience of readers (manufacturers, sponsors, quality teams, etc.) and appreciate
the care taken to be as broad as possible in this draft guidance.
The DiMe Society and our community have developed relevant resources, such as The
Playbook
5
that builds a shared foundation for developing and deploying digital clinical
measures using a step-wise approach and Digital Measures that Matter to Patients: A
Framework to Guide the Selection and Development of Digital Measures of Health
6
that
may be useful to FDA.
The Playbook’s step-wise approach for developing and deploying clinical measures
5
6
Manta C, Patrick-Lake B, Goldsack J, C. Digital Measures That Matter to Patients: A Framework to Guide the Selection and
Development of Digital Measures of Health. Digit Biomark 2020;4:69-77. https://www.karger.com/Article/Fulltext/509725#
5
https://playbook.dimesociety.org/
4
Through our and our collective digital medicine community’s experiences, we deem
measure development as a precursor to technology selection. We recognize the
discussion of endpoints in Section IV. D and recommend the addition of a section describing
measure development (inclusive of Section IV. D), prior to Section IV. A. Selection of a Digital
Health Technology and Rationale for use in a Clinical Investigation.
Measures collected with DHTs during clinical investigations may be collected to determine
study eligibility, assess efficacy, safety, and/or adherence to protocol.
Digital measures also vary by the type of underlying software/technology. To evaluate the
quality of the digital measurement product, it is valuable to distinguish between those tools
that rely upon sensor-generated data and those that rely on reported, survey data.
We use the term, “biometric monitoring technology” (BioMeT) to describe measurement
tools that process data captured by mobile sensors using algorithms to generate
measures of behavioral and/or physiological function. We suggest that this may be useful
language for the Agency to adopt in future guidance.
Evaluation of a digital measurement product that collects patient-generated data digitally
should be based on the context of use and should consider the type of technology:
Survey-derived measures (e.g., an ePRO): The evaluation process is already well
described by prior FDA guidelines (e.g., construct, context and content validation)
Sensor-derived measures (e.g., using a wearable): Evaluation should be based on the
verification, analytical validation, and clinical validation (V3)
7
process detailed further
on page 6 of this comment
As the recently released guidance on Patient-Focused Drug Development: Methods to
Identify What Is Important to Patients Guidance for Industry, Food and Drug Administration
Staff, and Other Stakeholders deals largely with identifying meaningful aspects of health
relevant to patients, citing that guidance would be beneficial to the readers.
As related to the selection of the DHT, we echo the fit-for-purpose definition (Line 150-152
and Glossary 824-824) of a conclusion that the level of validation associated with a DHT is
sufficient to support its context of use. Our digital medicine community recognizes the need
for clarification when considering the potential for a mixed technical capability of the
target study population, or at least the need to urge sponsors and investigators to consider
such a perspective when selecting a DHT.
7
Goldsack JC, Coravos A, Bakker JP, Bent B, Dowling AV, Fitzer-Attas C, Godfrey A, Godino JG, Gujar N, Izmailova E, Manta C.
Verification, analytical validation, and clinical validation (V3): the foundation of determining fit-for-purpose for biometric
monitoring technologies (BioMeTs). npj Digital Medicine. 2020 Apr 14;3(1):1-5.
https://www.nature.com/articles/s41746-020-0260-4
5
We appreciated the flexibility in DHT use in clinical investigation during the COVID-19
pandemic and encourage maintaining a similar approach in the future. This flexibility allows
for participants to have choice when enrolling in a trial and can encourage subject
participation and retention.
Lines 257-258 describe the use of technical specifications and descriptions provided by
the DHT manufacturer. While these lines indicate that these manufacturer specifications
and descriptions may be “sufficient,” further guidance is requested to define the exact
materials needed to meet this sufficiency and limit the variety of interpretations possible by
DHT manufacturers and sponsors. Additionally, we suggest the inclusion of clarifying
language on using a DHT in an alternate manner to the DHT manufacturer’s intended use.
In regards to the Verification, Validation, and Usability of Digital Health Technologies, we
champion the efforts to properly evaluate and document a digital measure and the DHTs
used to measure it. Our community has developed extensive, high-quality resources to this
end that have been adopted by nearly 100 companies and the European Medicines Agency.
We offer an overview of the resource here.
The evaluation framework for sensor-derived digital measures should encompass both
the product’s components (e.g., hardware, firmware, and software, including algorithms)
and the intended use of the product. Existing frameworks for new biotechnologies are not
sufficiently adaptable, but they can provide meaningful insight for developing new
evaluation frameworks for BioMeTs.
We propose a three-component framework to unite the different disciplinary experts who
should participate in the foundational evaluation of sensor-derived digital measures. This V3
framework includes (1) verification, (2) analytical validation, and (3) clinical validation.
V3 are foundational to determine whether a digital medicine tool is fit-for-purpose. An
evaluation of the usefulness and utility is only applicable after gaining evidence and
assurance that the underlying data and predictions are “valid” to answer a given question.
The V3 process is summarized in the figure below and described in detail in the manuscript,
“Verification, analytical validation, and clinical validation (V3): the foundation of determining
fit-for-purpose for Biometric Monitoring Technologies (BioMeTs)”. This manuscript was
developed by a multi-disciplinary group of experts with the goal of identifying
considerations for evaluating and documenting measurement performance of
technologies that generate sensor-derived digital measures. We hope it may be a useful
resource for the Agency.
6
Utilizing the definitions shown in the figure below provides increased flexibility to use parts
of the modular stack of a DHT and not others while maintaining necessary rigor.
The stages of the V3 framework for biometric monitoring technologies
8
In addition to the offer of this resource, our digital medicine community requests specifics
about what to include in the verification and validation plan, including the level of evidence
that is acceptable. We recognize that the variations in studies, measures, and technologies
makes for a particularly challenging request. Guidance around the general categories that
should be included in the verification and validation plan, whether there is an expectation to
reference a gold standard, and what makes for a good reference measure (or examples of
such) are, perhaps, specific enough to be actionable. We also would encourage providing
examples of the evidence required when a DHT is software alone and when it is a
software/hardware/firmware combination.
We are pleased that usability is referenced, along with the Applying Human Factors and
Usability Engineering to Medical Devices guidance. We suggest the inclusion of additional
guidance regarding the critical factors that should be included in usability testing for DHTs,
8
Goldsack JC, Coravos A, Bakker JP, Bent B, Dowling AV, Fitzer-Attas C, Godfrey A, Godino JG, Gujar N, Izmailova E, Manta C.
Verification, analytical validation, and clinical validation (V3): the foundation of determining fit-for-purpose for biometric
monitoring technologies (BioMeTs). npj Digital Medicine. 2020 Apr 14;3(1):1-5.
https://www.nature.com/articles/s41746-020-0260-4
7
such as environmental variables on the sensor performance or human biocompatibility of
the hardware.
We appreciate the inclusion of a section focused on Evaluation of Clinical Endpoints From
Data Collected Using Digital Health Technologies. As mentioned above, we recommend this
section to be moved prior to Selection of a Digital Health Technology and Rationale for Use
in a Clinical Investigation and including guidance on determining a meaningful aspect of
health, identification of concept of interest, followed by the development of the digital
endpoint.
We appreciate the approach the Agency takes in the draft guidance and agree that digital
medical product development tools should be no different than non-digital and request
clarification about how we justify use, specifically whether using existing biomarker and
COA guidance is sufficient. Should they be sufficient, a reference to these guidances would
be beneficial.
Perhaps out of scope for this guidance, DiMe’s community notes that efforts to develop new
digital clinical measures should return value over and above existing assessments, for
example reduction of participant burden, higher quality data, cost savings, participant
adherence, new information, and appeal to a more diverse population.
As we explore Risk Considerations When Using Digital Health Technologies, we encourage a
risk-based approach, similar to that used by the FDA in other guidances. for other
endpoints.
We suggest the inclusion of language around risks to equity or, if appropriate, references to
other guidances addressing equity and inclusion. The use of DHTs in clinical investigations is
at high risk of excluding populations based on socio-economic status. DiMe has convened a
Digital Health Measurement Collaborative Community (DATAcc) to develop and
demonstrate best practices and advance harmonized approaches to speed the use of digital
health measurement to improve health outcomes, health economics, and health equity.
9
We
hope the resources developed and pending release are useful to the readers of this
guidance.
Lines 458-486 discuss Statistical Analysis. There is an opportunity to conform the method
of defining endpoints to those of standard industry guidance. We recognize that the
diversity of sensors, algorithms, and methods make guidance difficult, yet realize the need
for specifics on the requirements and optional supporting elements for defining an
endpoint. For instance, defining meaningful change, particularly when access to the patient
population is not available, and indicating whether the change in a referenced traditional
9
http://datacc.dimesociety.org/
8
endpoint could be used as a benchmark would provide valuable insight to the audience of
this guidance.
We recommend the inclusion of guidance related to missing or changing data due to
software upgrades or technical failures within the Statistical Analysis section. Included in
the required statistical analysis plan should be documentation regarding these missing or
changing data, such as missing timestamps, duplicate timestamps (i.e., during daylight
savings), short and long-term gaps in data, and even partially uploaded files due to
technical trouble or connectivity issues.
We are pleased to see specific guidance, such as those around providing instructions to
participants for cleaning the DHT (lines 506-508). Based on our interactions with
individuals who have participated in trials using DHTs, we want to make a note that cleaning
procedures should take into consideration the burden on the trial participants.
Cumbersome, lengthy, or frequent cleaning procedures can result in non-compliance by the
participant, leading to a lack of or altered data collection. DHT manufacturers, sponsors, and
investigators should take this into account and consider evaluating the cleaning protocol as
a part of usability testing.
Lines 515-518 note cybersecurity risks as a part of the Clinical Risks section. FDA may wish
to consider either a separate risks section dedicated to cybersecurity or highlighting the
impact of cybersecurity in each risk category to clarify the extensive reach of
cybersecurity threats.
Lines 536-539, 567-569, and 582-589 discuss important points related to data privacy,
data access, and end-user license agreements. We agree that participants must be made
aware of all entities with access to the data, measures to keep data safe, and that sponsors
and investigators should attempt to work with DHT manufacturers to navigate this process.
We encourage the inclusion of guidance that recommends starting as early as possible
(long before preparing informed consent documents) when working with DHT
manufacturers in understanding the end-user license agreements and their impact on data
privacy and security. We also recommend that relevant highlights from the end-user
license agreements be included in the informed consent document using understandable
language for the participants.
In the Record Protection and Retention section, the guidance states the requirement of
record retention in clinical investigation and emphasizes the need for source data
retention "to reconstruct and evaluate the clinical investigation, and the data should be
available for inspection." There is an opportunity to clarify the definition of source data in
the context of continuous sensor-based DHT. To derive digital measures from continuous
time series of sensor data, a pipeline composed of signal processing and/or machine
9
learning steps is typically deployed to transform the raw sensor data to the discrete data
features so that they can be used as endpoints in statistical models for efficacy and safety
analysis. We believe that the retention of the raw sensor data, or its least processed form,
is essential to the use of sensor-based DHT in clinical investigation. This processing
pipeline, or algorithm, is not currently standardized for most sensor-based DHTs and
continues to iterate to keep pace with the rapidly evolving data science and technology
landscape. Processing pipelines can also be proprietary/black box and change during
firmware and software updates, imposing further challenges to data consistency.
As an example, sensors that use accelerometer data capture gravity (G) changes on multiple
axes. These micro-G measurements are captured many times per second and are the basis
for the resulting endpoints. The micro-G data would be considered the source data, not the
post-processed unit of measurement or calculated endpoint.
There is an opportunity to further clarify the definition of source data in the context of such
sensor-based DHT and recognize the importance of retaining minimally processed data, as
this offers the ability to investigate missing data, improve data consistency, and potentially
re-evaluate the clinical data when more accurate algorithms become available in the future.
Lines 616-624 discuss details around data outputs for the DHT. With a lack of
standardization around how source data is generated, we recommend that data with the
highest granularity, or least amount of processing, should be the default “source.” This
mitigates the most risk when manufacturers are generating data utilizing different
non-standard methods. The capture of the raw data or least processed data allows for all
other aspects of the data processing to be revisited or audited at any time.
In the DHT Updates and Other Changes section, lines 753-755 discuss the assessment of
updates to a DHT to ensure no significant impact on the measurements performed and
data collected. Our digital medicine community requests clarification on evidence needed
around the impact of updates when assessing for consistency. For instance, is
documentation from the software manufacturer stating backward and/or forward
compatibility sufficient or is additional evidence needed? Further clarification regarding the
difference between a manufacturer’s software update for something like a security patch
and an update impacting the use of the DHT for the trial purposes is requested in terms of
the evidence needed to support the “no significant impact” conclusion. Our community
follows the National Academy of Medicine definition of high quality data, which is data that
10
leads to the same conclusion.
10
We wonder if this aligns to your classification of “no
signi
ficant impact” and, if this language might provide clarity to readers.
Lines 762-772 discuss software and operating systems updates. Dedicated solutions are
preferred during active data collection of a DHT. Clear guidance on the types of updates a
manufacturer can push to production will allow sponsors to accept or decline these types of
updates. Certain updates that might be a "feature enhancement" could be opted out by the
sponsor to mitigate risk. While any updates on Security, Data Integrity, and Privacy are
clearly documented and approved by sponsors for "live" clinical trials using a DHT.
Conclusion
We recognize the effort on this draft guidance regarding DHT use in clinical investigations
as an impactful, helpful guidance that instructs and assists DHT manufacturers, sponsors,
investigators, and regulators alike. It is no small feat to provide clarity to a young industry
with a wealth of unknowns. We desire to assist the
field of digital medicine by providing
quality resources, best practices and use cases, and forums for valuable connections that
can drive the field forward productively and with patients always in mind.
We appreciate the efforts to create a broadly applicable guidance that does not overburden
the industry with regulations and we hope our comments are helpful in improving this
guidance. At DiMe, we are committed to the safe, effective, ethical, and equitable use of
digital technologies in clinical investigations and see through this guidance and our
interactions with FDA that the Agency shares this commitment.
Thank you,
Jennifer Goldsack, MChem, MA, MBA, OLY
CEO
Christina Marasco, PhD
Program Lead
10
Institute of Medicine (US) Roundtable on Research and Development of Drugs, Biologics, and Medical Devices; Davis JR,
Nolan VP, Woodcock J, et al., editors. Assuring Data Quality and Validity in Clinical Trials for Regulatory Decision Making:
Workshop Report. Washington (DC): National Academies Press (US); 1999. Executive Summary.
https://www.ncbi.nlm.nih.gov/books/NBK224572/
11