FUTURE-AI: International consensus guideline for trustworthy and deployable artificial intelligence in healthcare (2024)

Karim Lekadirkarim.lekadir@ub.eduUniversitat de BarcelonaArtificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer ScienceBarcelonaSpain,Aasa FeragenTechnical University of DenmarkDTU ComputeKgs LyngbyDenmark,Abdul Joseph FofanahMilton Margai Technical UniversityDepartment of Mathematics and Computer Science, Faculty of Science and TechnologyFreetownSierra Leone,Alejandro F FrangiUniversity of LeedsCentre for Computational Imaging & Simulation Technologies in Biomedicine, Schools of Computing and MedicineLeedsUnited KingdomKU LeuvenMedical Imaging Research Center (MIRC), Cardiovascular Science and Electronic Engineering DepartmentsLeuvenBelgium,Alena BuyxTechnical University of MunichInstitute of History and Ethics in MedicineMunichGermany,Anais EmelieUniversitat de BarcelonaArtificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer ScienceBarcelonaSpain,Andrea LaraGalileo UniversityFaculty of Engineering of Systems, Informatics and Sciences of ComputingGuatemala CityGuatemala,Antonio R PorrasUniversity of Colorado Anschutz Medical CampusDepartment of Biostatistics and Informatics, Colorado School of Public HealthAuroraUnited States,An-Wen ChanUniversity of TorontoDepartment of Medicine, Women’s College Research InstituteTorontoCanada,Arcadi NavarroUniversitat Pompeu FabraInstitució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpainPasqual Maragall FoundationBarcelonaBeta Brain Research CenterBarcelonaSpain,Ben GlockerImperial College LondonDepartment of ComputingLondonUnited Kingdom,Benard O BotweUniversity of GhanaSchool of Biomedical & Allied Health SciencesAccraGhanaUniversity of LondonDepartment of Midwifery & Radiography, School of Health & Psychological SciencesLondonUK,Bishesh KhanalNepAl Applied Mathematics and Informatics Institute for Research (NAAMII)KathmanduNepal,Brigit BegerEuropean Heart NetworkBrusselsBelgium,Carol C WuUniversity of Texas MD Anderson Cancer CenterDepartment of Thoracic ImagingHoustonUnited States,Celia CintasIBM Research AfricaNairobiKenya,Curtis P LanglotzStanford University School of MedicineCenter for Artificial Intelligence in Medicine & ImagingStanfordUnited States,Daniel RueckertTechnical University MunichInstitute for AI and Informatics in Medicine, Klinikum rechts der IsarMunichGermanyImperial College LondonDepartment of ComputingLondonUK,Deogratias MzurikwaoMuhimbili University of Health and Allied SciencesDar es SalaamTanzania,Dimitrios I FotiadisFoundation for Research and Technology - Hellas (FORTH)Unit of Medical Technology and Intelligent Information SystemsIoanninaGreece,Doszhan ZhussupovAlmaty AI LabAlmatyKazakhstan,Enzo FerranteUniversidad Nacional del LitoralCONICETSanta FeArgentina,Erik MeijeringUniversity of New South WalesSchool of Computer Science and EngineeringSydneyAustralia,Eva WeickenFraunhofer Heinrich Hertz InstituteBerlinGermany,Fabio A GonzálezUniversidad Nacional de ColombiaComputing Systems and Industrial Engineering Dept.BogotáColombia,Folkert W AsselbergsUniversity of AmsterdamAmsterdam University Medical Centers, Department of CardiologyAmsterdamThe NetherlandsUniversity College LondonHealth Data Research UK and Institute of Health InformaticsLondonUK,Fred PriorUniversity of Arkansas for Medical SciencesDepartment of Biomedical InformaticsLittle RockUnited States,Gabriel P KrestinErasmus MC University Medical CenterDepartment of Radiology & Nuclear MedicineRotterdamthe Netherlands,Gary CollinsUniversity of OxfordCentre for Statistics in MedicineOxfordUK,Geletaw S TegenawJimma UniversityFaculty of Computing and InformaticsJimmaEthiopia,Georgios KaissisTechnical University MunichInstitute for AI and Informatics in Medicine, Klinikum rechts der IsarMunichGermany,Gianluca MisuracaUniversidad Politécnica de MadridDepartment of Artificial IntelligenceMadridSpain,Gianna TsakouResearch and Development LabGruppo MaggioliAthensGreece,Girish DwivediThe University of Western AustraliaDepartment of Advanced Clinical and Translational Cardiovascular ImagingPerthAustralia,Haridimos KondylakisHellenic Mediterranean UniversityDepartment of Electrical and Computer Engineering, Foundation for Research and Technology - Hellas (FORTH)CreteGreece,Harsha JayakodyUniversity of ColomboPostgraduate Institute of MedicineColomboSri Lanka,Henry C WoodrufMaastricht UniversityThe D-lab, Department of Precision Medicine, GROW - School for Oncology and ReproductionMaastrichtthe Netherlands,Hugo JWL AertsHarvard Medical SchoolArtificial Intelligence in Medicine Program, Mass General BrighamBostonUnited States,Ian WalshTechnology and Research (A*STAR)Bioprocessing Technology Institute, Agency for ScienceSingaporeSingapore,Ioanna ChouvardaAristotle University of ThessalonikiSchool of MedicineThessalonikiGreece,Irène BuvatInsermInstitut CurieOrsayFrance,Islem RekikImperial College LondonBASIRA Lab, Imperial-X and Computing DepartmentLondonUKIstanbul Technical UniversityFaculty of Computer and Informatics EngineeringIstanbulTurkey,James DuncanYale UniversityDepartments of Biomedical Engineering and Radiology & Biomedical Imaging, Schools of Engineering & Applied Science and MedicineNew HavenUnited States,Jayashree Kalpathy-CramerHarvard Medical SchoolMassachusetts General HospitalMassachusettsUnited States,Jihad ZahirCadi Ayyad UniversityLISI Laboratory, Computer Science DepartmentMarrakechMorocco,Jinah ParkKorea Advanced Institute of Science and TechnologySchool of ComputingDaejeonSouth Korea,John MonganUniversity of California San FranciscoDepartment of Radiology and Biomedical ImagingSan FranciscoUnited States,Judy W GichoyaEmory UniversityDepartment of Radiology & Imaging SciencesAtlantaUnited States,Julia A SchnabelHelmholtz Center MunichInstitute of Machine Learning in Biomedical ImagingMunichGermany,Kaisar KushibarUniversitat de BarcelonaArtificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer ScienceBarcelonaSpain,Katrine RiklundUmeå UniversityDepartment of Radiation Sciences, Diagnostic RadiologyUmeåSweden,Kensaku MoriNagoya UniversityGraduate School of InformaticsNagoyaJapan,Kostas MariasHellenic Mediterranean UniversityDepartment of Electrical and Computer Engineering, Foundation for Research and Technology - Hellas (FORTH)CreteGreece,Lameck M AmugongoNamibia University of Science & TechnologyDepartment of Software EngineeringWindhoekNamibia,Lauren A FromontThe Barcelona Institute of Science and TechnologyCenter for Genomic RegulationBarcelonaSpain,Lena Maier-HeinGerman Cancer Research Center (DKFZ)Div. Intelligent Medical Systems (IMSY)HeidelbergGermany,Leonor Cerdá AlberichLa Fe Health Research InstituteBiomedical Imaging Research GroupValenciaSpain,Leticia RittnerUniversity of CampinasSchool of Electrical and Computer EngineeringCampinasBrazil,Lighton PhiriUniversity of ZambiaDepartment of Library Information ScienceLusakaZambia,Linda Marrakchi-KacemUniversity of Tunis El ManarNational Engineering School of TunisTunisTunisia,Lluís Donoso-BachHospital Clínic of BarcelonaClinical Advanced Technologies institute (CATI)BarcelonaSpain,Luis Martí-BonmatíHospital Universitario y Politécnico La FeMedical Imaging DepartmentValenciaSpain,M Jorge CardosoKing’s College LondonSchool of Biomedical Engineering & Imaging SciencesLondonUnited Kingdom,Maciej Bobowicz, MDMedical University of Gdansk2nd Division of RadiologyGdanskPoland,Mahsa ShabaniGhent UniversityFaculty of Law and CriminologyGhentBelgium,Manolis TsiknakisHellenic Mediterranean UniversityDepartment of Electrical and Computer Engineering, Foundation for Research and Technology - Hellas (FORTH)CreteGreece,Maria A ZuluagaEURECOMData Science DepartmentSophia AntipolisFrance,Maria BielikovaKempelen Institute of Intelligent TechnologiesBratislavaSlovakia,Marie-Christine FritzscheTechnical University of MunichInstitute of History and Ethics in MedicineMunichGermany,Marius George LinguraruChildren’s National Hospital Washington DCSheikh Zayed Institute for Pediatric Surgical InnovationWashington DCUnited States,Markus WenzelFraunhofer Heinrich Hertz InstituteBerlinGermany,Marleen De BruijneErasmus MC University Medical CenterDepartment of Radiology & Nuclear MedicineRotterdamthe Netherlands,Martin G TolsgaardUniversity of CopenhagenCopenhagen Academy for Medical Education and Simulation RigshospitaletCopenhagenDenmark,Marzyeh GhassemiMassachusetts Institute of TechnologyElectrical Engineering and Computer Science (EECS) and Institute for Medical Engineering & Science (IMES)CambridgeUnited States,Md AshrafuzzamanMilitary Institute of Science and TechnologyDepartment of Biomedical EngineeringDhakaBangladesh,Melanie GoisaufBBMRI-ERIC, ELSI Services & ResearchGrazAustria,Mohammad YaqubMohamed Bin Zayed University of Artificial IntelligenceAbu DhabiUnited Arab Emirates,Mohammed AmmarUniversity M’Hamed Bougarangineering Systems and Telecommunication LaboratoryBoumerdesAlgeria,Mónica Cano AbadíaBBMRI-ERIC, ELSI Services & ResearchGrazAustria,Mukhtar M E MahmoudUniversity of KassalaFaculty of Computer Science and Information TechnologyKassalaSudan,Mustafa ElattarNile UniversityCenter for Informatics ScienceSheikh Zayed CityEgypt,Nicola RiekeNVIDIA GmbHHealthcare & Life Science EMEAMunichGermany,Nikolaos PapanikolaouChampalimaud FoundationComputational Clinical Imaging GroupLisbonPortugal,Noussair LazrakNew York UniversityHealth, Environment and policyNew YorkUnited States,Oliver DíazUniversitat de BarcelonaArtificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer ScienceBarcelonaSpain,Olivier SalvadoData61, The Commonwealth Scientific and Industrial Research Organisation (CSIRO)CanberraAustralia,Oriol PujolUniversitat de BarcelonaArtificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer ScienceBarcelonaSpain,Ousmane SallUniversité Virtuelle du SénégalPôle Sciences, Technologies et NumériqueDiamniadioSenegal,Pamela GuevaraUniversidad de ConcepciónFaculty of EngineeringConcepciónChile,Peter GordebekeEuropean Institute for Biomedical Imaging ResearchViennaAustria,Philippe LambinMaastricht UniversityThe D-lab, Department of Precision Medicine, GROW - School for Oncology and ReproductionMaastrichtthe Netherlands,Pieta BrownOrion HealthAucklandNew Zealand,Purang Abolmaesumithe University of British ColumbiaDepartment of Electrical and Computer EngineeringVancouverCanada,Qi DouThe Chinese University of Hong KongDepartment of Computer Science and EngineeringHong KongChina,Qinghua LuData61, The Commonwealth Scientific and Industrial Research Organisation (CSIRO)CanberraAustralia,Richard OsualaUniversitat de BarcelonaArtificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer ScienceBarcelonaSpain,Rose NakasiMakerere UniversityMakerere Artificial Intelligence LabKampalaUganda,S Kevin ZhouUniversity of Science and Technology of ChinaSchool of Biomedical Engineering & Suzhou Institute for Advanced ResearchSuzhouChina,Sandy NapelStanford UniversityIntegrative Biomedical Imaging Informatics at Stanford (IBIIS), Department of RadiologyStanford CAUnited States,Sara ColantonioInstitute of Information Science and Technologies of the National Research Council of ItalyPisaItaly,Shadi AlbarqouniUniversity Hospital BonnDepartment of Diagnostic and Interventional RadiologyBonnGermany,Smriti JoshiUniversitat de BarcelonaArtificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer ScienceBarcelonaSpain,Stacy CarterUniversity of WollongongAustralian Centre for Health Engagement, Evidence and Values, School of Health and SocietyNew South WalesAustralia,Stefan KleinErasmus MC University Medical CenterDepartment of Radiology & Nuclear MedicineRotterdamthe Netherlands,Steffen E PetersenQueen Mary University of LondonWilliam Harvey Research InstituteLondonUK,Susanna AussóTIC Salut Social FoundationArtificial Intelligence in Healthcare ProgramBarcelonaSpain,Suyash AwateIndian Institute of Technology BombayComputer Science and Engineering DepartmentMumbaiIndia,Tammy Riklin RavivBen-Gurion UniversitySchool of Electrical and Computer EngineeringBeer ShebaIsrael,Tessa CookUniversity of PennsylvaniaDepartment of Radiology, Perelman School of MedicinePhiladelphiaUnited States,Tinashe E M MutsvangwaUniversity of Cape TownDepartment of Human BiologyCape TownSouth Africa,Wendy A RogersMacquarie UniversityDepartment of Philosophy, and School of MedicineSydneyAustralia,Wiro J NiessenErasmus MC University Medical CenterDepartment of Radiology & Nuclear MedicineRotterdamthe Netherlands,Xènia Puig-BoschUniversitat de BarcelonaArtificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer ScienceBarcelonaSpain,Yi ZengChinese Academy of Sciencesenter for Long-term AIBeijingChina,Yunusa G MohammedGombe State UniversityDepartment of Human AnatomyGombeNigeria,Yves Saint James AquinoUniversity of WollongongAustralian Centre for Health Engagement, Evidence and Values, School of Health and SocietyNew South WalesAustralia,Zohaib SalahuddinMaastricht UniversityThe D-lab, Department of Precision Medicine, GROW - School for Oncology and ReproductionMaastrichtthe NetherlandsandMartijn P A StarmansErasmus MC University Medical CenterDepartment of Radiology & Nuclear MedicineRotterdamthe Netherlands

Abstract.

Abstract:
Background: Despite major advances in artificial intelligence (AI) for medicine and healthcare, the deployment and adoption of AI technologies remain limited in real-world clinical practice. In recent years, concerns have been raised about the technical, clinical, ethical and legal risks associated with medical AI. To increase adoption in the real world, it is essential that medical AI tools are trusted and accepted by patients, clinicians, health organisations and authorities. This paper describes the FUTURE-AI guideline as the first international consensus framework for guiding the development and deployment of trustworthy AI tools in healthcare.
Methods: The FUTURE-AI consortium was founded in 2021 and currently comprises 118 inter-disciplinary experts from 51 countries representing all continents, including AI scientists, clinicians, ethicists, and social scientists. Over a two-year period, the consortium defined guiding principles and best practices for trustworthy AI through an iterative process comprising an in-depth literature review, a modified Delphi survey, and online consensus meetings.
Findings: The FUTURE-AI framework was established based on six guiding principles for trustworthy AI in healthcare, i.e. Fairness, Universality, Traceability, Usability, Robustness and Explainability. Through consensus, a set of 28 best practices were defined, addressing technical, clinical, legal and socio-ethical dimensions of trustworthy AI. The recommendations cover the entire lifecycle of medical AI, from design, development and validation to regulation, deployment, and monitoring.Interpretation: FUTURE-AI is a risk-informed, assumption-free guideline which provides a structured approach for constructing medical AI tools that will be trusted, deployed and adopted in real-world practice. Researchers are encouraged to take the recommendations into account in proof-of-concept stages to facilitate future translation towards clinical practice of medical AI.
Funding: Support for this work was partially provided by the European Union’s Horizon 2020 under Grant Agreement No. 952103 (EuCanImage), No.952159 (ProCAncer-I), No.952172
(CHAIMELEON), No. 826494 (PRIMAGE) and No. 952179 (INCISIVE).

1. Introduction

Despite major advances in the field of medical AI, the deployment and adoption of AI technologies remain limited in real-world clinical practice. In recent years, concerns have been raised about the technical, clinical, ethical and social risks associated with medical AI(Lekadir etal., 2022; Vollmer etal., 2020). In particular, existing research has shown that medical AI tools can be prone to errors and patient harm, biases and increased health inequalities, lack of transparency and accountability, as well as data privacy and security breaches(Challen etal., 2019; Celi etal., 2022; He etal., 2019; Haibe-Kains etal., 2020; Murdoch, 2021).

To increase adoption in the real world, it is essential that medical AI tools are trusted and accepted by patients, clinicians, health organisations and authorities. However, there is an absence of clear, widely accepted guidelines on how medical AI tools should be designed, developed, evaluated and deployed to be trustworthy, i.e. technically robust, clinically safe, ethically sound and legally compliant. To have a real impact at scale, such guidelines for trustworthy and responsible AI must be obtained through wide consensus involving international and inter-disciplinary experts.

In other domains, international consensus guidelines have made lasting impacts. For example, the FAIR guideline(Wilkinson etal., 2016) for data management has been widely adopted by researchers, organisations and authorities, as they provided a logical framework for standardising and enhancing the tasks of data collection, curation, organisation and storage. While it can be argued that the FAIR principles do not cover every aspect of data management, as they focus more on findability, accessibility, interoperability and reusability of the data, and less on privacy and security, they delivered a code of practice that is now widely accepted and applied.

For medical AI, initial efforts have focused on providing recommendations for the reporting of AI studies for different medical domains or clinical tasks (e.g. TRIPOD-AI(Collins etal., 2021a), CLAIM(Mongan etal., 2020a), CONSORT-AI(Liu etal., 2020a), DECIDE-AI(Vasey etal., 2022), PROBAST-AI(Collins etal., 2021a), CLEAR(Kocak etal., 2023)). These guidelines do not provide best practices for the actual development and deployment of the AI tools but promote standardised and complete reporting of their development and evaluation. Recently, several researchers have published promising ideas on possible best practices for medical AI(Larson etal., 2021a; Reddy etal., 2021; Park and Han, 2018; Walsh etal., 2021; Maier-Hein etal., 2022a; Bradshaw etal., 2022). However, these proposals have not been established through wide international consensus and do not cover the whole lifecycle of medical AI (i.e. from design, development and evaluation to deployment, usage and monitoring). In 2020, a comprehensive self-assessment checklist for trustworthy AI was defined through consensus by Europe’s High-Level Expert Group on Artificial Intelligence, but it covered AI in general and did not address the specific risks and challenges of AI in medicine and healthcare(Ala-Pietilä etal., 2020).

This paper addresses an important gap in the field of medical AI, by delivering the very first international, consensus guideline for trustworthy medical AI that covers the entire AI lifecycle (Figure1).

FUTURE-AI: International consensus guideline for trustworthy and deployable artificial intelligence in healthcare (1)

The FUTURE-AI consortium was initiated in 2021 and currently comprises 118 international and inter-disciplinary experts from 51 countries (Figure1), representing all continents (Europe, North America, South America, Asia, Africa, and Oceania). Additionally, the members represent a variety of disciplines (e.g. data science, medical research, healthcare, computer engineering, medical ethics, social sciences) and data domains (e.g. radiology, genomics, mobile health, electronic health records, surgery, pathology). To develop the FUTURE-AI framework, we drew inspiration from the FAIR principles for data management and defined concise recommendations structured according to six guiding principles, i.e. Fairness, Universality, Traceability, Usability, Robustness, and Explainability (Figure2).

FUTURE-AI: International consensus guideline for trustworthy and deployable artificial intelligence in healthcare (2)

2. Materials and Methods

The FUTURE-AI framework was defined over a 24-month period through a modified Delphi approach (Table1).

StepPeriod
1Founding of the FUTURE-AI consortium by members ofJune 2021
research projects in Europe(Commission, [n. d.])
2Creation of working groups for each of the six guiding principlesJuly 2021
3Proposal of a first set of 55 recommendations through a use-caseSeptember 2021
driven approach focused on AI in medical imaging
4Feedback gathering through a survey with ¿100 international andNovember 2021 – March 2022
multi-disciplinary experts from all continents
5Analysis of results and derivation of a more concise list of 22March 2022 – April 2022
recommendations, generalised to AI for healthcare
6Second round of feedback from the expertsMay 2022 – July 2022
7Analysis of results and derivation of an improved and extended listJune 2022 – July 2022
of 30 recommendations
8Third round of feedback, by focusing on the main disagreementsSeptember 2022 – February 2023
and the manuscript’s first draft
9Four consensus online meetings to discuss remainingJune 2023
disagreements, resulting in the final 28 recommendations
10Finalisation and presentation of the FUTURE-AI consensusJune 2023
guideline in a journal publication

FUTURE-AI was initiated with an in-depth literature review on medical AI and trustworthiness, which resulted in the identification of key dimensions of relevance to trustworthy AI, including robustness, safety, security, fairness, transparency, traceability, accountability, generalisability, explainability, usability and responsible AI. To facilitate the use of the framework, some keywords were grouped, selected and re-ordered to obtain a reduced set of six guiding principles (Fairness, Universality, Traceability, Usability, Robustness, Explainability), which form the basis of the FUTURE-AI acronym.

Working groups composed of three experts each (including clinicians, data scientists and computer engineers) explored the six principles separately and proposed an initial set of best practices, by using AI for medical imaging as an initial use case. Furthermore, the working groups discussed the proposed recommendations, and removed overlaps and redundancies across the six principles, resulting in a first comprehensive set of 54 recommendations.

Subsequently, the FUTURE-AI consortium members provided systematic feedback on the first version of the FUTURE-AI guideline through a comprehensive survey. The experts could comment on each recommendation, rate its importance, propose missing recommendations, and provide additional feedback in free text. Based on the results of the survey, the list of recommendations was deemed too extensive, and was thus substantially reduced from 54 to 22 recommendations, while the scope was carefully broadened from AI for medical imaging to AI for healthcare.

The reduced set of recommendations was sent out to the FUTURE-AI consortium, together with a list of major disagreements that arose, for a second round of feedback and comments. This step resulted in a new version consisting of 30 recommendations, with a new “General” category in addition to the six guiding principles to account for recommendations that are transversal across all the dimensions of trustworthy AI.

Based on the machine learning technology readiness level (ML-TRL)(Lavin etal., 2022), the FUTURE-AI guideline was refined by distinguishing between medical AI tools at the research or proof-of-concept stage (i.e. ML-TRL 1 to 4) and those intended for clinical deployment (i.e. ML-TRL 5 to 9), as they require different levels of compliance. Hence, we asked the members of the consortium to rate the recommendations as recommended vs. highly recommended, for both proof-of-concept (low ML-TRL) and deployable AI tools (high ML-TRL). Finally, iterative discussions on the guideline, disagreements and manuscript were held, including during four dedicated online meetings, resulting in a final set of 28 consensus recommendations, which are listed in Table2.

LowHigh
RecommendationsML-ML-
TRLTRL
Ff1Define any potential sources of bias from an early stage++++
F2Collect data on individuals’ attributes, when possible++
3Evaluate potential biases and bias correction measures++
1Define intended clinical settings and cross-setting variations++++
U2Use community-defined standards (e.g. clinical definitions, technical standards)++
3Evaluate using external datasets and/or multiple sites++++
4Evaluate and demonstrate local clinical validity+++
1Implement a risk management process throughout the AI lifecycle+++
2Provide documentation (e.g. technical, clinical)++++
T3Define mechanisms for quality control of the AI inputs and outputs+++
4Implement a system for periodic auditing and updating+++
5Implement a logging system for usage recording+++
6Establish mechanisms for human oversight and governance
1Define intended use and user requirements from an early stage++++
U2Provide training materials and activities (e.g. tutorials, hands-on sessions)+++
3Evaluate user experience and acceptance with independent end-users+++
4Evaluate clinical utility and safety (e.g. effectiveness, harm, cost-benefit)+++
1Define sources of data variation from an early stage++++
R2Train with representative real-world data++++
3Evaluate and optimise robustness against real-world variations++++
E1Define the need and requirements for explainability with end-users++++
2Evaluate explainability with end-users (e.g. correctness, impact on users)++
1Engage inter-disciplinary stakeholders throughout the AI lifecycle++++
2Implement measures for data privacy and security++++
3Define adequate evaluation plan (e.g. datasets, metrics, reference methods)++++
4Identify and comply with applicable AI regulatory requirements+++
5Investigate and address ethical issues+++
General6Investigate and address social and societal issues++

3. FUTURE-AI guideline

In this section, we provide definitions and justifications for each of the six guiding principles and give an overview of the FUTURE-AI recommendations. Table2 provides a summary of the recommendations, together with the proposed level of compliance (i.e. recommended vs. highly recommended). More detailed descriptions are provided in Table 3 in the Appendix for readers who may require more information on any recommendation(s). Note that a glossary of the main terms used in this paper is provided in Table 4 in the Appendix, while the main stakeholders of relevance to the FUTURE-AI framework are listed in Table 5 in the Appendix.

3.1. Fairness

The Fairness principle states that medical AI tools should maintain the same performance across individuals and groups of individuals (including under-represented and disadvantaged groups). AI-driven medical care should be provided equally for all citizens, independently of their sex, gender, ethnicity, age, socio-economic status and (dis)abilities, among other attributes. Fair medical AI tools should be developed such that potential AI biases are minimised as much as possible, or identified and reported.

To this end, three recommendations for Fairness are defined in the FUTURE-AI framework. First, AI developers together with domain experts should define fairness for their specific use case and make an inventory of potential sources of bias (Fairness 1). Accordingly, to facilitate verification of AI fairness and non-discrimination, information on the subjects’ relevant attributes should be included in the datasets (Fairness 2). Finally, whenever this data is available, the development team should apply bias detection and correction methods, to obtain the best possible trade-off between fairness and accuracy (Fairness 3).

3.2. Universality

The Universality principle states that a medical AI tool should be generalisable outside the controlled environment where it was built. Specifically, the AI tool should be able to generalise to new patients and new users (e.g. new clinicians), and when applicable, to new clinical sites. Depending on the intended radius of application, medical AI tools should be as interoperable and as transferable as possible, so they can benefit citizens and clinicians at scale.To this end, four recommendations for Universality are defined in the FUTURE-AI framework. First, the AI developers should define the requirements for universality, i.e. the radius of application of their medical AI tool (e.g. clinical centres, countries, clinical settings), and accordingly anticipate any potential obstacles to universality, such as differences in clinical workflows, medical equipment or digital infrastructures (Universality 1). To enhance interoperability, development teams should favour the use of established community-defined standards (e.g. clinical definitions, medical ontologies, data annotations, technical standards) throughout the AI tool’s production lifetime (Universality 2). To enhance generalisability, the medical AI tool should be tested with external datasets and, when applicable, across multiple sites (Universality 3). Finally, medical AI tools should be evaluated for their local clinical validity, and if necessary, calibrated so they perform well given the local populations and local clinical workflows (Universality 4).

3.3. Traceability

The Traceability principle states that medical AI tools should be developed together with mechanisms for documenting and monitoring the complete trajectory of the AI tool, from development and validation to deployment and usage. This will increase transparency and accountability by providing detailed and continuous information on the AI tools during their lifetime to clinicians, healthcare organisations, citizens and patients, AI developers and relevant authorities. AI traceability will also enable continuous auditing of AI models(Oala etal., 2021a), identify risks and limitations, and update the AI models when needed.To this end, six recommendations for Traceability are defined in the FUTURE-AI framework. First, a system for risk management should be implemented throughout the AI lifecycle, including risk identification, assessment, mitigation, monitoring and reporting (Traceability 1). To increase transparency, relevant documentation should be provided for the stakeholder groups of interest, including AI information leaflets, technical documentation, and/or scientific publications (Traceability 2). After deployment, continuous quality control of AI inputs and outputs should be implemented, to identify inconsistent input data and implausible AI outputs (e.g. using uncertainty estimation), and to implement necessary model updates (Traceability 3). Furthermore, periodic auditing and updating of AI tools should be implemented (e.g. yearly) to detect and address any potential issue or performance degradation (Traceability 4). To increase traceability and accountability, an AI logging system should be implemented to keep a record of the usage of the AI tool, including for instance, user actions, accessed and used datasets, and identified issues (Traceability 5). Finally, mechanisms for human oversight and governance should be implemented, to enable selected users to flag AI errors or risks, overrule AI decisions, use human judgment instead, assign roles and responsibilities, and maintain the AI system over time (Traceability 6).

3.4. Usability

The Usability principle states that the end-users should be able to use a medical AI tool to achieve a clinical goal efficiently and safely in their real-world environment. On one hand, this means that end-users should be able to use the AI tool’s functionalities and interfaces easily and with minimal errors. On the other hand, the AI tool should be clinically useful and safe, e.g. improve the clinicians’ productivity and/or lead to better health outcomes for the patients and avoid harm.

To this end, four recommendations for Usability are defined in the FUTURE-AI framework. First, through a human-centred approach, target end-users (e.g. general practitioners, specialists, nurses, patients, hospital managers) should be engaged from an early stage to define the AI tool’s intended use, user requirements and human-AI interfaces (Usability 1). Second, training materials and training activities should be provided for all intended end-users, to ensure adequate usage of the AI tool, minimise errors and thus patient harm, and increase AI literacy (Usability 2). At the evaluation stage, the usability within the local clinical workflows, including human factors that may impact the usage of the AI tool(Sujan etal., 2019a) (e.g. satisfaction, confidence, ergonomics, learnability), should be assessed with representative and diverse end-users (Usability 3). Furthermore, the clinical utility and safety of the AI tools should be evaluated and compared with the current standard of care, to estimate benefits as well as potential harms for the citizens, clinicians and/or health organisations (Usability 4).

3.5. Robustness

The Robustness principle refers to the ability of a medical AI tool to maintain its performance and accuracy under expected or unexpected variations in the input data. Existing research has shown that even small, imperceptible variations in the input data may lead AI models into incorrect decisions(Finlayson etal., 2019a). Biomedical and health data can be subject to significant variations in the real world (both expected and unexpected), which can affect the performance of AI tools. Hence, it is important that medical AI tools are designed and developed to be robust against real-world variations, as well as evaluated and optimised accordingly.

To this end, three recommendations for Robustness are defined in the FUTURE-AI framework. At the design phase, the development team should first define robustness requirements for the medical AI application in question, by making an inventory of the potential sources of variation e.g. data-, equipment-, clinician-, patient- and centre-related variations (Robustness 1). Accordingly, the training datasets should be carefully selected, analysed and enriched to reflect these real-world variations as much as possible (Robustness 2). Subsequently, the robustness of the AI tool, as well as measures to enhance robustness, should be iteratively evaluated under conditions that reflect the variations of real-world clinical practice (Robustness 3).

3.6. Explainability

The Explainability principle states that medical AI tools should provide clinically meaningful information about the logic behind the AI decisions. While medicine is a high-stake discipline that requires transparency, reliability and accountability, machine learning techniques often produce complex models which are black boxes in nature. Explainability is considered desirable from a technological, medical, ethical, legal as well as patient perspective(Amann etal., 2020). Explainability is a complex task which has challenges that need to be carefully addressed during AI development and evaluation to ensure that AI explanations are clinically meaningful and beneficial to the end-users(Ghassemi etal., 2021a).

To this end, two recommendations for Explainability are defined in the FUTURE-AI framework. At the design phase, it should be first established with end-users and domain experts whether explainable AI is needed for the medical AI tool in questions. In this case, the specific goal and approaches for explainability should be defined (Explainability 1). After their implementation, the selected approaches for explainability should be evaluated, both quantitatively using in silico methods(Hedström etal., 2023a), as well qualitatively with end-users to assess their impact on the user’s satisfaction and performance (Explainability 2).

3.7. General recommendations

Finally, six general recommendations are defined in the FUTURE-AI framework, which apply across all principles of trustworthy AI in healthcare. First, AI developers should actively engage inter-disciplinary stakeholders throughout the production lifecycle, including healthcare professionals, patient representatives, ethicists and social scientists, data managers and legal experts (General 1). During the whole lifecycle from development to deployment, adequate measures should be put in place to ensure data protection and security, such as data de-identification and minimisation, privacy-enhancing techniques, and defences against malicious attacks (General 2). During all evaluation tasks, appropriate evaluation datasets, metrics and reference methods should be carefully selected to gather strong evidence on the medical AI tool’s trustworthiness (General 3). The AI development teams should verify and understand the applicable AI regulations from an early stage, so they can anticipate and meet their legal obligations (General 4). All general and application-specific ethical issues should be investigated, discussed and integrated into the practical development of the AI tool, through continuous interactions with domain specialists and ethicists(McLennan etal., 2020) (General 5). Finally, to ensure a positive impact on citizens and society, social and societal issues should be investigated and addressed (e.g. the tool’s impact on working conditions, relationships between citizens and health services, upskilling or deskilling of citizens and healthcare professionals(Aquino etal., 2023), environmental sustainability) (General 6).

4. Discussion

Despite the tremendous amount of research in medical AI in recent years, currently, only a limited number of AI tools have made the transition to clinical practice. While many studies have demonstrated the huge potential of AI to improve healthcare, significant clinical, technical, socio-ethical and legal challenges persist.

In this paper, we presented the results of an international effort to establish a consensus guideline for developing trustworthy and deployable AI tools in healthcare. Through an iterative process that lasted 24 months, the FUTURE-AI framework was established, comprising a well-structured, self-contained set of 28 recommendations, which covers the whole lifecycle of medical AI. By dividing the recommendations across six guiding principles, the pathways towards trustworthy AI are clearly characterised to facilitate their use throughout the AI tool’s lifecycle.

By the end of the process, all the recommendations were approved with less than 5% disagreement among all FUTURE-AI members. The FUTURE-AI consortium provided knowledge and expertise across a wide range of disciplines and stakeholders, resulting in consensus and wide support, both geographically and across domains. Hence, the FUTURE-AI guideline can benefit a wide range of stakeholders, as detailed in Table 5 in the Appendix.

FUTURE-AI is a risk-informed framework. It proposes to assess application-specific risks and challenges early in the process (e.g. risk of discrimination, lack of generalisability, data drifts, lack of acceptance by end-users, potential harm for patients, lack of transparency, data security vulnerabilities, ethical risks), then implement tailored measures to reduce these risks (e.g. collect data on individuals’ attributes to assess and mitigate bias). This is also a risk-benefit balancing exercise, as the specific measures to be implemented have benefits and potential weaknesses that the developers need to assess and balance. For example, collecting data on individuals’ attributes may increase the risk of re-identification, but can enable to reduce the risk of bias and discrimination. Hence, in FUTURE-AI, risk management (as recommended in Traceability 1) must be a continuous and transparent process throughout the AI tool’s lifecycle.

Furthermore, FUTURE-AI is an assumption-free, highly collaborative framework. It recommends to continuously engage with multi-disciplinary stakeholders to understand application-specific needs, risks and solutions (General 1). This is crucial to remove assumptions and investigate all possible risks and factors that may reduce trust in a given AI tool. For example, instead of making any assumption on possible sources of bias (e.g. sex or age), FUTURE-AI recommends that the developers engage with healthcare professionals, domain experts, representative citizens, and/or ethicists early in the process to investigate in depth the application-specific sources of bias, that may include factors well beyond standards attributes (e.g. breast density for AI applications in breast cancer).

For deployable AI tools, 24 recommendations out of 28 are rated as highly recommended (Table2). For research and proof-of-concept AI tools, only 12 recommendations are rated as highly recommended, but we advise that researchers use as many elements as possible from the FUTURE-AI guideline to facilitate future transitions towards real-world practice.

The FUTURE-AI guideline was defined in a generic manner to ensure it can be applied across a variety of domains (e.g. radiology, genomics, mobile health, electronic health records). However, for many recommendations, their applicability varies across medical use cases. Hence, the first recommendation in each of the FUTURE-AI framework’s principles is to identify the specificities to be addressed, such as the types of biases (Fairness 1), the clinical settings (Universality 1), or the need and approaches for explainable AI (Explainability 1).

The FUTURE-AI framework provides a set of general recommendations on how to enhance the trustworthiness of medical AI tools but does not impose any specific techniques for implementing each recommendation. While some examples of techniques are provided in the Appendix (Table 3), the final implementations should be defined by the developers, who should carefully select the most adequate methods given the application domain, clinical use case and data characteristics, as well as the advantages and limitations of each method.While we obtained a large consensus, some AI experts may disagree with some of the recommendations or may consider that some recommendations are either missing or not fully addressed. For example, while we propose mechanisms to enhance traceability and governance (e.g. AI logging), the issue of liability is yet to be addressed (e.g. who should be responsible for periodic auditing of the AI tools, who should be accountable when there is an error). Some of these key issues will require further investigations by multi-disciplinary researchers in the field of trustworthy AI, as well as by legal experts, regulators and authorities.

Aware of this limitation, we propose FUTURE-AI as a dynamic, living framework. Progressive development and adoption of medical AI tools will lead to new needs, challenges, and opportunities. To refine the FUTURE-AI guideline and learn from other voices, we set up a dedicated webpage (www.future-ai.eu) through which we invite the community to join the FUTURE-AI network and provide feedback based on their own experience and perspective. On the website, we include a FUTURE-AI self-assessment checklist, which comprises a set of questions and examples to facilitate and illustrate the use of the FUTURE-AI recommendations.

Additionally, we plan to organise regular outreach events such as webinars and workshops to exchange with medical AI researchers, manufacturers, evaluators, end-users and regulators.

References

  • (1)
  • a30 ([n. d.])[n. d.].European Commission, Directorate-General forCommunications Networks, Content and Technology, The Assessment List forTrustworthy Artificial Intelligence (ALTAI) for self assessment, PublicationsOffice, 2020, https://data.europa.eu/doi/10.2759/002360.
  • a14 ([n. d.])[n. d.].ISO/IEC JTC 1/SC 42 - artificial intelligence. ISO.May 27, 2023. Accessed May 31, 2023.https://www.iso.org/committee/6794475.html.
  • Ala-Pietilä etal. (2020)Pekka Ala-Pietilä,Yann Bonnet, Urs Bergmann,Maria Bielikova, Cecilia Bonefeld-Dahl,Wilhelm Bauer, Loubna Bouarfa,Raja Chatila, Mark Coeckelbergh,Virginia Dignum, etal.2020.The assessment list for trustworthyartificial intelligence (ALTAI).European Commission.
  • Amann etal. (2020)Julia Amann, AlessandroBlasimme, Effy Vayena, Dietmar Frey,and VinceI Madai. 2020.Explainability for artificial intelligence inhealthcare: a multidisciplinary perspective.BMC medical informatics and decision making20, 1 (2020),1–9.
  • Aquino etal. (2023)Yves SaintJames Aquino,WendyA Rogers, Annette Braunack-Mayer,Helen Frazer, KhinThan Win,Nehmat Houssami, Christopher Degeling,Christopher Semsarian, and StacyMCarter. 2023.Utopia versus dystopia: Professional perspectiveson the impact of healthcare artificial intelligence on clinical roles andskills.International Journal of MedicalInformatics 169 (2023),104903.
  • Arnold etal. (2019)Matthew Arnold, RachelKEBellamy, Michael Hind, Stephanie Houde,Sameep Mehta, Aleksandra Mojsilović,Ravi Nair, KNatesan Ramamurthy,Alexandra Olteanu, David Piorkowski,etal. 2019.FactSheets: Increasing trust in AI services throughsupplier’s declarations of conformity.IBM Journal of Research and Development63, 4/5 (2019),6–1.
  • Arras etal. (2022)Leila Arras, Ahmed Osman,and Wojciech Samek. 2022.CLEVR-XAI: A benchmark dataset for the ground truthevaluation of neural network explanations.Information Fusion 81(2022), 14–40.
  • Barocas etal. (2017)Solon Barocas, MoritzHardt, and Arvind Narayanan.2017.Fairness in machine learning.Nips tutorial 1(2017), 2017.
  • Bellamy etal. (2018)RachelKE Bellamy, KuntalDey, Michael Hind, SamuelC Hoffman,Stephanie Houde, Kalapriya Kannan,Pranay Lohia, Jacquelyn Martino,Sameep Mehta, Aleksandra Mojsilovic,etal. 2018.AI Fairness 360: An extensible toolkit fordetecting, understanding, and mitigating unwanted algorithmic bias.arXiv preprint arXiv:1810.01943(2018).
  • Bodenreider etal. (2018)Oliver Bodenreider, RonaldCornet, and DanielJ Vreeman.2018.Recent developments in clinicalterminologies—SNOMED CT, LOINC, and RxNorm.Yearbook of medical informatics27, 01 (2018),129–139.
  • Bradshaw etal. (2022)TylerJ Bradshaw, RonaldBoellaard, Joyita Dutta, AbhinavK Jha,Paul Jacobs, Quanzheng Li,Chi Liu, Arkadiusz Sitek,Babak Saboury, PeterJH Scott,etal. 2022.Nuclear medicine and artificial intelligence: bestpractices for algorithm development.Journal of Nuclear Medicine63, 4 (2022),500–510.
  • Cabitza etal. (2021)Federico Cabitza, AndreaCampagner, Felipe Soares,LuisGarcía de Guadiana-Romualdo,Feyissa Challa, Adela Sulejmani,Michela Seghezzi, and Anna Carobene.2021.The importance of being external. methodologicalinsights for the external validation of machine learning models in medicine.Computer Methods and Programs inBiomedicine 208 (2021),106288.
  • Celi etal. (2022)LeoAnthony Celi,Jacqueline Cellini, Marie-LaureCharpignon, EdwardChristopher Dee,Franck Dernoncourt, Rene Eber,WilliamGreig Mitchell, Lama Moukheiber,Julian Schirmer, Julia Situ,etal. 2022.Sources of bias in artificial intelligence thatperpetuate healthcare disparities—A global review.PLOS Digital Health 1,3 (2022), e0000022.
  • Challen etal. (2019)Robert Challen, JoshuaDenny, Martin Pitt, Luke Gompels,Tom Edwards, and KrasimiraTsaneva-Atanasova. 2019.Artificial intelligence, bias and clinical safety.BMJ Quality & Safety 28,3 (2019), 231–237.
  • Channa etal. (2021)Roomasa Channa, RisaWolf, and MichaelD Abramoff.2021.Autonomous artificial intelligence in diabeticretinopathy: from algorithm to clinical application.Journal of diabetes science and technology15, 3 (2021),695–698.
  • Collins etal. (2021a)GaryS Collins, PaulaDhiman, Constanza LAndaur Navarro, JieMa, Lotty Hooft, JohannesB Reitsma,Patricia Logullo, AndrewL Beam,Lily Peng, Ben VanCalster,etal. 2021a.Protocol for development of a reporting guideline(TRIPOD-AI) and risk of bias tool (PROBAST-AI) for diagnostic and prognosticprediction model studies based on artificial intelligence.BMJ open 11,7 (2021), e048008.
  • Collins etal. (2021b)GaryS Collins, PaulaDhiman, Constanza LAndaur Navarro, JieMa, Lotty Hooft, JohannesB Reitsma,Patricia Logullo, AndrewL Beam,Lily Peng, Ben VanCalster,etal. 2021b.Protocol for development of a reporting guideline(TRIPOD-AI) and risk of bias tool (PROBAST-AI) for diagnostic and prognosticprediction model studies based on artificial intelligence.BMJ open 11,7 (2021), e048008.
  • Commission ([n. d.])European Commission.[n. d.].AI for Health Imaging — Programme — H2020 — CORDIS.https://cordis.europa.eu/programme/id/H2020_DT-TDS-05-2020
  • DeGrave etal. (2021)AlexJ DeGrave, JosephDJanizek, and Su-In Lee.2021.AI for radiographic COVID-19 detection selectsshortcuts over signal.Nature Machine Intelligence3, 7 (2021),610–619.
  • Dormann (2020)CarstenF Dormann.2020.Calibration of probability predictions frommachine-learning and statistical models.Global ecology and biogeography29, 4 (2020),760–765.
  • Feng etal. (2022)Jean Feng, RachaelVPhillips, Ivana Malenica, AndrewBishara, AlanE Hubbard, LeoA Celi,and Romain Pirracchio. 2022.Clinical artificial intelligence qualityimprovement: towards continual monitoring and updating of AI algorithms inhealthcare.npj Digital Medicine 5,1 (2022), 66.
  • Ferryman and Pitcan (2018)Kadija Ferryman andMikaela Pitcan. 2018.Fairness in precision medicine.(2018).
  • Finlayson etal. (2019a)SamuelG Finlayson, JohnDBowers, Joichi Ito, JonathanL Zittrain,AndrewL Beam, and IsaacS Kohane.2019a.Adversarial attacks on medical machine learning.Science 363,6433 (2019), 1287–1289.
  • Finlayson etal. (2019b)SamuelG Finlayson, JohnDBowers, Joichi Ito, JonathanL Zittrain,AndrewL Beam, and IsaacS Kohane.2019b.Adversarial attacks on medical machine learning.Science 363,6433 (2019), 1287–1289.
  • Ganapathi etal. (2022)Shaswath Ganapathi, JoPalmer, JosephE Alderman, MelanieCalvert, Cyrus Espinoza, Jacqui Gath,Marzyeh Ghassemi, Katherine Heller,Francis Mckay, Alan Karthikesalingam,etal. 2022.Tackling bias in AI health datasets through theSTANDING Together initiative.Nature Medicine 28,11 (2022), 2232–2233.
  • Gao etal. (2019)Yuan Gao, Yuanyuan Wang,and Jinhua Yu. 2019.Optimized Resolution-Oriented Many-to-One IntensityStandardization Method for Magnetic Resonance Images.Applied Sciences 9,24 (2019), 5531.
  • Garrucho etal. (2022)Lidia Garrucho, KaisarKushibar, Socayna Jouide, Oliver Diaz,Laura Igual, and Karim Lekadir.2022.Domain generalization in deep learning based massdetection in mammography: A large-scale multi-center study.Artificial Intelligence in Medicine132 (2022), 102386.
  • Gebru etal. (2021)Timnit Gebru, JamieMorgenstern, Briana Vecchione,JenniferWortman Vaughan, Hanna Wallach,HalDaumé Iii, and Kate Crawford.2021.Datasheets for datasets.Commun. ACM 64,12 (2021), 86–92.
  • Ghassemi etal. (2021a)Marzyeh Ghassemi, LukeOakden-Rayner, and AndrewL Beam.2021a.The false hope of current approaches to explainableartificial intelligence in health care.The Lancet Digital Health3, 11 (2021),e745–e750.
  • Ghassemi etal. (2021b)Marzyeh Ghassemi, LukeOakden-Rayner, and AndrewL Beam.2021b.The false hope of current approaches to explainableartificial intelligence in health care.The Lancet Digital Health3, 11 (2021),e745–e750.
  • Ghorbani etal. (2019)Amirata Ghorbani, AbubakarAbid, and James Zou. 2019.Interpretation of neural networks is fragile. InProceedings of the AAAI conference on artificialintelligence, Vol.33. 3681–3688.
  • Haibe-Kains etal. (2020)Benjamin Haibe-Kains,GeorgeAlexandru Adam, Ahmed Hosny,Farnoosh Khodakarami, Massive AnalysisQuality Control (MAQC) SocietyBoard of Directors Shraddha Thakkar 35 KuskoRebecca 36 Sansone Susanna-Assunta 37 Tong Weida 35 Wolfinger Russ D. 38Mason Christopher E. 39 Jones Wendell 40 Dopazo Joaquin 41 FurlanelloCesare42, Levi Waldron, Bo Wang,Chris McIntosh, Anna Goldenberg,Anshul Kundaje, etal.2020.Transparency and reproducibility in artificialintelligence.Nature 586,7829 (2020), E14–E16.
  • He etal. (2019)Jianxing He, SallyLBaxter, Jie Xu, Jiming Xu,Xingtao Zhou, and Kang Zhang.2019.The practical implementation of artificialintelligence technologies in medicine.Nature medicine 25,1 (2019), 30–36.
  • Hedström etal. (2023a)Anna Hedström, LeanderWeber, Daniel Krakowczyk, DilyaraBareeva, Franz Motzkus, Wojciech Samek,Sebastian Lapuschkin, and Marina M-CHöhne. 2023a.Quantus: An explainable ai toolkit for responsibleevaluation of neural network explanations and beyond.Journal of Machine Learning Research24, 34 (2023),1–11.
  • Hedström etal. (2023b)Anna Hedström, LeanderWeber, Daniel Krakowczyk, DilyaraBareeva, Franz Motzkus, Wojciech Samek,Sebastian Lapuschkin, and Marina M-CHöhne. 2023b.Quantus: An explainable ai toolkit for responsibleevaluation of neural network explanations and beyond.Journal of Machine Learning Research24, 34 (2023),1–11.
  • IEEE ([n. d.])IEEE. [n. d.].IEEE SA - IEEE Recommended Practice for the qualitymanagement of datasets for Medical Artificial Intelligence. IEEE StandardsAssociation. Accessed May 31, 2023.https://standards.ieee.org/ieee/2801/7459/.
  • Kapoor and Narayanan (2022)Sayash Kapoor and ArvindNarayanan. 2022.Leakage and the reproducibility crisis in ML-basedscience.arXiv preprint arXiv:2207.07048(2022).
  • Kaviani etal. (2022)Sara Kaviani, KiJin Han,and Insoo Sohn. 2022.Adversarial attacks and defenses on AI in medicalimaging informatics: A survey.Expert Systems with Applications198 (2022), 116815.
  • Kocak etal. (2023)Burak Kocak, BettinaBaessler, Spyridon Bakas, RenatoCuocolo, Andrey Fedorov, LenaMaier-Hein, Nathaniel Mercaldo, HenningMüller, Fanny Orlhac, DanielPintodos Santos, etal. 2023.CheckList for EvaluAtion of Radiomics research(CLEAR): a step-by-step reporting guideline for authors and reviewersendorsed by ESR and EuSoMII.Insights into Imaging 14,1 (2023), 1–13.
  • Kompa etal. (2021)Benjamin Kompa, JasperSnoek, and AndrewL Beam.2021.Second opinion needed: communicating uncertainty inmedical machine learning.NPJ Digital Medicine 4,1 (2021), 4.
  • Königstorfer and Thalmann (2022)Florian Königstorfer andStefan Thalmann. 2022.AI Documentation: A path to accountability.Journal of Responsible Technology11 (2022), 100043.
  • Larson etal. (2021a)DavidB Larson, HughHarvey, DanielL Rubin, Neville Irani,RTse Justin, and CurtisP Langlotz.2021a.Regulatory frameworks for development andevaluation of artificial intelligence–based diagnostic imaging algorithms:summary and recommendations.Journal of the American College ofRadiology 18, 3 (2021),413–424.
  • Larson etal. (2021b)DavidB Larson, HughHarvey, DanielL Rubin, Neville Irani,RTse Justin, and CurtisP Langlotz.2021b.Regulatory frameworks for development andevaluation of artificial intelligence–based diagnostic imaging algorithms:summary and recommendations.Journal of the American College ofRadiology 18, 3 (2021),413–424.
  • Lavin etal. (2022)Alexander Lavin,CiaránM Gilligan-Lee, AlessyaVisnjic, Siddha Ganju, Dava Newman,Sujoy Ganguly, Danny Lange,AtílímGüneş Baydin,Amit Sharma, Adam Gibson,etal. 2022.Technology readiness levels for machine learningsystems.Nature Communications 13,1 (2022), 6039.
  • Lekadir etal. (2022)Karim Lekadir, GianlucaQuaglio, ATselioudis Garmendia, andCatherine Gallin. 2022.Artificial Intelligence in Healthcare-Applications,Risks, and Ethical and Societal Impacts.European Parliament (2022).
  • Lemay etal. (2022)Andreanne Lemay, KatharinaHoebel, ChristopherP Bridge, BrianBefano, Silvia DeSanjosé, DidemEgemen, AnaCecilia Rodriguez, MarkSchiffman, JohnPeter Campbell, andJayashree Kalpathy-Cramer.2022.Improving the repeatability of deep learning modelswith Monte Carlo dropout.npj Digital Medicine 5,1 (2022), 174.
  • Li etal. (2021)Xiaoxiao Li, Ziteng Cui,Yifan Wu, Lin Gu, andTatsuya Harada. 2021.Estimating and improving fairness with adversariallearning.arXiv preprint arXiv:2103.04243(2021).
  • Lima etal. (2019)DanielM Lima, JoseFRodrigues-Jr, AgmaJM Traina, FabioAPires, and MarcoA Gutierrez.2019.Transforming two decades of ePR data to OMOP CDMfor clinical research.Stud Health Technol Inform264, August (2019),233–7.
  • Liu etal. (2020a)Xiaoxuan Liu,SamanthaCruz Rivera, David Moher,MelanieJ Calvert, AlastairK Denniston,Hutan Ashrafian, AndrewL Beam,An-Wen Chan, GaryS Collins,Ara DarziJonathanJ Deeks, etal.2020a.Reporting guidelines for clinical trial reports forinterventions involving artificial intelligence: the CONSORT-AI extension.The Lancet Digital Health2, 10 (2020),e537–e548.
  • Liu etal. (2020b)Xiaoxuan Liu,SamanthaCruz Rivera, David Moher,MelanieJ Calvert, AlastairK Denniston,Hutan Ashrafian, AndrewL Beam,An-Wen Chan, GaryS Collins,Ara DarziJonathanJ Deeks, etal.2020b.Reporting guidelines for clinical trial reports forinterventions involving artificial intelligence: the CONSORT-AI extension.The Lancet Digital Health2, 10 (2020),e537–e548.
  • Maier-Hein etal. (2022a)Lena Maier-Hein, BjoernMenze, etal. 2022a.Metrics reloaded: Pitfalls and recommendations forimage analysis validation.arXiv. org 2206.01653(2022).
  • Maier-Hein etal. (2022b)Lena Maier-Hein, BjoernMenze, etal. 2022b.Metrics reloaded: Pitfalls and recommendations forimage analysis validation.arXiv. org 2206.01653(2022).
  • McLennan etal. (2020)Stuart McLennan, AmeliaFiske, LeoAnthony Celi, RuthMüller, Jan Harder, Konstantin Ritt,Sami Haddadin, and Alena Buyx.2020.An embedded ethics approach for AI development.Nature Machine Intelligence2, 9 (2020),488–490.
  • McLennan etal. (2022)Stuart McLennan, AmeliaFiske, Daniel Tigard, Ruth Müller,Sami Haddadin, and Alena Buyx.2022.Embedded ethics: a proposal for integrating ethicsinto the development of medical AI.BMC Medical Ethics 23,1 (2022), 6.
  • Mikołajczyk and Grochowski (2018)Agnieszka Mikołajczyk andMichał Grochowski. 2018.Data augmentation for improving deep learning inimage classification problem. In 2018international interdisciplinary PhD workshop (IIPhDW). IEEE,117–122.
  • Mitchell etal. (2019)Margaret Mitchell, SimoneWu, Andrew Zaldivar, Parker Barnes,Lucy Vasserman, Ben Hutchinson,Elena Spitzer, InioluwaDeborah Raji,and Timnit Gebru. 2019.Model cards for model reporting. InProceedings of the conference on fairness,accountability, and transparency. 220–229.
  • Mohseni etal. (2021)Sina Mohseni, NiloofarZarei, and EricD Ragan.2021.A multidisciplinary survey and framework for designand evaluation of explainable AI systems.ACM Transactions on Interactive IntelligentSystems (TiiS) 11, 3-4(2021), 1–45.
  • Mongan etal. (2020a)John Mongan, Linda Moy,and CharlesE KahnJr. 2020a.Checklist for artificial intelligence in medicalimaging (CLAIM): a guide for authors and reviewers., e200029pages.
  • Mongan etal. (2020b)John Mongan, Linda Moy,and CharlesE KahnJr. 2020b.Checklist for artificial intelligence in medicalimaging (CLAIM): a guide for authors and reviewers., e200029pages.
  • Murdoch (2021)Blake Murdoch.2021.Privacy and artificial intelligence: challenges forprotecting health information in a new era.BMC Medical Ethics 22,1 (2021), 1–5.
  • Ngiam and Khor (2019)KeeYuan Ngiam and WeiKhor. 2019.Big data and machine learning algorithms forhealth-care delivery.The Lancet Oncology 20,5 (2019), e262–e273.
  • Oala etal. (2021a)Luis Oala, AndrewGMurchison, Pradeep Balachandran, ShrutiChoudhary, Jana Fehr, AlixandroWerneckLeite, PeterG Goldschmidt, ChristianJohner, EloraDM Schörverth, RoseNakasi, etal. 2021a.Machine learning for health: algorithm auditing &quality control.Journal of medical systems45 (2021), 1–8.
  • Oala etal. (2021b)Luis Oala, AndrewGMurchison, Pradeep Balachandran, ShrutiChoudhary, Jana Fehr, AlixandroWerneckLeite, PeterG Goldschmidt, ChristianJohner, EloraDM Schörverth, RoseNakasi, etal. 2021b.Machine learning for health: algorithm auditing &quality control.Journal of medical systems45 (2021), 1–8.
  • Park and Han (2018)SeongHo Park andKyunghwa Han. 2018.Methodologic guide for evaluating clinicalperformance and effect of artificial intelligence technology for medicaldiagnosis and prediction.Radiology 286,3 (2018), 800–809.
  • Pleiss etal. (2017)Geoff Pleiss, ManishRaghavan, Felix Wu, Jon Kleinberg, andKilianQ Weinberger. 2017.On fairness and calibration.Advances in neural information processingsystems 30 (2017).
  • Rafner etal. (2022)Janet Rafner, DominikDellermann, Arthur Hjorth, DoraVeraszto, Constance Kampf, Wendy MacKay,and Jacob Sherson. 2022.Deskilling, upskilling, and reskilling: a case forhybrid intelligence.Morals & Machines 1,2 (2022), 24–39.
  • Reddy etal. (2021)Sandeep Reddy, WendyRogers, Ville-Petteri Makinen, EnricoCoiera, Pieta Brown, Markus Wenzel,Eva Weicken, Saba Ansari,Piyush Mathur, Aaron Casey,etal. 2021.Evaluation framework to guide implementation of AIsystems into healthcare settings.BMJ health & care informatics28, 1 (2021).
  • Rouzrokh etal. (2022)Pouria Rouzrokh, BardiaKhosravi, Shahriar fa*ghani, ManaMoassefi, DianaV VeraGarcia, YashbirSingh, Kuan Zhang, GianMarco Conte,and BradleyJ Erickson. 2022.Mitigating bias in radiology machine learning: 1.Data handling.Radiology: Artificial Intelligence4, 5 (2022),e210290.
  • Sahiner etal. (2023)Berkman Sahiner, WeijieChen, RaviK Samala, and NicholasPetrick. 2023.Data drift in medical machine learning:implications and potential remedies.The British Journal of Radiology(2023), 20220878.
  • Selvan etal. (2022)Raghavendra Selvan, NikhilBhagwat, LasseF WolffAnthony, BenjaminKanding, and ErikB Dam.2022.Carbon footprint of selecting and training deeplearning models for medical image analysis. InInternational Conference on Medical Image Computingand Computer-Assisted Intervention. Springer, 506–516.
  • Sperrin etal. (2022)Matthew Sperrin, RichardDRiley, GaryS Collins, and GlenPMartin. 2022.Targeted validation: validating clinical predictionmodels in their intended population and setting.Diagnostic and Prognostic Research6, 1 (2022),24.
  • Sujan etal. (2019a)Mark Sujan, DominicFurniss, Kath Grundy, Howard Grundy,David Nelson, Matthew Elliott,Sean White, Ibrahim Habli, andNick Reynolds. 2019a.Human factors challenges for the safe use ofartificial intelligence in patient care.BMJ health & care informatics26, 1 (2019).
  • Sujan etal. (2019b)Mark Sujan, DominicFurniss, Kath Grundy, Howard Grundy,David Nelson, Matthew Elliott,Sean White, Ibrahim Habli, andNick Reynolds. 2019b.Human factors challenges for the safe use ofartificial intelligence in patient care.BMJ health & care informatics26, 1 (2019).
  • Tian and Zhang (2022)Yingjie Tian and YuqiZhang. 2022.A comprehensive survey on regularization strategiesin machine learning.Information Fusion 80(2022), 146–166.
  • Tjoa and Guan (2020)Erico Tjoa and c*ntaiGuan. 2020.A survey on explainable artificial intelligence(xai): Toward medical xai.IEEE transactions on neural networks andlearning systems 32, 11(2020), 4793–4813.
  • Varoquaux and Cheplygina (2022)Gaël Varoquaux andVeronika Cheplygina. 2022.Machine learning for medical imaging:methodological failures and recommendations for the future.NPJ digital medicine 5,1 (2022), 48.
  • Vasey etal. (2022)Baptiste Vasey, MyuraNagendran, Bruce Campbell, DavidAClifton, GaryS Collins, Spiros Denaxas,AlastairK Denniston, Livia Faes,Bart Geerts, Mudathir Ibrahim,etal. 2022.Reporting guideline for the early-stage clinicalevaluation of decision support systems driven by artificial intelligence:DECIDE-AI.Nature medicine 28,5 (2022), 924–933.
  • Vokinger etal. (2021)KerstinN Vokinger, StefanFeuerriegel, and AaronS Kesselheim.2021.Mitigating bias in machine learning for medicine.Communications medicine1, 1 (2021),25.
  • Vollmer etal. (2020)Sebastian Vollmer, BilalAMateen, Gergo Bohner, FranzJKirály, Rayid Ghani, Pall Jonsson,Sarah Cumbers, Adrian Jonas,KatherineSL McAllister, Puja Myles,etal. 2020.Machine learning and artificial intelligenceresearch for patient benefit: 20 critical questions on transparency,replicability, ethics, and effectiveness.bmj 368(2020).
  • Walsh etal. (2021)Ian Walsh, DmytroFishman, Dario Garcia-Gasulla, TiinaTitma, Gianluca Pollastri, JenniferHarrow, FotisE Psomopoulos, andSilvioCE Tosatto. 2021.DOME: recommendations for supervised machinelearning validation in biology.Nature methods 18,10 (2021), 1122–1127.
  • Wilkinson etal. (2016)MarkD Wilkinson, MichelDumontier, IjJ Aalbersberg, G Appleton,M Axton, A Baak, NBlomberg, JW Boiten, LB da SilvaSantos,PE Bourne, etal.2016.The FAIR Guiding Principles for scientific datamanagement and stewardship. Sci Data 3: 160018.
  • Zhang etal. (2022)Kuan Zhang, BardiaKhosravi, Sanaz Vahdati, Shahriarfa*ghani, Fred Nugen, SeyedMoeinRassoulinejad-Mousavi, Mana Moassefi,Jaidip ManikraoM Jagtap, Yashbir Singh,Pouria Rouzrokh, etal.2022.Mitigating bias in radiology machine learning: 2.Model development.Radiology: Artificial Intelligence4, 5 (2022),e220010.
  • Zhou etal. (2021)Qian Zhou, Zhi-hang Chen,Yi-heng Cao, and Sui Peng.2021.Clinical impact and quality of randomizedcontrolled trials involving interventions evaluating artificial intelligenceprediction tools: a systematic review.NPJ digital medicine 4,1 (2021), 154.

APPENDIX

Appendix A TABLES

RecommendationDescription
Fairness 1.
Define sources of bias
Bias in medical AI is application-specific(Ferryman and Pitcan, 2018). At the design phase, the development teamshould identify possible types and sources of bias for their AI tool(Ganapathi etal., 2022). These may includegroup attributes (e.g. sex, gender, age, ethnicity, socioeconomics, geography), themedical profiles of the individuals (e.g. with comorbidities or disability), as well as human biases during data labeling, data curation, or the selection of the input features.
Fairness 2.
Collect data on attributes
To identify biases and apply measures for increased fairness, relevant attributes of the individuals, such as sex, gender, age, ethnicity, risk factors, comorbidities or disabilities, should be collected. This should be subject to informed consent and approval by ethics committees to ensure an appropriate balance between the benefits for non-discrimination and risks for re-identification.
Fairness 3.
Evaluate & correct biases
When possible, i.e. the individuals’ attributes are included in the data, bias detection methods should be applied by using fairness metrics(Barocas etal., 2017; Bellamy etal., 2018). To correct for any identified biases, mitigation measures should be applied (e.g. data re-sampling, bias-free representations, equalised odds post-processing)(Vokinger etal., 2021; Li etal., 2021; Pleiss etal., 2017; Rouzrokh etal., 2022; Zhang etal., 2022) and tested to verify their impact on both the tool’s fairness and the model’s accuracy. Importantly, any potential bias should be documented and reported to inform the end-users and citizens (see Traceability 2).
Universality 1.
Define clinical settings
At the design phase, the development team should specify the clinical settings in which the AI tool will be applied (e.g. primary healthcare centres, hospitals, home care, low vs. high-resource settings, one or multiple countries), and anticipate potential obstacles to universality (e.g. differences in clinical definitions, medical equipment or IT infrastructures across settings).
Universality 2.
Use existing standards
To ensure the quality and interoperability of the AI tool, it should be developed based on existing community-defined standards. These may include clinical definitions, medical ontologies (e.g. SNOMED CT(Bodenreider etal., 2018), OMOP(Lima etal., 2019)), interface standards (e.g. DICOM, FHIR HL7), data annotations, evaluation criteria(Maier-Hein etal., 2022b), and technical standards (e.g. IEEE(IEEE, [n. d.]) or ISO(a14, [n. d.])).
Universality 3.
Evaluate using external data
To assess generalisability, technical validation of the AI tools should be performed with external datasets that are distinct from those used for training(Cabitza etal., 2021). These may include reference or benchmarking datasets which are representative for the task in question (i.e. approximating the expected real-world variations). Except for AI tools intended for single centres, the clinical evaluation studies should be performed at multiple sites(Sperrin etal., 2022) to assess performance and interoperability across clinical workflows. If the tool’s generalisability is limited, mitigation measures (e.g. transfer learning or domain adaptation) should be considered, applied and tested.
Universality 4.
Evaluate local clinical
validity
Clinical settings vary in many aspects, such as populations, equipment, clinical workflows, and end-users. Hence to ensure trust at each site, the AI tools should be evaluated for their local clinical validity(Larson etal., 2021b). In particular, the AI tool should fit the local clinical workflows and perform well on the local populations. If the performance is decreased when evaluated locally, re-calibration of the AI model should be performed (e.g., through model fine-tunning or retraining).
Traceability 1.
Implement risk management
Throughout the AI tool’s lifecycle, the development team should analyse potential risks, assess each risk’s likelihood, effects and risk-benefit balance, define risk mitigation measures, monitor the risks and mitigations continuously, and maintain a risk management file. The risks may include those explicitly covered by the FUTURE-AI guiding principles (e.g. bias, harm), but also application-specific risks. Other risks to consider include human factors that may lead to misuse of the AI tool (e.g. not following the instructions, receiving insufficient training), application of the AI tool to individuals who are not within the target population, use of the tool by others than the target end-users (e.g. technician instead of physician), hardware failure, incorrect data annotations or input values, and adversarial attacks. Mitigation measures may include warnings to the users, system shutdown, re-processing of the input data, the acquisition of new input data, or the use of an alternative procedure or human judgment only.
Traceability 2.
Provide documentation
To increase transparency, traceability, and accountability, adequate documentation should be created and maintained for the AI tool(Königstorfer and Thalmann, 2022), which may include (i) an AI information leaflet to inform citizens and healthcare professionals about the tool’s intended use, risks (e.g. biases) and instructions for use; (ii) a technical document(Mitchell etal., 2019; Arnold etal., 2019; Gebru etal., 2021) to inform AI developers, health organisations and regulators about the AI model’s properties (e.g. hyperparameters), training and testing data, evaluation criteria and results, biases and other limitations, and periodic audits and updates; (iii) a publication based on existing AI reporting standards(Collins etal., 2021b; Mongan etal., 2020b; Liu etal., 2020b), and (iv) a risk management file (see Traceability 1).
Traceability 3.
Implement continuous
quality control
The AI tool should be developed and deployed with mechanisms for continuous monitoring and quality control of the AI inputs and outputs(Oala etal., 2021b), such as to identify missing or out-of-range input variables, inconsistent data formats or units, incorrect annotations or data pre-processing, and erroneous or implausible AI outputs. For quality control of the AI decisions, uncertainty estimates should be provided (and calibrated(Dormann, 2020)) to inform the end-users on the degree of confidence in the results(Kompa etal., 2021). Finally, when necessary, model updates should be applied to address any identified limitations and enhance the AI models over time(Feng etal., 2022).
Traceability 4.
Implement periodic auditing
The AI tool should be developed and deployed with a configurable system for periodic auditing(Oala etal., 2021b), which should define site-specific datasets and timelines for periodic evaluations (e.g. every year). The periodic auditing should enable the identification of data or concept drifts, newly occurring biases, performance degradation(Sahiner etal., 2023) or changes in the decision making of the end-users. Accordingly, necessary updates to the AI models or AI tools should be applied(Feng etal., 2022).
Traceability 5.
Implement AI logging
To increase traceability and accountability, an AI logging system should be implemented to trace the user’s main actions in a privacy-preserving manner, specify the data that is accessed and used, record the AI predictions and clinical decisions, and log any encountered issues. Time-series statistics and visualisations should be used to inspect the usage of the AI tool over time.
Traceability 6.
Implement human oversight
Given the high-stake nature of medical AI, human oversight is essential and increasingly required by policy makers and regulators(Larson etal., 2021b; a30, [n. d.]). Human-AI interfaces and human-in-the-loop mechanisms should be designed and implemented to perform specific quality checks (e.g. to flag biases, errors or implausible explanations), and to overrule the AI decisions when necessary. Furthermore, governance of the AI tool in the health organisation should be specified, including roles and responsibilities for performing risk management, periodic auditing, human oversight, and AI tool maintenance.
Usability 1.
Define user requirements
The AI developers should engage clinical experts, end-users (e.g. patients, physicians) and other relevant stakeholders (e.g. data managers, administrators) from an early stage, to compile information on the AI tool’s intended use and end-user requirements (e.g. human-AI interfaces), as well as on human factors that may impact the usage of the AI tool(Sujan etal., 2019b) (e.g. ergonomics, intuitiveness, experience, learnability).
Usability 2.
Provide training
To facilitate best usage of the AI tool, minimise errors and harm, and increase AI literacy, the developers should provide training materials (e.g. tutorials, manuals, examples) in accessible language and/or training activities (e.g. hands-on sessions), taking into account the diversity of end-users (e.g. clinical specialists, nurses, technicians, citizens or administrators).
Usability 3.
Evaluate clinical usability
To facilitate adoption, the usability of the AI tool should be evaluated in the real world with representative and diverse end-users (e.g. with respect to sex, gender, age, clinical role, digital proficiency, (dis)ability). The usability tests should gather evidence on the user’s satisfaction, performance and productivity. These tests should also verify whether the AI tool impacts the behaviour and decision making of the end-users.
Usability 4.
Evaluate clinical utility
The AI tool should be evaluated for its clinical utility and safety. The clinical evaluations of the AI tool should show benefits for the clinician (e.g. increased productivity, improved care), for the patient (e.g. earlier diagnosis, better outcomes), and/or for the healthcare organisation (e.g. reduced costs, optimised workflows), when compared to the current standard of care. Additionally, it is important to show that the AI tool is safe and does not cause harm to individuals (or specific groups), such as through a randomised clinical trial(Zhou etal., 2021).
Robustness 1.
Define sources of data
variation
At the design phase, an inventory should be made of the application-specific sources of variation that may impact the AI tool’s robustness in the real world. These may include differences in equipment, technical fault of a machine, data heterogeneities during data acquisition or annotation, and/or adversarial attacks(Finlayson etal., 2019b).
Robustness 2.
Train with representative
data
Clinicians, citizens and other stakeholders are more likely to trust the AI tool if it is trained on data that adequately represents the variations encountered in real-world clinical practice(Ngiam and Khor, 2019). Hence, the training datasets should be carefully selected, analysed and enriched according to the sources of variation identified at the design phase (see Robustness 1).
Robustness 3.
Evaluate & optimise
robustness
Evaluation studies should be implemented to evaluate the AI tool’s robustness (including stress tests and repeatability tests(Lemay etal., 2022)), by considering all potential sources of variation (see Robustness 1), such as data-, equipment-, clinician-, patient- and centre-related variations. Depending on the results, mitigation measures should be implemented to optimise the robustness of the AI model, such as regularisation(Tian and Zhang, 2022), data augmentation(Mikołajczyk and Grochowski, 2018), data harmonisation(Gao etal., 2019), or domain adaptation(Garrucho etal., 2022).
Explainability 1.
Define explainability needs
At the design phase, it should be established if explainability is required for the AI tool. In this case, the specific requirements for explainability should be defined with representative experts and end-users, including (i) the goal of the explanations (e.g. global description of the model’s behaviour vs. local explanation of each AI decision), (ii) the most suitable approach for AI explainability(Tjoa and Guan, 2020), and (iii) the potential limitations to anticipate and monitor (e.g. over-reliance of the end-users on the AI decision(Ghassemi etal., 2021b)).
Explainability 2.
Evaluate explainability
The explainable AI methods should be evaluated, first quantitatively by using in silico methods to assess the correctness of the explanations(Arras etal., 2022; Hedström etal., 2023b), then qualitatively with end-users to assess their impact on user satisfaction, confidence and clinical performance(Mohseni etal., 2021). The evaluations should also identify any limitations of the AI explanations (e.g. they are clinically incoherent(DeGrave etal., 2021) or sensitive to noise or adversarial attacks(Ghorbani etal., 2019), they unreasonably increase the confidence in the AI-generated results(Channa etal., 2021)).
General 1.
Engage stakeholders
continuously
Throughout the AI tool’s lifecycle, the AI developers should continuously engage with inter-disciplinary stakeholders, such as healthcare professionals, citizens, patient representatives, expert ethicists, data managers and legal experts. This interaction will facilitate the understanding and anticipation of the needs, obstacles and pathways towards acceptance and adoption.
General 2.
Ensure data protection
Adequate measures to ensure data privacy and security should be put in place throughout the AI lifecycle. These may include privacy-enhancing techniques (e.g. differential privacy, encryption), data protection impact assessment and appropriate data governance after deployment (e.g. logging system for data access, see Traceability 5). If de-identification is implemented (e.g. pseudonymisation, k-anonymity), the balance between the health benefits for citizens and the risks for re-identification should be carefully assessed and considered. Furthermore, the manufacturers and deployers should implement and regularly evaluate measures for protecting the AI tool against malicious attacks, such as by using system-level cybersecurity solutions or application-specific defense mechanisms(Kaviani etal., 2022) (e.g. attack detection or mitigation).
General 3.
Define adequate
evaluation plan
To increase trust and adoption, an appropriate evaluation plan should be defined (including test data, metrics and reference methods). First, adequate test data should be selected for assessing each dimension of trustworthy AI. In particular, the test data should be well separated from the training to prevent data leakage(Kapoor and Narayanan, 2022). Furthermore, adequate evaluation metrics should be carefully selected, taking into account their benefits and potential flaws(Varoquaux and Cheplygina, 2022). Finally, benchmarking with respect to reference AI tools or standard practice should be performed to enable a comparative assessment of model performance.
General 4.
Comply with AI regulations
The development team should identify the applicable AI regulations depending on the relevant jurisdictions. This should be done at an early stage to anticipate regulatory obligations based on the medical AI tool’s intended classification and risks.
General 5.
Investigate ethical issues
In addition to the well-known ethical issues that arise in medical AI (e.g. privacy, transparency, equity, autonomy), AI developers, domain specialists and professional ethicists should identify, discuss and address all application-specific ethical, social and societal issues as an integral part of the development and deployment of the AI too(McLennan etal., 2022).
General 6.
Investigatesocial issues
Social and societal implications should be considered and addressed when developing the AI tool, to ensure a positive impact on citizens and society. Relevant issues include the impact of the AI tool on the working conditions and power relations, on the new skills (or deskilling) of the healthcare professionals and citizens(Rafner etal., 2022), and on future interactions between citizens, health professionals and social carers. Furthermore, for environmental sustainability, AI developers should consider strategies to reduce the carbon footprint of the AI tool(Selvan etal., 2022).
TermDefinition
AI auditingA periodic evaluation of an AI tool to assess its performance and working conditions over time, and to identify potential problems.
AI deploymentThe process of placing a completed AI tool into a live clinical environment where it can be used for its intended purpose.
AI designEarly stage of an AI’s production lifetime, during which specifications and plans are defined for the subsequent development of the AI tool
AI developmentThe process of training AI models and building AI-human interfaces, based on the specifications and plans from the AI design phase.
AI evaluationThe assessment of an AI tool’s added value in its intended clinical setting.
AI modelA program trained using a machine learning algorithm to perform a given task based on specific input data.
AI monitoringThe process of tracking the behavior of a deployed AI tool over time, to identify potential degradation in performance and implement mitigation measures such as model updating.
AI regulationA set of requirements and obligations defined by public authorities, that AI developers, deployers and users must adhere to.
AI riskAny negative effect that may occur when using an AI tool.
AI toolA software that comprises the AI model plus a user interface that can be used by the end-users to perform a given AI-powered clinical task.
AI trainingThe process of using machine learning algorithms to build AI models that learn to perform specific tasks based on existing data samples.
AI updatingThe process of re-training or fine-tuning the AI model after some time to improve its performance and correct identified issues.
AI validationThe assessment of an AI model’s performance.
AttributePersonal quality, trait or characteristic of an individual or group of individuals, such as sex, gender, age, ethnicity, socioeconomic status or disability. Protected attributes refer to those attributes that, by law, cannot be discriminated against (i.e. attributes that are protected by law).
BenchmarkingThe practice of comparing the performance of multiple AI tools (or an AI tool against the standard practice) based on a common reference dataset and a set of predefined performance criteria and metrics.
BiasSystematic, prejudiced errors by an AI tool against certain individuals or subgroups due to inadequate data or assumptions used during the training of the machine learning model.
Clinical safetyThe capability of an AI tool to keep individuals and patients safe and not to cause them any harm.
Clinical settingThe environment or location where the AI tool will be used, such as a hospital, a radiology department, a primary care centre, or for home-based care.
Clinical utilityThe capability of an AI tool to be useful in its intended clinical settings, such as to improve clinical outcomes, to increase the clinicians’ productivity, or to reduce healthcare costs.
Concept driftChanges in relationship between AI model inputs and outputs.
Data driftChanges in the distribution of the AI model’s input data over time.
Data quality controlThe process of assessing the quality of the input data, to identify potential defects that may affect the correct functioning of the AI tool.
Deployable AIAI developed with a high technology readiness level (TRL) (5-9) intended for deployment in clinical practice.
Ethical AIAI that adheres to key ethical values and human rights, such as the rights to privacy, equity and autonomy.
ExplainabilityThe ability of an AI tool to provide clinically meaningful information about the logic behind the AI decisions.
FairnessThe ability of an AI tool to treat equally individuals with similar characteristics or subgroups of individuals including under-represented groups.
Human oversightA procedure or set of procedures put in place to ensure an AI tool is used under the supervision of a human (e.g. a clinician), who is able to overrule the AI decisions and take the final clinical decision.
Intended useClinical purpose or clinical task that the AI tool aims to realise in its intended clinical setting.
LoggingThe process of keeping a log of events that occur while using an AI tool, such as user actions, accessed and used datasets, clinical decisions, and identified issues.
Proof-of-concept AIAI developed with a low machine learning technology readiness level (ML-TRL) (1-4) to demonstrate the feasibility of a new AI method or new AI concept.
Real worldThe clinical environment in which AI tools will be applied in practice, outside the controlled environment of research labs.
Responsible AIAI that is designed, developed, evaluated, and monitored by employing an appropriate code of conduct and appropriate methods to achieve technical, clinical, ethical, and legal requirements (e.g. efficacy, safety, fairness, robustness, transparency).
RobustnessThe ability of an AI tool to overcome expected or unexpected variations, such as due to noise or artefacts in the data.
Third-party evaluatorAn independent evaluator who did not participate in any way in the design or development of the AI tool to be evaluated.
TraceabilityThe ability of an AI tool to be monitored over its complete lifecycle.
Trustworthy AIAI with proven characteristics such as efficacy, safety, fairness, robustness, transparency, which enable relevant stakeholders such as citizens, clinicians, health organisations and authorities to rely on it and adopt it in real-world practice.
Trustworthy AIvs.
Responsible AI
For trustworthy AI, the emphasis is on the characteristics of the AI tool and how they are perceived by the stakeholders of interest (e.g. patients, clinicians), while for responsible AI, the emphasis is on the developers, evaluators and managers of the AI tool, and the code of conduct and methods they employ to obtain trustworthy AI tools.
UniversalityThe ability of an AI tool to generalise across clinical settings.
UsabilityThe degree to which an AI tool is fit to be used by end-users in the intended clinical setting.
StakeholdersFUTURE-AI usage
AI ethicistsTo embed ethics into the development of medical AI tools.
AI evaluators/clinical trialistsTo perform more comprehensive, multi-faceted evaluations of medical AI tools based on the principles of trustworthy AI.To assess the trustworthiness of AI tools.
Citizens and patientsTo increase literacy about medical AI and trustworthy AI.To increase engagement in the production and evaluation of medical AI tools.
Conferences/journalsTo promote best practices and new methods for trustworthy AI among researchers reading or publishing scientific papers.
Data managersTo support the development and deployment of medical AI tools that are compliant with data protection/governance principles.
Educational institutionsTo educate students from all disciplines (machine learning, computer science, medicine, ethics, social sciences) on the principles and approaches for trustworthy AI.
Funding agenciesTo promote new research projects that integrate best practices and new approaches for responsible AI.
Health organisationsTo guide healthcare organisations in the evaluation, deployment and monitoring of medical AI tools.To verify the trustworthiness of AI tools.
Healthcare professionalsTo adopt the principles of trustworthy AI and best practices among the healthcare professions.To engage clinicians in the design, development, evaluation and monitoring of medical AI tools.
IT managersTo promote IT solutions for the deployment and monitoring of trustworthy and secure AI tools in clinical practice.
Legal expertsTo ensure compliance with applicable laws and regulations related to medical AI and data protection.
Manufacturers of medical AI devicesTo adopt best practices for responsible AI within companies.To develop and/or commercialise new AI tools that will be accepted, certified and deployed for clinical use.
Public authoritiesTo adapt existing regulations and policies on medical AI.
Regulatory bodiesTo enhance the procedures for the evaluation, certification and monitoring of AI tools as medical devices.
Researchers and developers in medical AI.To investigate new methods according to the recommendations for trustworthy AI.To develop proof-of-concepts that can more easily transition into deployable AI tools for clinical practice.
Scientific/medical societiesTo promote the principles of trustworthy AI and best practices among scientific and medical communities.
Social scientistsTo ensure social and societal dimensions of medical AI are considered.
Standardisation bodiesTo develop new standards that facilitate the implementation, evaluation and adoption of trustworthy AI tools in healthcare.

Appendix B FULL AUTHOR AFFILIATIONS

Karim Lekadir, Artificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer Science, Universitat de Barcelona, Barcelona, Spain
Aasa Feragen, DTU Compute, Technical University of Denmark, Kgs Lyngby, Denmark
Abdul Joseph Fofanah, Department of Mathematics and Computer Science, Faculty of Science and Technology, Milton Margai Technical University, Freetown, Sierra Leone
Alejandro F Frangi, Centre for Computational Imaging & Simulation Technologies in Biomedicine, Schools ofComputing and Medicine, University of Leeds, Leeds, United Kingdom and Medical Imaging Research Center (MIRC), Cardiovascular Science and Electronic Engineering Departments, KU Leuven, Leuven, Belgium
Alena Buyx Institute of History and Ethics in Medicine, Technical University of Munich, Munich, Germany
Anais Emelie Artificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer Science, Universitat de Barcelona, Barcelona, Spain
Andrea Lara Faculty of Engineering of Systems, Informatics and Sciences of Computing, Galileo University, Guatemala City, Guatemala
Antonio R Porras Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
An-Wen Chan Department of Medicine, Women’s College Research Institute, University of Toronto, Toronto, Canada
Arcadi Navarro Institució Catalana de Recerca i Estudis Avançats (ICREA) and Universitat Pompeu Fabra, Barcelona, Spain andBarcelona Beta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
Ben Glocker Department of Computing, Imperial College London, London, United Kingdom
Benard O Botwe School of Biomedical & Allied Health Sciences, University of Ghana, Accra, Ghana and Department of Midwifery & Radiography, School of Health & Psychological Sciences, City University of London, UK
Bishesh Khanal NepAl Applied Mathematics and Informatics Institute for research (NAAMII), Kathmandu, Nepal
Brigit Beger European Heart Network, Brussels, Belgium
Carol C Wu Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, United States
Celia Cintas IBM Research Africa, Nairobi, Kenya
Curtis P Langlotz Center for Artificial Intelligence in Medicine & Imaging, Stanford University School of Medicine, Stanford, United States
Daniel Rueckert Institute for AI and Informatics in Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany and Department of Computing, Imperial College London, London, UK
Deogratias Mzurikwao Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
Dimitrios I Fotiadis Unit of Medical Technology and Intelligent Information Systems, Foundation for Research and Technology - Hellas (FORTH), Ioannina, Greece
Doszhan Zhussupov Almaty AI Lab, Almaty, Kazakhstan
Enzo Ferrante CONICET, Universidad Nacional del Litoral, Santa Fe, Argentina
Erik Meijering School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
Eva Weicken Fraunhofer Heinrich Hertz Institute, Berlin, Germany
Fabio A González Computing Systems and Industrial Engineering Dept., Universidad Nacional de Colombia, Bogotá, Colombia
Folkert W Asselbergs Amsterdam University Medical Centers, Department of Cardiology, University of Amsterdam, Amsterdam, The Netherlands and Health Data Research UK and Institute of Health Informatics, University College London, London, United Kingdom
Fred Prior Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, United States
Gabriel P Krestin Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
Gary Collins Centre for Statistics in Medicine, University of Oxford, Oxford, United Kingdom
Geletaw S Tegenaw Faculty of Computing and Informatics, JiT, Jimma University, Jimma, Ethiopia
Georgios Kaissis Institute for AI and Informatics in Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
Gianluca Misuraca Department of Artificial Intelligence, Universidad Politécnica de Madrid, Madrid, Spain
Gianna Tsakou Gruppo Maggioli, Research and Development Lab, Athens, Greece
Girish Dwivedi Department of Advanced Clinical and Translational Cardiovascular Imaging, The University of Western Australia, Perth, Australia
Haridimos Kondylakis Department of Electrical and Computer Engineering, Hellenic Mediterranean University and Foundation for Research and Technology - Hellas (FORTH), Crete, Greece
Harsha Jayakody Postgraduate Institute of Medicine, University of Colombo, Colombo, Sri Lanka
Henry C Woodruf The D-lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
Hugo JWL Aerts Artificial Intelligence in Medicine Program, Mass General Brigham, Harvard Medical School, Boston, United States
Ian Walsh Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
Ioanna Chouvarda School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
Irène Buvat Institut Curie, Inserm, Orsay, France
Islem Rekik BASIRA Lab, Imperial-X and Computing Department, Imperial College London, UK and ]Faculty of Computer and Informatics Engineering, Istanbul Technical University, Turkey
James Duncan Departments of Biomedical Engineering and Radiology & Biomedical Imaging, Schools of Engineering & Applied Science and Medicine, Yale University, New Haven, United States
Jayashree Kalpathy-Cramer Massachusetts General Hospital, Harvard Medical School, Massachusetts, United States
Jihad Zahir LISI Laboratory, Computer Science Department, Cadi Ayyad University, Marrakech, Morocco
Jinah Park School of Computing, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
John Mongan Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, United States
Judy W Gichoya Department of Radiology & Imaging Sciences, Emory University, Atlanta, United States
Julia A Schnabel Institute of Machine Learning in Biomedical Imaging, Helmholtz Center Munich, Munich, Germany
Kaisar Kushibar Artificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer Science, Universitat de Barcelona, Barcelona, Spain
Katrine Riklund Department of Radiation Sciences, Diagnostic Radiology, Umeå University, Umeå, Sweden
Kensaku Mori Graduate School of Informatics, Nagoya University, Nagoya, Japan
Kostas Marias Department of Electrical and Computer Engineering, Hellenic Mediterranean University and Foundation for Research and Technology - Hellas (FORTH), Crete, Greece
Lameck M Amugongo Department of Software Engineering, Namibia University of Science & Technology, Windhoek, Namibia
Lauren A Fromont Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
Lena Maier-Hein Div. Intelligent Medical Systems (IMSY), German Cancer Research Center (DKFZ), Heidelberg, Germany
Leonor Cerdá Alberich Biomedical Imaging Research Group, La Fe Health Research Institute, Valencia, Spain
Leticia Rittner School of Electrical and Computer Engineering, University of Campinas, Campinas, Brazil
Lighton Phiri Department of Library Information Science, University of Zambia, Lusaka, Zambia
Linda Marrakchi-Kacem National Engineering School of Tunis, University of Tunis El Manar, Tunis, Tunisia
Lluís Donoso-Bach Clinical Advanced Technologies institute (CATI), Hospital Clínic of Barcelona, Barcelona, Spain
Luis Martí-Bonmatí Medical Imaging Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
M Jorge Cardoso School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
Maciej Bobowicz 2nd Division of Radiology, Medical University of Gdansk, Gdansk, Poland
Mahsa Shabani Faculty of Law and Criminology, Ghent University, Ghent, Belgium
Manolis Tsiknakis Department of Electrical and Computer Engineering, Hellenic Mediterranean University and Foundation for Research and Technology - Hellas (FORTH), Crete, Greece
Maria A Zuluaga Data Science Department, EURECOM, Sophia Antipolis, France
Maria Bielikova Kempelen Institute of Intelligent Technologies, Bratislava, Slovakia
Marie-Christine Fritzsche Institute of History and Ethics in Medicine, Technical University of Munich, Munich, Germany
Marius George Linguraru Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital Washington DC, United States
Markus Wenzel Fraunhofer Heinrich Hertz Institute, Berlin, Germany
Marleen De Bruijne Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
Martin G Tolsgaard Copenhagen Academy for Medical Education and Simulation Rigshospitalet, University of, Copenhagen, Copenhagen, Denmark
Marzyeh Ghassemi Electrical Engineering and Computer Science (EECS) and Institute for Medical Engineering & Science (IMES), Massachusetts Institute of Technology, Cambridge, United States
Md Ashrafuzzaman Department of Biomedical Engineering, Military Institute of Science and Technology, Dhaka, Bangladesh
Melanie Goisauf BBMRI-ERIC, ELSI Services & Research, Graz, Austria
Mohammad Yaqub Mohamed Bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
Mohammed Ammar Engineering Systems and Telecommunication Laboratory, University M’Hamed Bougara, Boumerdes, Algeria
Mónica Cano Abadía BBMRI-ERIC, ELSI Services & Research, Graz, Austria
Mukhtar M E Mahmoud Faculty of Computer Science and Information Technology, University of Kassala, Kassala, Sudan
Mustafa Elattar Center for Informatics Science, Nile University, Sheikh Zayed City, Egypt
Nicola Rieke Healthcare & Life Science EMEA, NVIDIA GmbH, Munich, Germany
Nikolaos Papanikolaou Computational Clinical Imaging Group, Champalimaud Foundation, Lisbon, Portugal
Noussair Lazrak Health, Environment and policy, New York University, New York, United States
Oliver Díaz Artificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer Science, Universitat de Barcelona, Barcelona, Spain
Olivier Salvado Data61, The Commonwealth Scientific and Industrial Research Organisation (CSIRO), Canberra, Australia
Oriol Pujol Artificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer Science, Universitat de Barcelona, Barcelona, Spain
Ousmane Sall Pôle Sciences, Technologies et Numérique, Université Virtuelle du Sénégal, Diamniadio, Senegal
Pamela Guevara Faculty of Engineering, Universidad de Concepción, Concepción, Chile
Peter Gordebeke European Institute for Biomedical Imaging Research, Vienna, Austria
Philippe Lambin The D-lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
Pieta Brown Orion Health, Auckland, New Zealand
Purang Abolmaesumi Department of Electrical and Computer Engineering, the University of British Columbia, Vancouver, BC, Canada
Qi Dou Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
Qinghua Lu Data61, The Commonwealth Scientific and Industrial Research Organisation (CSIRO), Canberra, Australia
Richard Osuala Artificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer Science, Universitat de Barcelona, Barcelona, Spain
Rose Nakasi Makerere Artificial Intelligence Lab, Makerere University, Kampala, Uganda
S Kevin Zhou School of Biomedical Engineering & Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China
Sandy Napel Integrative Biomedical Imaging Informatics at Stanford (IBIIS), Department of Radiology, Stanford University, Stanford CA, United States
Sara Colantonio Institute of Information Science and Technologies of the National Research Council of Italy, Pisa, Italy
Shadi Albarqouni Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
Smriti Joshi Artificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer Science, Universitat de Barcelona, Barcelona, Spain
Stacy Carter Australian Centre for Health Engagement, Evidence and Values, School of Health and Society, University of Wollongong, New South Wales, Australia
Stefan Klein Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
Steffen E Petersen William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
Susanna Aussó Artificial Intelligence in Healthcare Program, TIC Salut Social Foundation, Barcelona, Spain
Suyash Awate, Computer Science and Engineering Department, Indian Institute of Technology Bombay, Mumbai, India
Tammy Riklin Raviv School of Electrical and Computer Engineering, Ben-Gurion University, Beer Sheba, Israel
Tessa Cook Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
Tinashe E M Mutsvangwa Department of Human Biology, University of Cape Town, Cape Town, South Africa
Wendy A Rogers Department of Philosophy, and School of Medicine, Macquarie University, Sydney, Australia
Wiro J Niessen Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
Xènia Puig-Bosch Artificial Intelligence in Medicine Lab (BCN-AIM), Department of Mathematics and Computer Science, Universitat de Barcelona, Barcelona, Spain
Yi Zeng Center for Long-term AI, Chinese Academy of Sciences, Beijing, China
Yunusa G Mohammed Department of Human Anatomy, Gombe State University, Gombe, Nigeria
Yves Saint James Aquino Australian Centre for Health Engagement, Evidence and Values, School of Health and Society, University of Wollongong, New South Wales, Australia
Zohaib Salahuddin The D-lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
Martijn P A Starmans Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands

FUTURE-AI: International consensus guideline for trustworthy and deployable artificial intelligence in healthcare (2024)
Top Articles
Target Extends Enhancements to Pay and Benefits and Provides COVID-19 Business Update
What are your target compensation expectations? | Tandym Group
Https //Paperlesspay.talx.com/Gpi
Swgoh Darth Vader Mods
Netlearning Login Rwjbh
Sssniperwolf Number 2023
Bookmark Cshive
S10 Mpg
Uta Frontrunner Twitter
Ellaeats Tumblr
Gay Pnp Zoom Meetings
6023445010
How Much Is Cvs Sports Physical
PNC Bank Review 2024
Sandra Sancc
Demystifying the C-Suite: A Close Look at the Top Executive Roles - 33rd Square
Eztv Ig
Lighthouse Diner Taylorsville Menu
Baca's Funeral Chapels & Sunset Crematory Las Cruces Obituaries
FREE Printable Pets Animal Playdough Mats
Bx11
Nephi Veterinarian
Craigs List Plattsburgh Ny
Horseware Deken Amigo Bravo 100gr Donkerblauw - 130/183 | bol
Morgan Plus Four 2024 review
Ihub Kblb
1773X To
Maine Marine Forecast Gyx
Sun Commercial Obituaries
R/Maddenultimateteam
Pokerev Telegram
Royal Carting Holidays 2022
Mrballen Political Views
9044906381
Sam's Club Stafford Gas Price
Withers Not In Sarcophagus
Etfh Hatchery
Danielle Longet
Culver's Flavor Of The Day Whitewater
Boostmaster Lin Yupoo
Craigslist In Visalia California
The Menu Showtimes Near Regal Edwards Ontario Mountain Village
Uw Oshkosh Wrestling
Sarah Snook Weight Gain
Destep: 10 tips voor de scherpste destep-analyse ooit!
Roman Numerals Chart, Translation Tips & History
Art Labeling Activity The Big Picture Of Nutrient Catabolism — I Hate CBT's
Birmingham National Weather Service
Good Number To Shoot For
Basketball Defense: 1-3-1 half court trap
Pollen Count Butler Pa
Latest Posts
Article information

Author: Reed Wilderman

Last Updated:

Views: 5689

Rating: 4.1 / 5 (72 voted)

Reviews: 87% of readers found this page helpful

Author information

Name: Reed Wilderman

Birthday: 1992-06-14

Address: 998 Estell Village, Lake Oscarberg, SD 48713-6877

Phone: +21813267449721

Job: Technology Engineer

Hobby: Swimming, Do it yourself, Beekeeping, Lapidary, Cosplaying, Hiking, Graffiti

Introduction: My name is Reed Wilderman, I am a faithful, bright, lucky, adventurous, lively, rich, vast person who loves writing and wants to share my knowledge and understanding with you.