101
|
Li M, Jiang Z, Shen W, Liu H. Deep learning in bladder cancer imaging: A review. Front Oncol 2022; 12:930917. [PMID: 36338676 PMCID: PMC9631317 DOI: 10.3389/fonc.2022.930917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
Deep learning (DL) is a rapidly developing field in machine learning (ML). The concept of deep learning originates from research on artificial neural networks and is an upgrade of traditional neural networks. It has achieved great success in various domains and has shown potential in solving medical problems, particularly when using medical images. Bladder cancer (BCa) is the tenth most common cancer in the world. Imaging, as a safe, noninvasive, and relatively inexpensive technique, is a powerful tool to aid in the diagnosis and treatment of bladder cancer. In this review, we provide an overview of the latest progress in the application of deep learning to the imaging assessment of bladder cancer. First, we review the current deep learning approaches used for bladder segmentation. We then provide examples of how deep learning helps in the diagnosis, staging, and treatment management of bladder cancer using medical images. Finally, we summarize the current limitations of deep learning and provide suggestions for future improvements.
Collapse
Affiliation(s)
- Mingyang Li
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zekun Jiang
- Ministry of Education (MoE) Key Lab of Artificial Intelligence, Artificial Intelligence (AI) Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Shen
- Ministry of Education (MoE) Key Lab of Artificial Intelligence, Artificial Intelligence (AI) Institute, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Haitao Liu, ; Wei Shen,
| | - Haitao Liu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Haitao Liu, ; Wei Shen,
| |
Collapse
|
102
|
Loménie N, Bertrand C, Fick RH, Ben Hadj S, Tayart B, Tilmant C, Farré I, Azdad SZ, Dahmani S, Dequen G, Feng M, Xu K, Li Z, Prevot S, Bergeron C, Bataillon G, Devouassoux-Shisheboran M, Glaser C, Delaune A, Valmary-Degano S, Bertheau P. Can AI predict epithelial lesion categories via automated analysis of cervical biopsies: The TissueNet challenge? J Pathol Inform 2022; 13:100149. [PMID: 36605109 PMCID: PMC9808029 DOI: 10.1016/j.jpi.2022.100149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 12/26/2022] Open
Abstract
The French Society of Pathology (SFP) organized its first data challenge in 2020 with the help of the Health Data Hub (HDH). The organization of this event first consisted of recruiting nearly 5000 cervical biopsy slides obtained from 20 pathology centers. After ensuring that patients did not refuse to include their slides in the project, the slides were anonymized, digitized, and annotated by expert pathologists, and finally uploaded to a data challenge platform for competitors from around the world. Competing teams had to develop algorithms that could distinguish 4 diagnostic classes in cervical epithelial lesions. Among the many submissions from competitors, the best algorithms achieved an overall score close to 95%. The final part of the competition lasted only 6 weeks, and the goal of SFP and HDH is now to allow for the collection to be published in open access for the scientific community. In this report, we have performed a "post-competition analysis" of the results. We first described the algorithmic pipelines of 3 top competitors. We then analyzed several difficult cases that even the top competitors could not predict correctly. A medical committee of several expert pathologists looked for possible explanations for these erroneous results by reviewing the images, and we present their findings here targeted for a large audience of pathologists and data scientists in the field of digital pathology.
Collapse
Affiliation(s)
- Nicolas Loménie
- LIPADE, UFR Mathématiques-Informatiques, Université Paris Cité, 45 rue des Saints-Pères, 75006 Paris, France,Corresponding author.
| | | | | | | | | | | | | | | | - Samy Dahmani
- Algoscope, 9 rue Gaspard Monge, 60200 Compiègne, France
| | - Gilles Dequen
- Laboratoire Modélisation, Information, Systèmes (MIS), Université de Picardie Jules Verne, 80080 Amiens, France
| | | | - Kele Xu
- Tongji University, Shanghai, China
| | - Zimu Li
- Tongji University, Shanghai, China
| | - Sophie Prevot
- Pathologie, CHU Bicêtre, APHP, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | | | | | - Mojgan Devouassoux-Shisheboran
- Centre de Pathologie Sud des Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, 165 chemin du grand Revoyet, 69495 Pierre Bénite Cedex, France
| | - Claire Glaser
- Pathologie, CHG Versailles, 177 Rue de Versailles, 78150 Le Chesnay-Rocquencourt, France
| | - Agathe Delaune
- Plateforme de données de santé - Health Data Hub, 9 rue Georges Pitard, 75015 Paris, France
| | - Séverine Valmary-Degano
- Pathologie, Université Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, CHU, Grenoble 38000, France
| | - Philippe Bertheau
- Pathologie, CHU Saint-Louis, APHP, Université Paris Cité, 1 avenue Claude Vellefaux, 75010 Paris, France
| |
Collapse
|
103
|
Borgohain DJ, Bhardwaj RK, Verma MK. Mapping the literature on the application of artificial intelligence in libraries (AAIL): a scientometric analysis. LIBRARY HI TECH 2022. [DOI: 10.1108/lht-07-2022-0331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PurposeArtificial Intelligence (AI) is an emerging technology and turned into a field of knowledge that has been consistently displacing technologies for a change in human life. It is applied in all spheres of life as reflected in the review of the literature section here. As applicable in the field of libraries too, this study scientifically mapped the papers on AAIL and analyze its growth, collaboration network, trending topics, or research hot spots to highlight the challenges and opportunities in adopting AI-based advancements in library systems and processes.Design/methodology/approachThe study was developed with a bibliometric approach, considering a decade, 2012 to 2021 for data extraction from a premier database, Scopus. The steps followed are (1) identification, selection of keywords, and forming the search strategy with the approval of a panel of computer scientists and librarians and (2) design and development of a perfect algorithm to verify these selected keywords in title-abstract-keywords of Scopus (3) Performing data processing in some state-of-the-art bibliometric visualization tools, Biblioshiny R and VOSviewer (4) discussing the findings for practical implications of the study and limitations.FindingsAs evident from several papers, not much research has been conducted on AI applications in libraries in comparison to topics like AI applications in cancer, health, medicine, education, and agriculture. As per the Price law, the growth pattern is exponential. The total number of papers relevant to the subject is 1462 (single and multi-authored) contributed by 5400 authors with 0.271 documents per author and around 4 authors per document. Papers occurred mostly in open-access journals. The productive journal is the Journal of Chemical Information and Modelling (NP = 63) while the highly consistent and impactful is the Journal of Machine Learning Research (z-index=63.58 and CPP = 56.17). In the case of authors, J Chen (z-index=28.86 and CPP = 43.75) is the most consistent and impactful author. At the country level, the USA has recorded the highest number of papers positioned at the center of the co-authorship network but at the institutional level, China takes the 1st position. The trending topics of research are machine learning, large dataset, deep learning, high-level languages, etc. The present information system has a high potential to improve if integrated with AI technologies.Practical implicationsThe number of scientific papers has increased over time. The evolution of themes like machine learning implicates AI as a broad field of knowledge that converges with other disciplines. The themes like large datasets imply that AI may be applied to analyze and interpret these data and support decision-making in public sector enterprises. Theme named high-level language emerged as a research hotspot which indicated that extensive research has been going on in this area to improve computer systems for facilitating the processing of data with high momentum. These implications are of high strategic worth for policymakers, library stakeholders, researchers and the government as a whole for decision-making.Originality/valueThe analysis of collaboration, prolific authors/journals using consistency factor and CPP, testing the relationship between consistency (z-index) and impact (h-index), using state-of-the-art network visualization and cluster analysis techniques make this study novel and differentiates it from the traditional bibliometric analysis. To the best of the author's knowledge, this work is the first attempt to comprehend the research streams and provide a holistic view of research on the application of AI in libraries. The insights obtained from this analysis are instrumental for both academics and practitioners.
Collapse
|
104
|
Wang K, Ren Y, Ma L, Fan Y, Yang Z, Yang Q, Shi J, Sun Y. Deep Learning-Based Prediction of Treatment Prognosis from Nasal Polyp Histology Slides. Int Forum Allergy Rhinol 2022; 13:886-898. [PMID: 36066094 DOI: 10.1002/alr.23083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Histopathology of nasal polyps contains rich prognostic information, which is difficult to objectively extract. In the present study, we aimed to develop a prognostic indicator of patient outcomes by analyzing scanned conventional haematoxylin and eosin (H&E) -stained slides alone using deep learning. METHODS An interpretable supervised deep learning model was developed using 185 H&E-stained whole-slide images (WSIs) of nasal polyps, each from a patient randomly selected from the pool of 232 patients who underwent endoscopic sinus surgery at the First Affiliated Hospital of Sun Yat-sen University (internal cohort). We internally validated the model on a holdout dataset from the internal cohort (47 H&E-stained WSIs) and externally validated the model on 122 H&E-stained WSIs from the Seventh Affiliated Hospital of Sun Yat-sen University and the University of Hong Kong-Shenzhen Hospital (external cohort). A poor prognosis score (PPS) was established to evaluate patient outcomes, and then risk activation mapping was applied to visualize the histopathological features underlying PPS. RESULTS The model yielded a patient-level sensitivity of 79.5%, and specificity of 92.3%, with areas under the receiver operating characteristic curve of 0.943, on the multi-center external cohort. The predictive ability of PPS was superior to that of conventional tissue eosinophil number. Notably, eosinophil infiltration, goblet cell hyperplasia, glandular hyperplasia, squamous metaplasia, and fibrin deposition were identified as the main underlying features of PPS. CONCLUSIONS Our deep learning model is an effective method for decoding pathological images of nasal polyps, providing a valuable solution for disease prognosis prediction and precise patient treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kanghua Wang
- Department of Otolaryngology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.,Department of Otolaryngology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yong Ren
- Center for Digestive Disease, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.,Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Ling Ma
- Department of Otorhinolaryngology, the University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Yunping Fan
- Department of Otolaryngology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zheng Yang
- Department of Pathology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Qintai Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianbo Shi
- Department of Otolaryngology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yueqi Sun
- Department of Otolaryngology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.,Department of Otolaryngology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
105
|
Kleppe A, Skrede OJ, De Raedt S, Hveem TS, Askautrud HA, Jacobsen JE, Church DN, Nesbakken A, Shepherd NA, Novelli M, Kerr R, Liestøl K, Kerr DJ, Danielsen HE. A clinical decision support system optimising adjuvant chemotherapy for colorectal cancers by integrating deep learning and pathological staging markers: a development and validation study. Lancet Oncol 2022; 23:1221-1232. [PMID: 35964620 DOI: 10.1016/s1470-2045(22)00391-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND The DoMore-v1-CRC marker was recently developed using deep learning and conventional haematoxylin and eosin-stained tissue sections, and was observed to outperform established molecular and morphological markers of patient outcome after primary colorectal cancer resection. The aim of the present study was to develop a clinical decision support system based on DoMore-v1-CRC and pathological staging markers to facilitate individualised selection of adjuvant treatment. METHODS We estimated cancer-specific survival in subgroups formed by pathological tumour stage (pT<4 or pT4), pathological nodal stage (pN0, pN1, or pN2), number of lymph nodes sampled (≤12 or >12) if not pN2, and DoMore-v1-CRC classification (good, uncertain, or poor prognosis) in 997 patients with stage II or III colorectal cancer considered to have no residual tumour (R0) from two community-based cohorts in Norway and the UK, and used these data to define three risk groups. An external cohort of 1075 patients with stage II or III R0 colorectal cancer from the QUASAR 2 trial was used for validation; these patients were treated with single-agent capecitabine. The proposed risk stratification system was evaluated using Cox regression analysis. We similarly evaluated a risk stratification system intended to reflect current guidelines and clinical practice. The primary outcome was cancer-specific survival. FINDINGS The new risk stratification system provided a hazard ratio of 10·71 (95% CI 6·39-17·93; p<0·0001) for high-risk versus low-risk patients and 3·06 (1·73-5·42; p=0·0001) for intermediate versus low risk in the primary analysis of the validation cohort. Estimated 3-year cancer-specific survival was 97·2% (95% CI 95·1-98·4; n=445 [41%]) for the low-risk group, 94·8% (91·7-96·7; n=339 [32%]) for the intermediate-risk group, and 77·6% (72·1-82·1; n=291 [27%]) for the high-risk group. The guideline-based risk grouping was observed to be less prognostic and informative (the low-risk group comprised only 142 [13%] of the 1075 patients). INTERPRETATION Integrating DoMore-v1-CRC and pathological staging markers provided a clinical decision support system that risk stratifies more accurately than its constituent elements, and identifies substantially more patients with stage II and III colorectal cancer with similarly good prognosis as the low-risk group in current guidelines. Avoiding adjuvant chemotherapy in these patients might be safe, and could reduce morbidity, mortality, and treatment costs. FUNDING The Research Council of Norway.
Collapse
Affiliation(s)
- Andreas Kleppe
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Department of Informatics, University of Oslo, Oslo, Norway
| | - Ole-Johan Skrede
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Department of Informatics, University of Oslo, Oslo, Norway
| | - Sepp De Raedt
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Department of Informatics, University of Oslo, Oslo, Norway
| | - Tarjei S Hveem
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Hanne A Askautrud
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Jørn E Jacobsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Department of Research and Development, Vestfold Hospital Trust, Tønsberg, Norway
| | - David N Church
- National Institute of Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Arild Nesbakken
- Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Colorectal Cancer Research Centre, Oslo, Norway
| | - Neil A Shepherd
- Gloucestershire Cellular Pathology Laboratory, Cheltenham General Hospital, Cheltenham, UK
| | - Marco Novelli
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Research Department of Pathology, University College London, London, UK
| | - Rachel Kerr
- Department of Oncology, University of Oxford, Oxford, UK
| | - Knut Liestøl
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Department of Informatics, University of Oslo, Oslo, Norway
| | - David J Kerr
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK
| | - Håvard E Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Department of Informatics, University of Oslo, Oslo, Norway; Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
106
|
Shmatko A, Ghaffari Laleh N, Gerstung M, Kather JN. Artificial intelligence in histopathology: enhancing cancer research and clinical oncology. NATURE CANCER 2022; 3:1026-1038. [PMID: 36138135 DOI: 10.1038/s43018-022-00436-4] [Citation(s) in RCA: 138] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 08/03/2022] [Indexed: 06/16/2023]
Abstract
Artificial intelligence (AI) methods have multiplied our capabilities to extract quantitative information from digital histopathology images. AI is expected to reduce workload for human experts, improve the objectivity and consistency of pathology reports, and have a clinical impact by extracting hidden information from routinely available data. Here, we describe how AI can be used to predict cancer outcome, treatment response, genetic alterations and gene expression from digitized histopathology slides. We summarize the underlying technologies and emerging approaches, noting limitations, including the need for data sharing and standards. Finally, we discuss the broader implications of AI in cancer research and oncology.
Collapse
Affiliation(s)
- Artem Shmatko
- Division of AI in Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
| | | | - Moritz Gerstung
- Division of AI in Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK.
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.
- Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
107
|
Diaz-Uriarte R, Gómez de Lope E, Giugno R, Fröhlich H, Nazarov PV, Nepomuceno-Chamorro IA, Rauschenberger A, Glaab E. Ten quick tips for biomarker discovery and validation analyses using machine learning. PLoS Comput Biol 2022; 18:e1010357. [PMID: 35951526 PMCID: PMC9371329 DOI: 10.1371/journal.pcbi.1010357] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Ramon Diaz-Uriarte
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (UAM-CSIC), Madrid, Spain
| | - Elisa Gómez de Lope
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg
| | - Rosalba Giugno
- Department of Computer Science, University of Verona, Verona, Italy
| | - Holger Fröhlich
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Centre for IT (b-it), Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Petr V. Nazarov
- Department of Cancer Research, Luxembourg Institute of Health, Strassen, Luxembourg
| | | | - Armin Rauschenberger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg
- * E-mail:
| |
Collapse
|
108
|
Xiong Y, Wang S, Wei H, Li H, Lv Y, Chi M, Su D, Lu Q, Yu Y, Zuo Y, Yang L. Deep learning-based transcription factor activity for stratification of breast cancer patients. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194838. [PMID: 35690313 DOI: 10.1016/j.bbagrm.2022.194838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/19/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Transcription factors directly bind to DNA and regulate the expression of the gene, causing epigenetic modification of the DNA. They often mediate epigenetic parameters of transcriptional and posttranscriptional mechanisms, and their expression activities can be used to characterize genomic aberrations in cancer cell. In this study, the activity profile of transcription factors inferred by VIPER algorithm. The autoencoder model was applied for compressing the transcription factor activity profile for obtaining more useful transformed features for stratifying patients into two different breast cancer subtypes. The deep learning-based subtypes exhibited superior prognostic value and yielded better risk-stratification than the transcription factor activity-based method. Importantly, according to transformed features, a deep neural network was constructed to predict the subtypes, and achieved the accuracy of 94.98% and area under the ROC curve of 0.9663, respectively. The proposed subtypes were found to be significantly associated with immune infiltration, tumor immunogenicity and so on. Furthermore, the ceRNA network was constructed for the breast cancer subtypes. Besides, 11 master regulators were found to be associated with patients in cluster 1. Given the robustness performance of our deep learning model over multiple breast cancer cohorts, we expected this model may be useful in the area of prognosis prediction and lead some possibility for personalized medicine in breast cancer patients.
Collapse
Affiliation(s)
- Yuqiang Xiong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Shiyuan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Haodong Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Hanshuang Li
- The State key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Yingli Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Meng Chi
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Dongqing Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Qianzi Lu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yao Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yongchun Zuo
- The State key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China; Digital College, Inner Mongolia Intelligent Union Big Data Academy, Inner Mingolia Wesure Date Technology Co., Ltd., Hohhot 010010, China.
| | - Lei Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
109
|
Medical domain knowledge in domain-agnostic generative AI. NPJ Digit Med 2022; 5:90. [PMID: 35817798 PMCID: PMC9273760 DOI: 10.1038/s41746-022-00634-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 06/15/2022] [Indexed: 11/25/2022] Open
|
110
|
Waljee AK, Weinheimer-Haus EM, Abubakar A, Ngugi AK, Siwo GH, Kwakye G, Singal AG, Rao A, Saini SD, Read AJ, Baker JA, Balis U, Opio CK, Zhu J, Saleh MN. Artificial intelligence and machine learning for early detection and diagnosis of colorectal cancer in sub-Saharan Africa. Gut 2022; 71:1259-1265. [PMID: 35418482 PMCID: PMC9177787 DOI: 10.1136/gutjnl-2022-327211] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/17/2022] [Indexed: 01/05/2023]
Affiliation(s)
- Akbar K Waljee
- Veterans Affairs Center for Clinical Management Research, Ann Arbor, Michigan, USA .,Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA.,Center for Global Health Equity, University of Michigan, Ann Arbor, Michigan, USA.,Michigan Integrated Center for Health Analytics and Medical Prediction (MiCHAMP), University of Michigan, Ann Arbor, Michigan, USA
| | - Eileen M Weinheimer-Haus
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA,Center for Global Health Equity, University of Michigan, Ann Arbor, Michigan, USA,Michigan Integrated Center for Health Analytics and Medical Prediction (MiCHAMP), University of Michigan, Ann Arbor, Michigan, USA
| | - Amina Abubakar
- Institute for Human Development, The Aga Khan University, Nairobi, Kenya
| | - Anthony K Ngugi
- Department of Population Health, The Aga Khan University, Nairobi, Kenya
| | - Geoffrey H Siwo
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA,Center for Global Health Equity, University of Michigan, Ann Arbor, Michigan, USA,Eck Institute for Global Health, University of Notre Dame, South Bend, Indiana, USA,Center for Research Computing, University of Notre Dame, South Bend, Indiana, USA
| | - Gifty Kwakye
- Department of Surgery, Division of Colorectal Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Amit G Singal
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA,Department of Internal Medicine, Division of Digestive and Liver Diseases, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Arvind Rao
- Michigan Integrated Center for Health Analytics and Medical Prediction (MiCHAMP), University of Michigan, Ann Arbor, Michigan, USA,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA,Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sameer D Saini
- Veterans Affairs Center for Clinical Management Research, Ann Arbor, Michigan, USA,Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew J Read
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA,Michigan Integrated Center for Health Analytics and Medical Prediction (MiCHAMP), University of Michigan, Ann Arbor, Michigan, USA
| | - Jessica A Baker
- Veterans Affairs Center for Clinical Management Research, Ann Arbor, Michigan, USA,Center for Global Health Equity, University of Michigan, Ann Arbor, Michigan, USA,Michigan Integrated Center for Health Analytics and Medical Prediction (MiCHAMP), University of Michigan, Ann Arbor, Michigan, USA
| | - Ulysses Balis
- Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Christopher K Opio
- Department of Medicine, Aga Khan University Hospital Nairobi, Nairobi, Kenya
| | - Ji Zhu
- Center for Global Health Equity, University of Michigan, Ann Arbor, Michigan, USA,Michigan Integrated Center for Health Analytics and Medical Prediction (MiCHAMP), University of Michigan, Ann Arbor, Michigan, USA,Department of Statistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Mansoor N Saleh
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama, USA,Department of Hematology-Oncology, Aga Khan University Hospital Nairobi, Nairobi, Kenya
| |
Collapse
|
111
|
Daveau RS, Law I, Henriksen OM, Hasselbalch SG, Andersen UB, Anderberg L, Højgaard L, Andersen FL, Ladefoged CN. Deep learning based low-activity PET reconstruction of [ 11C]PiB and [ 18F]FE-PE2I in neurodegenerative disorders. Neuroimage 2022; 259:119412. [PMID: 35753592 DOI: 10.1016/j.neuroimage.2022.119412] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
PURPOSE Positron Emission Tomography (PET) can support a diagnosis of neurodegenerative disorder by identifying disease-specific pathologies. Our aim was to investigate the feasibility of using activity reduction in clinical [18F]FE-PE2I and [11C]PiB PET/CT scans, simulating low injected activity or scanning time reduction, in combination with AI-assisted denoising. METHODS A total of 162 patients with clinically uncertain Alzheimer's disease underwent amyloid [11C]PiB PET/CT and 509 patients referred for clinically uncertain Parkinson's disease underwent dopamine transporter (DAT) [18F]FE-PE2I PET/CT. Simulated low-activity data were obtained by random sampling of 5% of the events from the list-mode file and a 5% time window extraction in the middle of the scan. A three-dimensional convolutional neural network (CNN) was trained to denoise the resulting PET images for each disease cohort. RESULTS Noise reduction of low-activity PET images was successful for both cohorts using 5% of the original activity with improvement in visual quality and all similarity metrics with respect to the ground-truth images. Clinically relevant metrics extracted from the low-activity images deviated <2% compared to ground-truth values, which were not significantly changed when extracting the metrics from the denoised images. CONCLUSION The presented models were based on the same network architecture and proved to be a robust tool for denoising brain PET images with two widely different tracer distributions (delocalized, ([11C]PiB, and highly localized, [18F]FE-PE2I). This broad and robust application makes the presented network a good choice for improving the quality of brain images to the level of the standard-activity images without degrading clinical metric extraction. This will allow for reduced dose or scan time in PET/CT to be implemented clinically.
Collapse
Affiliation(s)
- Raphaël S Daveau
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, University of Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, University of Copenhagen, Denmark
| | - Otto Mølby Henriksen
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, University of Copenhagen, Denmark
| | | | - Ulrik Bjørn Andersen
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, University of Copenhagen, Denmark
| | - Lasse Anderberg
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, University of Copenhagen, Denmark
| | - Liselotte Højgaard
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, University of Copenhagen, Denmark
| | - Flemming Littrup Andersen
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, University of Copenhagen, Denmark
| | - Claes Nøhr Ladefoged
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, University of Copenhagen, Denmark.
| |
Collapse
|
112
|
Security Evaluation of Financial and Insurance and Ruin Probability Analysis Integrating Deep Learning Models. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:1857100. [PMID: 35720881 PMCID: PMC9200529 DOI: 10.1155/2022/1857100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/11/2022] [Accepted: 05/21/2022] [Indexed: 11/17/2022]
Abstract
To ensure safe development of the financial and insurance industry and promote the continuous growth of the social economy, the theory and its role of deep learning are firstly analyzed. Secondly, the security of financial and insurance and bankruptcy probability are discussed. Finally, an analytical model of the security bankruptcy probability of financial and insurance is designed through a deep learning model, and the model is evaluated comprehensively. The research results manifest that first, the designed security evaluation of the financial and insurance industry based on the deep learning and bankruptcy probability analysis model not only has strong learning ability but also can effectively reduce its own calculation error through short-time learning. Then, by comparing with other models, it is found that the designed model has a stronger ability to control various errors than other models, and the overall error rate of the model can be reduced to about 20%. At last, the data training indicates that the model designed by the deep learning method can accurately and effectively predict the basic situation of the financial and insurance industry, the minimum error can reach 0, and the highest is only about 3. The research provides a technical reference for the development of the financial and insurance industry and contributes to the prosperity of the social economy.
Collapse
|
113
|
Letters to the Editor. J R Coll Physicians Edinb 2022; 52:80-85. [DOI: 10.1177/14782715221089063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
114
|
George RS, Htoo A, Cheng M, Masterson TM, Huang K, Adra N, Kaimakliotis HZ, Akgul M, Cheng L. Artificial intelligence in prostate cancer: Definitions, current research, and future directions. Urol Oncol 2022; 40:262-270. [PMID: 35430139 DOI: 10.1016/j.urolonc.2022.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/19/2022] [Accepted: 03/10/2022] [Indexed: 12/24/2022]
Abstract
Multiple novel modalities tasking artificial intelligence based computational pathology applications and integrating other variables, such as risk factors, tumor microenvironment, genomic testing data, laboratory findings, clinical history, and radiology findings, will improve diagnostic consistency and generate a synergistic diagnostic workflow. In this article, we present the concise and contemporary review on the utilization of artificial intelligence in prostate cancer and identify areas for possible future applications.
Collapse
Affiliation(s)
- Rose S George
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY
| | - Arkar Htoo
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY
| | - Michael Cheng
- Department of Medicine, Indianapolis, Indianapolis, IN
| | | | - Kun Huang
- Department of Medicine, Indianapolis, Indianapolis, IN; Department of Biostatistics and Health Data Science, Indianapolis, IN; Regenstrief Institute, Indianapolis, IN
| | - Nabil Adra
- Department of Medicine, Indianapolis, Indianapolis, IN; Department of Urology, Indianapolis, IN
| | | | - Mahmut Akgul
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY.
| | - Liang Cheng
- Department of Urology, Indianapolis, IN; Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN.
| |
Collapse
|
115
|
Raza R, Ijaz Bajwa U, Mehmood Y, Waqas Anwar M, Hassan Jamal M. dResU-Net: 3D deep residual U-Net based brain tumor segmentation from multimodal MRI. Biomed Signal Process Control 2022. [DOI: 10.1016/j.bspc.2022.103861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
116
|
Barragán-Montero A, Bibal A, Dastarac MH, Draguet C, Valdés G, Nguyen D, Willems S, Vandewinckele L, Holmström M, Löfman F, Souris K, Sterpin E, Lee JA. Towards a safe and efficient clinical implementation of machine learning in radiation oncology by exploring model interpretability, explainability and data-model dependency. Phys Med Biol 2022; 67:10.1088/1361-6560/ac678a. [PMID: 35421855 PMCID: PMC9870296 DOI: 10.1088/1361-6560/ac678a] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/14/2022] [Indexed: 01/26/2023]
Abstract
The interest in machine learning (ML) has grown tremendously in recent years, partly due to the performance leap that occurred with new techniques of deep learning, convolutional neural networks for images, increased computational power, and wider availability of large datasets. Most fields of medicine follow that popular trend and, notably, radiation oncology is one of those that are at the forefront, with already a long tradition in using digital images and fully computerized workflows. ML models are driven by data, and in contrast with many statistical or physical models, they can be very large and complex, with countless generic parameters. This inevitably raises two questions, namely, the tight dependence between the models and the datasets that feed them, and the interpretability of the models, which scales with its complexity. Any problems in the data used to train the model will be later reflected in their performance. This, together with the low interpretability of ML models, makes their implementation into the clinical workflow particularly difficult. Building tools for risk assessment and quality assurance of ML models must involve then two main points: interpretability and data-model dependency. After a joint introduction of both radiation oncology and ML, this paper reviews the main risks and current solutions when applying the latter to workflows in the former. Risks associated with data and models, as well as their interaction, are detailed. Next, the core concepts of interpretability, explainability, and data-model dependency are formally defined and illustrated with examples. Afterwards, a broad discussion goes through key applications of ML in workflows of radiation oncology as well as vendors' perspectives for the clinical implementation of ML.
Collapse
Affiliation(s)
- Ana Barragán-Montero
- Molecular Imaging, Radiation and Oncology (MIRO) Laboratory, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium
| | - Adrien Bibal
- PReCISE, NaDI Institute, Faculty of Computer Science, UNamur and CENTAL, ILC, UCLouvain, Belgium
| | - Margerie Huet Dastarac
- Molecular Imaging, Radiation and Oncology (MIRO) Laboratory, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium
| | - Camille Draguet
- Molecular Imaging, Radiation and Oncology (MIRO) Laboratory, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium
- Department of Oncology, Laboratory of Experimental Radiotherapy, KU Leuven, Belgium
| | - Gilmer Valdés
- Department of Radiation Oncology, Department of Epidemiology and Biostatistics, University of California, San Francisco, United States of America
| | - Dan Nguyen
- Medical Artificial Intelligence and Automation (MAIA) Laboratory, Department of Radiation Oncology, UT Southwestern Medical Center, United States of America
| | - Siri Willems
- ESAT/PSI, KU Leuven Belgium & MIRC, UZ Leuven, Belgium
| | | | | | | | - Kevin Souris
- Molecular Imaging, Radiation and Oncology (MIRO) Laboratory, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium
| | - Edmond Sterpin
- Molecular Imaging, Radiation and Oncology (MIRO) Laboratory, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium
- Department of Oncology, Laboratory of Experimental Radiotherapy, KU Leuven, Belgium
| | - John A Lee
- Molecular Imaging, Radiation and Oncology (MIRO) Laboratory, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium
| |
Collapse
|
117
|
Swarm learning for decentralized artificial intelligence in cancer histopathology. Nat Med 2022; 28:1232-1239. [PMID: 35469069 PMCID: PMC9205774 DOI: 10.1038/s41591-022-01768-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/02/2022] [Indexed: 02/07/2023]
Abstract
Artificial intelligence (AI) can predict the presence of molecular alterations directly from routine histopathology slides. However, training robust AI systems requires large datasets for which data collection faces practical, ethical and legal obstacles. These obstacles could be overcome with swarm learning (SL), in which partners jointly train AI models while avoiding data transfer and monopolistic data governance. Here, we demonstrate the successful use of SL in large, multicentric datasets of gigapixel histopathology images from over 5,000 patients. We show that AI models trained using SL can predict BRAF mutational status and microsatellite instability directly from hematoxylin and eosin (H&E)-stained pathology slides of colorectal cancer. We trained AI models on three patient cohorts from Northern Ireland, Germany and the United States, and validated the prediction performance in two independent datasets from the United Kingdom. Our data show that SL-trained AI models outperform most locally trained models, and perform on par with models that are trained on the merged datasets. In addition, we show that SL-based AI models are data efficient. In the future, SL can be used to train distributed AI models for any histopathology image analysis task, eliminating the need for data transfer. A decentralized, privacy-preserving machine learning framework used to train a clinically relevant AI system identifies actionable molecular alterations in patients with colorectal cancer by use of routine histopathology slides collected in real-world settings.
Collapse
|
118
|
Qi W, Chen R, Chen M, Zhao M, Wang M. Evaluation Analysis of the Nephrotoxicity of Tripterygium wilfordii Preparations with CONSORT Harms Statement Based on Deep Learning. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:5054932. [PMID: 35432821 PMCID: PMC9010145 DOI: 10.1155/2022/5054932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 03/12/2022] [Indexed: 11/17/2022]
Abstract
In this paper, the safety of Tripterygium wilfordii polyglycoside (TW) preparation was evaluated by combining literature research and evidence-based evaluation research, so as to provide evidence-based safety information of Tripterygium wilfordii polyglycoside preparation (nephroptosis) for government decision making and clinical application. In this paper, we propose a network structure inspired by the LSTM gate mechanism. All the research methods of the included references are evaluated by internationally recognized evaluation tools or standards. Prevalence was analyzed according to the type of intervention (e.g., time of administration) and route of administration. The results of this experiment provide methods and suggestions for the evaluation of traditional Chinese medicine nephroptosis in the future.
Collapse
Affiliation(s)
- Wuqiang Qi
- Pharmacy Department of Baojl, Hospital of Traditional Chinese Medicine, Baoji, Shaanxi 721001, China
| | - Rui Chen
- Pharmacy Department of Baojl, Hospital of Traditional Chinese Medicine, Baoji, Shaanxi 721001, China
| | - Minghui Chen
- Pharmacy Department of Hanzhong, People's Hospital, Hanzhong, Shaanxi 723000, China
| | - Meng Zhao
- Pharmacy Department of Xi'an, Central Hospital, Xi'an, Shaanxi 710003, China
| | - Mingzhao Wang
- Pharmacy Department of Affiliated Hospital of Shaanxi, University of Chinese Medicine, Xianyang, Shaanxi 712000, China
| |
Collapse
|
119
|
Area under the curve may hide poor generalisation to external datasets. ESMO Open 2022; 7:100429. [PMID: 35397433 PMCID: PMC9006654 DOI: 10.1016/j.esmoop.2022.100429] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 11/23/2022] Open
|
120
|
Marshall JL, Peshkin BN, Yoshino T, Vowinckel J, Danielsen HE, Melino G, Tsamardinos I, Haudenschild C, Kerr DJ, Sampaio C, Rha SY, FitzGerald KT, Holland EC, Gallagher D, Garcia-Foncillas J, Juhl H. The Essentials of Multiomics. Oncologist 2022; 27:272-284. [PMID: 35380712 PMCID: PMC8982374 DOI: 10.1093/oncolo/oyab048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
Within the last decade, the science of molecular testing has evolved from single gene and single protein analysis to broad molecular profiling as a standard of care, quickly transitioning from research to practice. Terms such as genomics, transcriptomics, proteomics, circulating omics, and artificial intelligence are now commonplace, and this rapid evolution has left us with a significant knowledge gap within the medical community. In this paper, we attempt to bridge that gap and prepare the physician in oncology for multiomics, a group of technologies that have gone from looming on the horizon to become a clinical reality. The era of multiomics is here, and we must prepare ourselves for this exciting new age of cancer medicine.
Collapse
Affiliation(s)
- John L Marshall
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Beth N Peshkin
- Georgetown University, Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | | | | | - Håvard E Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Radiumhospitalet, Montebello, Oslo, Norway
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Ioannis Tsamardinos
- JADBio Gnosis DA, N. Plastira 100, Science and Technology Park of Crete and Institute of Applied and Computational Mathematics, Foundation for Research and Technology Hellas, Heraklion, GR, Greece
| | | | - David J Kerr
- Nuffield Division of Clinical and Laboratory Sciences, Level 4, Academic Block, John Radcliffe Infirmary, Headington, Oxford, UK
| | | | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University College of Medicine, Seodaemun-Ku, Seoul, Korea
| | - Kevin T FitzGerald
- Department of Medical Humanities in the School of Medicine, Creighton University, Omaha, NE, USA
| | - Eric C Holland
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - David Gallagher
- St. James’s Hospital/Trinity College Dublin, St. Raphael’s House, Dublin, Ireland
| | - Jesus Garcia-Foncillas
- Cancer Institute, Fundacion Jimenez Diaz University Hospital, Autonomous University, Madrid, Spain
| | | |
Collapse
|
121
|
Heinz CN, Echle A, Foersch S, Bychkov A, Kather JN. The future of artificial intelligence in digital pathology - results of a survey across stakeholder groups. Histopathology 2022; 80:1121-1127. [PMID: 35373378 DOI: 10.1111/his.14659] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/17/2022] [Accepted: 04/02/2022] [Indexed: 11/30/2022]
Abstract
AIMS Artificial intelligence (AI) provides a powerful tool to extract information from digitized histopathology whole slide images. In the last five years, academic and commercial actors have developed new technical solutions for a diverse set of tasks, including tissue segmentation, cell detection, mutation prediction, prognostication and prediction of treatment response. In the light of limited overall resources, it is presently unclear for researchers, practitioners and policymakers which of these topics are stable enough for clinical use in the near future and which topics are still experimental, but worth investing time and effort into. METHODS To identify potentially promising applications of AI in pathology, we performed an anonymous online survey of 75 computational pathology domain experts from academia and industry. Participants enrolled in 2021 were queried about their subjective opinion on promising and appealing sub-fields of computational pathology with a focus on solid tumors. RESULTS The results of this survey indicate that the prediction of treatment response directly from routine pathology slides is regarded as the most promising future application. This item was ranked highest in the overall analysis and in sub-groups by age and professional background. Furthermore, prediction of genetic alterations, gene expression and survival directly from routine pathology images scored consistently high across subgroups. CONCLUSIONS Together, these data demonstrate a possible direction for the development of computational pathology systems in clinical, academic and industrial research in the near future.
Collapse
Affiliation(s)
- Céline N Heinz
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Amelie Echle
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Sebastian Foersch
- Department of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Andrey Bychkov
- Department of Pathology, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.,Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.,Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| |
Collapse
|
122
|
He X, Lee B, Jiang Y. Extracellular matrix in cancer progression and therapy. MEDICAL REVIEW (2021) 2022; 2:125-139. [PMID: 37724245 PMCID: PMC10471113 DOI: 10.1515/mr-2021-0028] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/31/2022] [Indexed: 09/20/2023]
Abstract
The tumor ecosystem with heterogeneous cellular compositions and the tumor microenvironment has increasingly become the focus of cancer research in recent years. The extracellular matrix (ECM), the major component of the tumor microenvironment, and its interactions with the tumor cells and stromal cells have also enjoyed tremendously increased attention. Like the other components of the tumor microenvironment, the ECM in solid tumors differs significantly from that in normal organs and tissues. We review recent studies of the complex roles the tumor ECM plays in cancer progression, from tumor initiation, growth to angiogenesis and invasion. We highlight that the biomolecular, biophysical, and mechanochemical interactions between the ECM and cells not only regulate the steps of cancer progression, but also affect the efficacy of systemic cancer treatment. We further discuss the strategies to target and modify the tumor ECM to improve cancer therapy.
Collapse
Affiliation(s)
- Xiuxiu He
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Byoungkoo Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
123
|
Brozgol E, Kumar P, Necula D, Bronshtein-Berger I, Lindner M, Medalion S, Twito L, Shapira Y, Gondra H, Barshack I, Garini Y. Cancer detection from stained biopsies using high-speed spectral imaging. BIOMEDICAL OPTICS EXPRESS 2022; 13:2503-2515. [PMID: 35519262 PMCID: PMC9045910 DOI: 10.1364/boe.445782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/31/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023]
Abstract
The escalating demand for diagnosing pathological biopsies requires the procedures to be expedited and automated. The existing imaging systems for measuring biopsies only measure color, and even though a lot of effort is invested in deep learning analysis, there are still serious challenges regarding the performance and validity of the data for the intended medical setting. We developed a system that rapidly acquires spectral images from biopsies, followed by spectral classification algorithms. The spectral information is remarkably more informative than the color information, and leads to very high accuracy in identifying cancer cells, as tested on tens of cancer cases. This was improved even more by using artificial intelligence algorithms that required a rather small training set, indicating the high level of information that exists in the spectral images. The most important spectral differences are observed in the nucleus and they are related to aneuploidy in tumor cells. Rapid spectral imaging measurement therefore can bridge the gap in the machine-aided diagnostics of whole biopsies, thus improving patient care, and expediting the treatment procedure.
Collapse
Affiliation(s)
- Eugene Brozgol
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
- Contributed equally
| | - Pramod Kumar
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
- Contributed equally
| | - Daniela Necula
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
| | | | - Moshe Lindner
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
| | | | - Lee Twito
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
| | - Yotam Shapira
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
| | - Helena Gondra
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Equal supervision
| | - Yuval Garini
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
- Biomedical Engineering Faculty, Technion − Israel Institute of Technology, Haifa, Israel
- Equal supervision
| |
Collapse
|
124
|
Echle A, Ghaffari Laleh N, Quirke P, Grabsch HI, Muti HS, Saldanha OL, Brockmoeller SF, van den Brandt PA, Hutchins GGA, Richman SD, Horisberger K, Galata C, Ebert MP, Eckardt M, Boutros M, Horst D, Reissfelder C, Alwers E, Brinker TJ, Langer R, Jenniskens JCA, Offermans K, Mueller W, Gray R, Gruber SB, Greenson JK, Rennert G, Bonner JD, Schmolze D, Chang-Claude J, Brenner H, Trautwein C, Boor P, Jaeger D, Gaisa NT, Hoffmeister M, West NP, Kather JN. Artificial intelligence for detection of microsatellite instability in colorectal cancer-a multicentric analysis of a pre-screening tool for clinical application. ESMO Open 2022; 7:100400. [PMID: 35247870 PMCID: PMC9058894 DOI: 10.1016/j.esmoop.2022.100400] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Microsatellite instability (MSI)/mismatch repair deficiency (dMMR) is a key genetic feature which should be tested in every patient with colorectal cancer (CRC) according to medical guidelines. Artificial intelligence (AI) methods can detect MSI/dMMR directly in routine pathology slides, but the test performance has not been systematically investigated with predefined test thresholds. METHOD We trained and validated AI-based MSI/dMMR detectors and evaluated predefined performance metrics using nine patient cohorts of 8343 patients across different countries and ethnicities. RESULTS Classifiers achieved clinical-grade performance, yielding an area under the receiver operating curve (AUROC) of up to 0.96 without using any manual annotations. Subsequently, we show that the AI system can be applied as a rule-out test: by using cohort-specific thresholds, on average 52.73% of tumors in each surgical cohort [total number of MSI/dMMR = 1020, microsatellite stable (MSS)/ proficient mismatch repair (pMMR) = 7323 patients] could be identified as MSS/pMMR with a fixed sensitivity at 95%. In an additional cohort of N = 1530 (MSI/dMMR = 211, MSS/pMMR = 1319) endoscopy biopsy samples, the system achieved an AUROC of 0.89, and the cohort-specific threshold ruled out 44.12% of tumors with a fixed sensitivity at 95%. As a more robust alternative to cohort-specific thresholds, we showed that with a fixed threshold of 0.25 for all the cohorts, we can rule-out 25.51% in surgical specimens and 6.10% in biopsies. INTERPRETATION When applied in a clinical setting, this means that the AI system can rule out MSI/dMMR in a quarter (with global thresholds) or half of all CRC patients (with local fine-tuning), thereby reducing cost and turnaround time for molecular profiling.
Collapse
Affiliation(s)
- A Echle
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - N Ghaffari Laleh
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - P Quirke
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - H I Grabsch
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK; Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - H S Muti
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - O L Saldanha
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - S F Brockmoeller
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - P A van den Brandt
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - G G A Hutchins
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - S D Richman
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - K Horisberger
- Department of Abdominal and Transplantation Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - C Galata
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Division of Thoracic Surgery, Academic Thoracic Center Mainz, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - M P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Institute for Innate Immunoscience (MI3) and Clinical Cooperation Unit Healthy Metabolism, Center of Preventive Medicine and Digital Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - M Eckardt
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - M Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - D Horst
- Institut für Pathologie Charité, Berlin, Germany
| | - C Reissfelder
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - E Alwers
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - T J Brinker
- Digital Biomarkers for Oncology Group, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - R Langer
- Institute of Pathology, Inselspital, University of Bern, Bern, Switzerland
| | - J C A Jenniskens
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - K Offermans
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - W Mueller
- Gemeinschaftspraxis Pathologie, Starnberg, Germany
| | - R Gray
- Clinical Trial Service Unit, University of Oxford, Oxford, UK
| | - S B Gruber
- Center for Precision Medicine and Department of Medical Oncology, City of Hope National Medical Center, Duarte, USA
| | - J K Greenson
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, USA
| | - G Rennert
- Department of Community Medicine & Epidemiology, Lady Davis Carmel Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Steve and Cindy Rasmussen Institute for Genomic Medicine, Lady Davis Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - J D Bonner
- Center for Precision Medicine and Department of Medical Oncology, City of Hope National Medical Center, Duarte, USA
| | - D Schmolze
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, USA
| | - J Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Cancer Epidemiology Group, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - H Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - P Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany; Department of Nephrology and Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - D Jaeger
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - N T Gaisa
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - M Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - N P West
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - J N Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany; Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
125
|
Cifci D, Foersch S, Kather JN. Artificial intelligence to identify genetic alterations in conventional histopathology. J Pathol 2022; 257:430-444. [PMID: 35342954 DOI: 10.1002/path.5898] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 11/10/2022]
Abstract
Precision oncology relies on the identification of targetable molecular alterations in tumor tissues. In many tumor types, a limited set of molecular tests is currently part of standard diagnostic workflows. However, universal testing for all targetable alterations, especially rare ones, is limited by the cost and availability of molecular assays. From 2017 to 2021, multiple studies have shown that artificial intelligence (AI) methods can predict the probability of specific genetic alterations directly from conventional hematoxylin and eosin (H&E) tissue slides. Although these methods are currently less accurate than gold-standard testing (e.g. immunohistochemistry, polymerase chain reaction or next-generation sequencing), they could be used as pre-screening tools to reduce the workload of genetic analyses. In this systematic literature review, we summarize the state of the art in predicting molecular alterations from H&E using AI. We found that AI methods perform reasonably well across multiple tumor types, although few algorithms have been broadly validated. In addition, we found that genetic alterations in FGFR, IDH, PIK3CA, BRAF, TP53 and DNA repair pathways are predictable from H&E in multiple tumor types, while many other genetic alterations have rarely been investigated or were only poorly predictable. Finally, we discuss the next steps for the implementation of AI-based surrogate tests in diagnostic workflows. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Didem Cifci
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.,Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.,Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
126
|
Afshar P, Rafiee MJ, Naderkhani F, Heidarian S, Enshaei N, Oikonomou A, Babaki Fard F, Anconina R, Farahani K, Plataniotis KN, Mohammadi A. Human-level COVID-19 diagnosis from low-dose CT scans using a two-stage time-distributed capsule network. Sci Rep 2022; 12:4827. [PMID: 35318368 PMCID: PMC8940967 DOI: 10.1038/s41598-022-08796-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/01/2022] [Indexed: 01/01/2023] Open
Abstract
Reverse transcription-polymerase chain reaction is currently the gold standard in COVID-19 diagnosis. It can, however, take days to provide the diagnosis, and false negative rate is relatively high. Imaging, in particular chest computed tomography (CT), can assist with diagnosis and assessment of this disease. Nevertheless, it is shown that standard dose CT scan gives significant radiation burden to patients, especially those in need of multiple scans. In this study, we consider low-dose and ultra-low-dose (LDCT and ULDCT) scan protocols that reduce the radiation exposure close to that of a single X-ray, while maintaining an acceptable resolution for diagnosis purposes. Since thoracic radiology expertise may not be widely available during the pandemic, we develop an Artificial Intelligence (AI)-based framework using a collected dataset of LDCT/ULDCT scans, to study the hypothesis that the AI model can provide human-level performance. The AI model uses a two stage capsule network architecture and can rapidly classify COVID-19, community acquired pneumonia (CAP), and normal cases, using LDCT/ULDCT scans. Based on a cross validation, the AI model achieves COVID-19 sensitivity of [Formula: see text], CAP sensitivity of [Formula: see text], normal cases sensitivity (specificity) of [Formula: see text], and accuracy of [Formula: see text]. By incorporating clinical data (demographic and symptoms), the performance further improves to COVID-19 sensitivity of [Formula: see text], CAP sensitivity of [Formula: see text], normal cases sensitivity (specificity) of [Formula: see text] , and accuracy of [Formula: see text]. The proposed AI model achieves human-level diagnosis based on the LDCT/ULDCT scans with reduced radiation exposure. We believe that the proposed AI model has the potential to assist the radiologists to accurately and promptly diagnose COVID-19 infection and help control the transmission chain during the pandemic.
Collapse
Affiliation(s)
- Parnian Afshar
- Concordia Institute for Information Systems Engineering (CIISE), Concordia University, Montreal, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Canada
| | - Moezedin Javad Rafiee
- Department of Medicine and Diagnostic Radiology, McGill University Health Center-Research Institute, Montreal, QC, Canada
| | - Farnoosh Naderkhani
- Concordia Institute for Information Systems Engineering (CIISE), Concordia University, Montreal, Canada
| | - Shahin Heidarian
- Department of Electrical and Computer Engineering, Concordia University, Montreal, QC, Canada
| | - Nastaran Enshaei
- Concordia Institute for Information Systems Engineering (CIISE), Concordia University, Montreal, Canada
| | - Anastasia Oikonomou
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | | | - Reut Anconina
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Keyvan Farahani
- Center for Biomedical Informatics and Information Technology, National Cancer Institute (NCI), Rockville, MD, USA
| | | | - Arash Mohammadi
- Concordia Institute for Information Systems Engineering (CIISE), Concordia University, Montreal, Canada.
| |
Collapse
|
127
|
Assessing radiomics feature stability with simulated CT acquisitions. Sci Rep 2022; 12:4732. [PMID: 35304508 PMCID: PMC8933485 DOI: 10.1038/s41598-022-08301-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 03/03/2022] [Indexed: 11/29/2022] Open
Abstract
Medical imaging quantitative features had once disputable usefulness in clinical studies. Nowadays, advancements in analysis techniques, for instance through machine learning, have enabled quantitative features to be progressively useful in diagnosis and research. Tissue characterisation is improved via the “radiomics” features, whose extraction can be automated. Despite the advances, stability of quantitative features remains an important open problem. As features can be highly sensitive to variations of acquisition details, it is not trivial to quantify stability and efficiently select stable features. In this work, we develop and validate a Computed Tomography (CT) simulator environment based on the publicly available ASTRA toolbox (www.astra-toolbox.com). We show that the variability, stability and discriminative power of the radiomics features extracted from the virtual phantom images generated by the simulator are similar to those observed in a tandem phantom study. Additionally, we show that the variability is matched between a multi-center phantom study and simulated results. Consequently, we demonstrate that the simulator can be utilised to assess radiomics features’ stability and discriminative power.
Collapse
|
128
|
Schwendicke F, Mertens S, Cantu AG, Chaurasia A, Meyer-Lueckel H, Krois J. Cost-effectiveness of AI for Caries Detection: Randomized Trial. J Dent 2022; 119:104080. [PMID: 35245626 DOI: 10.1016/j.jdent.2022.104080] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVES We assessed the cost-effectiveness of AI-supported detection of proximal caries in a randomized controlled clustered cross-over superiority trial. METHODS Twenty-three dentists were sampled to assess 20 bitewings; 10 were randomly evaluated supported by an AI-based software (dentalXrai Pro 1.0.4, dentalXrai Ltd, Berlin, Germany) and the other 10 without AI support. The reference test had been established by four independent experts and an additional review. We evaluated the proportion of true and false positive and negative detections and the treatment decisions assigned to each detection (non-invasive, micro-invasive, invasive). Cost-effectiveness was assessed using a mixed public-private-payer perspective in German healthcare. Using the accuracy and treatment decision data from the trial, a Markov simulation model was populated and posterior permanent teeth in initially 31-years old individuals followed over their lifetime. The model allowed extrapolation from the initial detection and therapy to treatment success, re-treatments and, eventually, tooth loss and replacement, capturing long-term effectiveness (tooth retention) and costs (cumulative in Euro). Costs were estimated using the German public and private fee catalogues. Monte-Carlo microsimulations were used and incremental cost-effectiveness at different willingness-to-pay ceiling thresholds assessed. RESULTS In the trial, AI-supported detection was significantly more sensitive than detection without AI. However, in the AI group, lesions were more often treated invasively. As a result, AI and no AI showed identical effectiveness (tooth retention for a mean (2.5-97.5%) 49 (48-51)) and nearly identical costs (AI: 330 (250-409) Euro, no AI: 330 (248-410) Euro). 41% simulations found AI and 43% no AI to be more cost-effective. The resulting cost-effectiveness remained uncertain regardless of a payer's willingness-to-pay. CONCLUSIONS Higher accuracy of AI did not lead to higher cost-effectiveness, as more invasive treatment approaches generated costs and diminished possible effectiveness advantages. CLINICAL SIGNIFICANCE The cost-effectiveness of AI could be improved by supporting not only caries detection, but also subsequent management.
Collapse
Affiliation(s)
- Falk Schwendicke
- Department of Oral Diagnostics, Digital Health and Health Services Research, Charité - Universitätsmedizin Berlin, Germany.
| | - Sarah Mertens
- Department of Oral Diagnostics, Digital Health and Health Services Research, Charité - Universitätsmedizin Berlin, Germany
| | - Anselmo Garcia Cantu
- Department of Oral Diagnostics, Digital Health and Health Services Research, Charité - Universitätsmedizin Berlin, Germany
| | | | | | - Joachim Krois
- Department of Oral Diagnostics, Digital Health and Health Services Research, Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
129
|
Deep learning in image-based breast and cervical cancer detection: a systematic review and meta-analysis. NPJ Digit Med 2022; 5:19. [PMID: 35169217 PMCID: PMC8847584 DOI: 10.1038/s41746-022-00559-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Accurate early detection of breast and cervical cancer is vital for treatment success. Here, we conduct a meta-analysis to assess the diagnostic performance of deep learning (DL) algorithms for early breast and cervical cancer identification. Four subgroups are also investigated: cancer type (breast or cervical), validation type (internal or external), imaging modalities (mammography, ultrasound, cytology, or colposcopy), and DL algorithms versus clinicians. Thirty-five studies are deemed eligible for systematic review, 20 of which are meta-analyzed, with a pooled sensitivity of 88% (95% CI 85–90%), specificity of 84% (79–87%), and AUC of 0.92 (0.90–0.94). Acceptable diagnostic performance with analogous DL algorithms was highlighted across all subgroups. Therefore, DL algorithms could be useful for detecting breast and cervical cancer using medical imaging, having equivalent performance to human clinicians. However, this tentative assertion is based on studies with relatively poor designs and reporting, which likely caused bias and overestimated algorithm performance. Evidence-based, standardized guidelines around study methods and reporting are required to improve the quality of DL research.
Collapse
|
130
|
Nam D, Chapiro J, Paradis V, Seraphin TP, Kather JN. Artificial intelligence in liver diseases: improving diagnostics, prognostics and response prediction. JHEP REPORTS : INNOVATION IN HEPATOLOGY 2022; 4:100443. [PMID: 35243281 PMCID: PMC8867112 DOI: 10.1016/j.jhepr.2022.100443] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/26/2021] [Accepted: 01/11/2022] [Indexed: 12/18/2022]
Abstract
Clinical routine in hepatology involves the diagnosis and treatment of a wide spectrum of metabolic, infectious, autoimmune and neoplastic diseases. Clinicians integrate qualitative and quantitative information from multiple data sources to make a diagnosis, prognosticate the disease course, and recommend a treatment. In the last 5 years, advances in artificial intelligence (AI), particularly in deep learning, have made it possible to extract clinically relevant information from complex and diverse clinical datasets. In particular, histopathology and radiology image data contain diagnostic, prognostic and predictive information which AI can extract. Ultimately, such AI systems could be implemented in clinical routine as decision support tools. However, in the context of hepatology, this requires further large-scale clinical validation and regulatory approval. Herein, we summarise the state of the art in AI in hepatology with a particular focus on histopathology and radiology data. We present a roadmap for the further development of novel biomarkers in hepatology and outline critical obstacles which need to be overcome.
Collapse
|
131
|
Boehm KM, Khosravi P, Vanguri R, Gao J, Shah SP. Harnessing multimodal data integration to advance precision oncology. Nat Rev Cancer 2022; 22:114-126. [PMID: 34663944 PMCID: PMC8810682 DOI: 10.1038/s41568-021-00408-3] [Citation(s) in RCA: 169] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
Advances in quantitative biomarker development have accelerated new forms of data-driven insights for patients with cancer. However, most approaches are limited to a single mode of data, leaving integrated approaches across modalities relatively underdeveloped. Multimodal integration of advanced molecular diagnostics, radiological and histological imaging, and codified clinical data presents opportunities to advance precision oncology beyond genomics and standard molecular techniques. However, most medical datasets are still too sparse to be useful for the training of modern machine learning techniques, and significant challenges remain before this is remedied. Combined efforts of data engineering, computational methods for analysis of heterogeneous data and instantiation of synergistic data models in biomedical research are required for success. In this Perspective, we offer our opinions on synthesizing complementary modalities of data with emerging multimodal artificial intelligence methods. Advancing along this direction will result in a reimagined class of multimodal biomarkers to propel the field of precision oncology in the coming decade.
Collapse
Affiliation(s)
- Kevin M Boehm
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pegah Khosravi
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rami Vanguri
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jianjiong Gao
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sohrab P Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
132
|
PrACTiC: A Predictive Algorithm for Chemoradiotherapy-Induced Cytopenia in Glioblastoma Patients. JOURNAL OF ONCOLOGY 2022; 2022:1438190. [PMID: 35111223 PMCID: PMC8803420 DOI: 10.1155/2022/1438190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/06/2022] [Indexed: 11/18/2022]
Abstract
Background Chemotherapy-induced cytopenia is the most frequent side effect of chemoradiotherapy in glioblastoma patients which may lead to reduced delivery of treatment. This study aims to develop a predictive model that is able to forecast the cytopenia induced by temozolomide (TMZ) during concomitant chemoradiotherapy. Methods Medical records of 128 patients with newly diagnosed glioblastoma were evaluated to extract the baseline complete blood test before and during the six weeks of chemoradiotherapy to create a dataset for the development of ML models. Using the constructed dataset, different ML algorithms were trained and tested. Results Our proposed algorithm achieved accuracies of 85.6%, 88.7%, and 89.3% in predicting thrombocytopenia, lymphopenia, and neutropenia, respectively. Conclusions The algorithm designed and developed in this study, called PrACTiC, showed promising results in the accurate prediction of thrombocytopenia, neutropenia, and lymphopenia induced by TMZ in glioblastoma patients. PrACTiC can provide valuable insight for physicians and help them to make the necessary treatment modifications and prevent the toxicities.
Collapse
|
133
|
Alabi RO, Almangush A, Elmusrati M, Mäkitie AA. Deep Machine Learning for Oral Cancer: From Precise Diagnosis to Precision Medicine. FRONTIERS IN ORAL HEALTH 2022; 2:794248. [PMID: 35088057 PMCID: PMC8786902 DOI: 10.3389/froh.2021.794248] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most prevalent cancers worldwide and its incidence is on the rise in many populations. The high incidence rate, late diagnosis, and improper treatment planning still form a significant concern. Diagnosis at an early-stage is important for better prognosis, treatment, and survival. Despite the recent improvement in the understanding of the molecular mechanisms, late diagnosis and approach toward precision medicine for OSCC patients remain a challenge. To enhance precision medicine, deep machine learning technique has been touted to enhance early detection, and consequently to reduce cancer-specific mortality and morbidity. This technique has been reported to have made a significant progress in data extraction and analysis of vital information in medical imaging in recent years. Therefore, it has the potential to assist in the early-stage detection of oral squamous cell carcinoma. Furthermore, automated image analysis can assist pathologists and clinicians to make an informed decision regarding cancer patients. This article discusses the technical knowledge and algorithms of deep learning for OSCC. It examines the application of deep learning technology in cancer detection, image classification, segmentation and synthesis, and treatment planning. Finally, we discuss how this technique can assist in precision medicine and the future perspective of deep learning technology in oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Rasheed Omobolaji Alabi
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Industrial Digitalization, School of Technology and Innovations, University of Vaasa, Vaasa, Finland
| | - Alhadi Almangush
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pathology, University of Helsinki, Helsinki, Finland
- Institute of Biomedicine, Pathology, University of Turku, Turku, Finland
| | - Mohammed Elmusrati
- Department of Industrial Digitalization, School of Technology and Innovations, University of Vaasa, Vaasa, Finland
| | - Antti A. Mäkitie
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Otorhinolaryngology – Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
134
|
Delaune A, Valmary-Degano S, Loménie N, Zryouil K, Benyahia N, Trassard O, Eraville V, Bergeron C, Devouassoux-Shisheboran M, Glaser C, Bataillon G, Bacry E, Combes S, Prevot S, Bertheau P. Le premier data challenge organisé par la Société Française de Pathologie : une compétition internationale en 2020, un outil de recherche en intelligence artificielle pour l’avenir ? Ann Pathol 2022; 42:119-128. [DOI: 10.1016/j.annpat.2021.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/08/2021] [Accepted: 10/09/2021] [Indexed: 10/19/2022]
|
135
|
Wang Y, Tan H, Yu T, Ma X, Chen X, Jing F, Zou L, Shi H. The identification of gene signatures in patients with extranodal NK/T-cell lymphoma from a pair of twins. BMC Cancer 2021; 21:1303. [PMID: 34872521 PMCID: PMC8650233 DOI: 10.1186/s12885-021-09023-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/17/2021] [Indexed: 02/08/2023] Open
Abstract
Background There is no unified treatment standard for patients with extranodal NK/T-cell lymphoma (ENKTL). Cancer neoantigens are the result of somatic mutations and cancer-specific. Increased number of somatic mutations are associated with anti-cancer effects. Screening out ENKTL-specific neoantigens on the surface of cancer cells relies on the understanding of ENKTL mutation patterns. Hence, it is imperative to identify ENKTL-specific genes for ENKTL diagnosis, the discovery of tumor-specific neoantigens and the development of novel therapeutic strategies. We investigated the gene signatures of ENKTL patients. Methods We collected the peripheral blood of a pair of twins for sequencing to identify unique variant genes. One of the twins is diagnosed with ENKTL. Seventy samples were analyzed by Robust Multi-array Analysis (RMA). Two methods (elastic net and Support Vector Machine-Recursive Feature Elimination) were used to select unique genes. Next, we performed functional enrichment analysis and pathway enrichment analysis. Then, we conducted single-sample gene set enrichment analysis of immune infiltration and validated the expression of the screened markers with limma packages. Results We screened out 126 unique variant genes. Among them, 11 unique genes were selected by the combination of elastic net and Support Vector Machine-Recursive Feature Elimination. Subsequently, GO and KEGG analysis indicated the biological function of identified unique genes. GSEA indicated five immunity-related pathways with high signature scores. In patients with ENKTL and the group with high signature scores, a proportion of functional immune cells are all of great infiltration. We finally found that CDC27, ZNF141, FCGR2C and NES were four significantly differential genes in ENKTL patients. ZNF141, FCGR2C and NES were upregulated in patients with ENKTL, while CDC27 was significantly downregulated. Conclusion We identified four ENKTL markers (ZNF141, FCGR2C, NES and CDC27) in patients with extranodal NK/T-cell lymphoma.
Collapse
Affiliation(s)
- Yang Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Huaicheng Tan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Ting Yu
- Department of Pathology and Laboratory of Pathology, State Key Laboratory of Biotherapy, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,Department of Radiotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxuan Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Fangqi Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Liqun Zou
- Department of Head and Neck Cancer, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China. .,Department of Radiotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
136
|
Ramesh S, Chokkara S, Shen T, Major A, Volchenboum SL, Mayampurath A, Applebaum MA. Applications of Artificial Intelligence in Pediatric Oncology: A Systematic Review. JCO Clin Cancer Inform 2021; 5:1208-1219. [PMID: 34910588 PMCID: PMC8812636 DOI: 10.1200/cci.21.00102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/05/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE There is a need for an improved understanding of clinical and biologic risk factors in pediatric cancer to improve patient outcomes. Machine learning (ML) represents the application of computational inference from advanced statistical methods that can be applied to increasing amount of data available for study in pediatric oncology. The goal of this systematic review was to systematically characterize the state of ML in pediatric oncology and highlight advances and opportunities in the field. METHODS We conducted a systematic review of the Embase, Scopus, and MEDLINE databases for applications of ML in pediatric oncology. Query results from all three databases were aggregated and duplicate studies were removed. RESULTS A total of 42 unique articles that examined the applications of ML in pediatric oncology met inclusion criteria for review. We identified 20 studies of CNS tumors, 13 of solid tumors, and nine of leukemia. ML tasks included classification, prediction of treatment response, and dose optimization with a variety of methods being used including neural network, k-nearest neighbor, random forest, naive Bayes, and support vector machines. Strengths of the identified studies included matching or outperforming physician comparators via automated analysis and predicting therapeutic response. Common limitations included significant heterogeneity in reporting standards, clinical applicability, small sample sizes, and missing external validation cohorts. CONCLUSION We identified areas where ML can enhance clinical care in ways that may not otherwise be achievable. Although ML promises enormous potential in improving diagnostics, decision making, and monitoring for children with cancer, the field remains in early stages and future work will be aided by standards and guidelines to ensure rigorous methodologic design and maximizing clinical utility.
Collapse
Affiliation(s)
- Siddhi Ramesh
- Pritzker School of Medicine, University of Chicago, Chicago, IL
| | - Sukarn Chokkara
- Pritzker School of Medicine, University of Chicago, Chicago, IL
| | - Timothy Shen
- Pritzker School of Medicine, University of Chicago, Chicago, IL
| | - Ajay Major
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | - Samuel L. Volchenboum
- Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | - Anoop Mayampurath
- Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | - Mark A. Applebaum
- Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL
| |
Collapse
|
137
|
Huang X, Huang K, Johnson T, Radovich M, Zhang J, Ma J, Wang Y. ParsVNN: parsimony visible neural networks for uncovering cancer-specific and drug-sensitive genes and pathways. NAR Genom Bioinform 2021; 3:lqab097. [PMID: 34729476 PMCID: PMC8557386 DOI: 10.1093/nargab/lqab097] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/07/2021] [Accepted: 10/08/2021] [Indexed: 11/23/2022] Open
Abstract
Prediction of cancer-specific drug responses as well as identification of the corresponding drug-sensitive genes and pathways remains a major biological and clinical challenge. Deep learning models hold immense promise for better drug response predictions, but most of them cannot provide biological and clinical interpretability. Visible neural network (VNN) models have emerged to solve the problem by giving neurons biological meanings and directly casting biological networks into the models. However, the biological networks used in VNNs are often redundant and contain components that are irrelevant to the downstream predictions. Therefore, the VNNs using these redundant biological networks are overparameterized, which significantly limits VNNs' predictive and explanatory power. To overcome the problem, we treat the edges and nodes in biological networks used in VNNs as features and develop a sparse learning framework ParsVNN to learn parsimony VNNs with only edges and nodes that contribute the most to the prediction task. We applied ParsVNN to build cancer-specific VNN models to predict drug response for five different cancer types. We demonstrated that the parsimony VNNs built by ParsVNN are superior to other state-of-the-art methods in terms of prediction performance and identification of cancer driver genes. Furthermore, we found that the pathways selected by ParsVNN have great potential to predict clinical outcomes as well as recommend synergistic drug combinations.
Collapse
Affiliation(s)
- Xiaoqing Huang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kun Huang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Travis Johnson
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Milan Radovich
- Division of General Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
| | - Jianzhu Ma
- Institute for Artificial Intelligence, Peking University, China
| | - Yijie Wang
- Department of Computer Science, Indiana University, Bloomington, IN 47408, USA
| |
Collapse
|
138
|
Mertens S, Krois J, Cantu AG, Arsiwala LT, Schwendicke F. Artificial intelligence for caries detection: Randomized trial. J Dent 2021; 115:103849. [PMID: 34656656 DOI: 10.1016/j.jdent.2021.103849] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES We aimed to assess the impact of an artificial intelligence (AI)-based diagnostic-support software for proximal caries detection on bitewing radiographs. METHODS A cluster-randomized cross-over controlled trial was conducted. A commercially available software employing a fully convolutional neural network for caries detection (dentalXrai Pro, dentalXrai Ltd.) was randomly employed by 22 dentists, supporting their caries detection on 20 bitewings randomly chosen from a pool of 140 bitewings, with 10 bitewings randomly being supported by AI and 10 not. The reference test had been established by 4 + 1 independent experts in a pixelwise fashion. Caries was subgrouped as enamel, early dentin and advanced dentin caries, and accuracy and treatment decisions for each caries lesion assessed. RESULTS Dentists with AI showed a significantly higher mean (95% CI) area under the Receiver-Operating-Characteristics curve (0.89; 0.87-0.90) than those without AI (0.85; 0.83-0.86; p<0.05), mainly as their sensitivity was significantly higher (0.81; 0.74-0.87 compared with 0.72; 0.64-0.79; p<0.05) while the specificity was not significantly affected (p>0.05). This increase in sensitivity was found for enamel, but not early or advanced dentin lesions. Higher sensitivity came with an increase in non-invasive, but also invasive treatment decisions (p<0.05). CONCLUSION AI can increase dentists' diagnostic accuracy but may also increase invasive treatment decisions. CLINICAL SIGNIFICANCE AI can increase dentists' diagnostic accuracy, mainly via increasing their sensitivity for detecting enamel lesions, but may also increase invasive therapy decisions. Differences in the effects of AI for different dentists should be explored, and dentists should be guided as to which therapy to choose when detecting caries lesions using AI support.
Collapse
Affiliation(s)
- Sarah Mertens
- Department of Oral Diagnostics, Digital Health and Health Services Research, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Operative and Preventive Dentistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Joachim Krois
- Department of Oral Diagnostics, Digital Health and Health Services Research, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Anselmo Garcia Cantu
- Department of Oral Diagnostics, Digital Health and Health Services Research, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Lubaina T Arsiwala
- Department of Oral Diagnostics, Digital Health and Health Services Research, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Falk Schwendicke
- Department of Oral Diagnostics, Digital Health and Health Services Research, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany.
| |
Collapse
|
139
|
Chen P, Chen Xu R, Chen N, Zhang L, Zhang L, Zhu J, Pan B, Wang B, Guo W. Detection of Metastatic Tumor Cells in the Bone Marrow Aspirate Smears by Artificial Intelligence (AI)-Based Morphogo System. Front Oncol 2021; 11:742395. [PMID: 34646779 PMCID: PMC8503678 DOI: 10.3389/fonc.2021.742395] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/06/2021] [Indexed: 11/17/2022] Open
Abstract
Introduction Metastatic carcinomas of bone marrow (MCBM) are characterized as tumors of non-hematopoietic origin spreading to the bone marrow through blood or lymphatic circulation. The diagnosis is critical for tumor staging, treatment selection and prognostic risk stratification. However, the identification of metastatic carcinoma cells on bone marrow aspiration smears is technically challenging by conventional microscopic screening. Objective The aim of this study is to develop an automatic recognition system using deep learning algorithms applied to bone marrow cells image analysis. The system takes advantage of an artificial intelligence (AI)-based method in recognizing metastatic atypical cancer clusters and promoting rapid diagnosis. Methods We retrospectively reviewed metastatic non-hematopoietic malignancies in bone marrow aspirate smears collected from 60 cases of patients admitted to Zhongshan Hospital. High resolution digital bone marrow aspirate smear images were generated and automatically analyzed by Morphogo AI based system. Morphogo system was trained and validated using 20748 cell cluster images from randomly selected 50 MCBM patients. 5469 pre-classified cell cluster images from the remaining 10 MCBM patients were used to test the recognition performance between Morphogo and experienced pathologists. Results Morphogo exhibited a sensitivity of 56.6%, a specificity of 91.3%, and an accuracy of 82.2% in the recognition of metastatic cancer cells. Morphogo’s classification result was in general agreement with the conventional standard in the diagnosis of metastatic cancer clusters, with a Kappa value of 0.513. The test results between Morphogo and pathologists H1, H2 and H3 agreement demonstrated a reliability coefficient of 0.827. The area under the curve (AUC) for Morphogo to diagnose the cancer cell clusters was 0.865. Conclusion In patients with clinical history of cancer, the Morphogo system was validated as a useful screening tool in the identification of metastatic cancer cells in the bone marrow aspirate smears. It has potential clinical application in the diagnostic assessment of metastatic cancers for staging and in screening MCBM during morphology examination when the symptoms of the primary site are indolent.
Collapse
Affiliation(s)
- Pu Chen
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Run Chen Xu
- Department of Medical Development, Hangzhou ZhiWei Information Technology Co. Ltd., Hangzhou, China
| | - Nan Chen
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianfeng Zhu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Baishen Pan
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China.,Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China.,Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China.,Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
140
|
Cyll K, Kleppe A, Kalsnes J, Vlatkovic L, Pradhan M, Kildal W, Tobin KAR, Reine TM, Wæhre H, Brennhovd B, Askautrud HA, Skaaheim Haug E, Hveem TS, Danielsen HE. PTEN and DNA Ploidy Status by Machine Learning in Prostate Cancer. Cancers (Basel) 2021; 13:cancers13174291. [PMID: 34503100 PMCID: PMC8428363 DOI: 10.3390/cancers13174291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 12/05/2022] Open
Abstract
Simple Summary Molecular tissue-based prognostic biomarkers are anticipated to complement the current risk stratification systems in prostate cancer, but their manual assessment is subjective and time-consuming. Objective assessment of such biomarkers by machine learning-based methods could advance their adoption in a clinical workflow. PTEN and DNA ploidy status are well-studied biomarkers, which can provide clinically relevant information in prostate cancer at a low cost. Using a cohort of 253 patients who received radical prostatectomy, we developed a novel, fully-automated PTEN scoring in immunohistochemically-stained tissue slides, which could be used to assess PTEN status in a reliable and reproducible manner. In an independent validation cohort of 259 patients, automatically assessed PTEN status was significantly associated with time to biochemical recurrence after radical prostatectomy, and the combination of PTEN and DNA ploidy status further improved risk stratification. These results demonstrate the utility of machine learning in biomarker assessment. Abstract Machine learning (ML) is expected to improve biomarker assessment. Using convolution neural networks, we developed a fully-automated method for assessing PTEN protein status in immunohistochemically-stained slides using a radical prostatectomy (RP) cohort (n = 253). It was validated according to a predefined protocol in an independent RP cohort (n = 259), alone and by measuring its prognostic value in combination with DNA ploidy status determined by ML-based image cytometry. In the primary analysis, automatically assessed dichotomized PTEN status was associated with time to biochemical recurrence (TTBCR) (hazard ratio (HR) = 3.32, 95% CI 2.05 to 5.38). Patients with both non-diploid tumors and PTEN-low had an HR of 4.63 (95% CI 2.50 to 8.57), while patients with one of these characteristics had an HR of 1.94 (95% CI 1.15 to 3.30), compared to patients with diploid tumors and PTEN-high, in univariable analysis of TTBCR in the validation cohort. Automatic PTEN scoring was strongly predictive of the PTEN status assessed by human experts (area under the curve 0.987 (95% CI 0.968 to 0.994)). This suggests that PTEN status can be accurately assessed using ML, and that the combined marker of automatically assessed PTEN and DNA ploidy status may provide an objective supplement to the existing risk stratification factors in prostate cancer.
Collapse
Affiliation(s)
- Karolina Cyll
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
| | - Andreas Kleppe
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
- Department of Informatics, University of Oslo, NO-0316 Oslo, Norway
| | - Joakim Kalsnes
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
| | - Ljiljana Vlatkovic
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
| | - Manohar Pradhan
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
| | - Wanja Kildal
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
| | - Kari Anne R. Tobin
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
| | - Trine M. Reine
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
| | - Håkon Wæhre
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
| | - Bjørn Brennhovd
- Department of Urology, Oslo University Hospital, NO-0424 Oslo, Norway;
| | - Hanne A. Askautrud
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
| | - Erik Skaaheim Haug
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
- Department of Urology, Vestfold Hospital Trust, NO-3103 Tønsberg, Norway
| | - Tarjei S. Hveem
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
| | - Håvard E. Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424 Oslo, Norway; (K.C.); (A.K.); (J.K.); (L.V.); (M.P.); (W.K.); (K.A.R.T.); (T.M.R.); (H.W.); (H.A.A.); (E.S.H.); (T.S.H.)
- Department of Informatics, University of Oslo, NO-0316 Oslo, Norway
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford OX3 9DU, UK
- Correspondence: ; Tel.: +47-22-78-23-20
| |
Collapse
|
141
|
Abstract
Artificial intelligence (AI) has seen an explosion in interest within nuclear medicine. This interest is driven by the rapid progress and eye-catching achievements of machine learning algorithms. The growing foothold of AI in molecular imaging is exposing nuclear medicine personnel to new technology and terminology. Clinicians and researchers can be easily overwhelmed by numerous architectures and algorithms that have been published. This article dissects the backbone of most AI algorithms: the convolutional neural network. The algorithm training workflow and the key ingredients and operations of a convolutional neural network are described in detail. Finally, the ubiquitous U-Net is explained step-by-step.
Collapse
Affiliation(s)
- Tyler J Bradshaw
- Department of Radiology, University of Wisconsin, 3252 Clinical Science Center, 600 Highland Avenue, Madison, WI 53792, USA.
| | - Alan B McMillan
- Department of Radiology, University of Wisconsin, 3252 Clinical Science Center, 600 Highland Avenue, Madison, WI 53792, USA. https://twitter.com/alan_b_mcmillan
| |
Collapse
|
142
|
Mosallaie S, Rad M, Schiffauerova A, Ebadi A. Discovering the evolution of artificial intelligence in cancer research using dynamic topic modeling. COLLNET JOURNAL OF SCIENTOMETRICS AND INFORMATION MANAGEMENT 2021. [DOI: 10.1080/09737766.2021.1958659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Shahab Mosallaie
- Concordia Institute for Information Systems Engineering (CIISE), Concordia University, H3G 1M8, Montreal, QC, Canada
| | - Mahdi Rad
- Concordia Institute for Information Systems Engineering (CIISE), Concordia University, H3G 1M8, Montreal, QC, Canada
| | - Andrea Schiffauerova
- Concordia Institute for Information Systems Engineering (CIISE), Concordia University, H3G 1M8, Montreal, QC, Canada
| | - Ashkan Ebadi
- Digital Technologies Research Centre, National Research Council Canada, H3T 2B2, Montreal, QC, Canada
- Concordia Institute for Information Systems Engineering, Concordia University, H3G 1M8, Montreal, QC, Canada
| |
Collapse
|
143
|
Reichert G, Bellamine A, Fontaine M, Naipeanu B, Altar A, Mejean E, Javaud N, Siauve N. How Can a Deep Learning Algorithm Improve Fracture Detection on X-rays in the Emergency Room? J Imaging 2021; 7:105. [PMID: 39080893 PMCID: PMC8321374 DOI: 10.3390/jimaging7070105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 11/17/2022] Open
Abstract
The growing need for emergency imaging has greatly increased the number of conventional X-rays, particularly for traumatic injury. Deep learning (DL) algorithms could improve fracture screening by radiologists and emergency room (ER) physicians. We used an algorithm developed for the detection of appendicular skeleton fractures and evaluated its performance for detecting traumatic fractures on conventional X-rays in the ER, without the need for training on local data. This algorithm was tested on all patients (N = 125) consulting at the Louis Mourier ER in May 2019 for limb trauma. Patients were selected by two emergency physicians from the clinical database used in the ER. Their X-rays were exported and analyzed by a radiologist. The prediction made by the algorithm and the annotation made by the radiologist were compared. For the 125 patients included, 25 patients with a fracture were identified by the clinicians, 24 of whom were identified by the algorithm (sensitivity of 96%). The algorithm incorrectly predicted a fracture in 14 of the 100 patients without fractures (specificity of 86%). The negative predictive value was 98.85%. This study shows that DL algorithms are potentially valuable diagnostic tools for detecting fractures in the ER and could be used in the training of junior radiologists.
Collapse
Affiliation(s)
- Guillaume Reichert
- Radiology Department, Louis Mourier Hospital, Assistance Publique-Hôpitaux de Paris (APHP), University of Paris, 92700 Colombes, France; (A.B.); (M.F.); (B.N.)
| | - Ali Bellamine
- Radiology Department, Louis Mourier Hospital, Assistance Publique-Hôpitaux de Paris (APHP), University of Paris, 92700 Colombes, France; (A.B.); (M.F.); (B.N.)
| | - Matthieu Fontaine
- Radiology Department, Louis Mourier Hospital, Assistance Publique-Hôpitaux de Paris (APHP), University of Paris, 92700 Colombes, France; (A.B.); (M.F.); (B.N.)
| | - Beatrice Naipeanu
- Radiology Department, Louis Mourier Hospital, Assistance Publique-Hôpitaux de Paris (APHP), University of Paris, 92700 Colombes, France; (A.B.); (M.F.); (B.N.)
| | - Adrien Altar
- Emergency Department, Louis Mourier Hospital, AP-HP, 92700 Colombes, France; (A.A.); (N.J.)
| | - Elodie Mejean
- Emergency Department, Foch Hospital, 92150 Suresnes, France;
| | - Nicolas Javaud
- Emergency Department, Louis Mourier Hospital, AP-HP, 92700 Colombes, France; (A.A.); (N.J.)
| | - Nathalie Siauve
- Radiology Department, Louis Mourier Hospital, Assistance Publique-Hôpitaux de Paris (APHP), University of Paris, 92700 Colombes, France; (A.B.); (M.F.); (B.N.)
- INSERM, U970, Paris Cardiovascular Research Center—PARCC, 75015 Paris, France
| |
Collapse
|
144
|
Sun X, Feinberg MW. Vascular Endothelial Senescence: Pathobiological Insights, Emerging Long Noncoding RNA Targets, Challenges and Therapeutic Opportunities. Front Physiol 2021; 12:693067. [PMID: 34220553 PMCID: PMC8242592 DOI: 10.3389/fphys.2021.693067] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/07/2021] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a stable form of cell cycle arrest in response to various stressors. While it serves as an endogenous pro-resolving mechanism, detrimental effects ensue when it is dysregulated. In this review, we introduce recent advances for cellular senescence and inflammaging, the underlying mechanisms for the reduction of nicotinamide adenine dinucleotide in tissues during aging, new knowledge learned from p16 reporter mice, and the development of machine learning algorithms in cellular senescence. We focus on pathobiological insights underlying cellular senescence of the vascular endothelium, a critical interface between blood and all tissues. Common causes and hallmarks of endothelial senescence are highlighted as well as recent advances in endothelial senescence. The regulation of cellular senescence involves multiple mechanistic layers involving chromatin, DNA, RNA, and protein levels. New targets are discussed including the roles of long noncoding RNAs in regulating endothelial cellular senescence. Emerging small molecules are highlighted that have anti-aging or anti-senescence effects in age-related diseases and impact homeostatic control of the vascular endothelium. Lastly, challenges and future directions are discussed including heterogeneity of endothelial cells and endothelial senescence, senescent markers and detection of senescent endothelial cells, evolutionary differences for immune surveillance in mice and humans, and long noncoding RNAs as therapeutic targets in attenuating cellular senescence. Accumulating studies indicate that cellular senescence is reversible. A better understanding of endothelial cellular senescence through lifestyle and pharmacological interventions holds promise to foster a new frontier in the management of cardiovascular disease risk.
Collapse
Affiliation(s)
- Xinghui Sun
- Department of Biochemistry, University of Nebraska–Lincoln, Lincoln, NE, United States
- Nebraska Center for the Prevention of Obesity Diseases Through Dietary Molecules, University of Nebraska–Lincoln, Lincoln, NE, United States
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska–Lincoln, Lincoln, NE, United States
| | - Mark W. Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
145
|
van der Laak J, Litjens G, Ciompi F. Deep learning in histopathology: the path to the clinic. Nat Med 2021; 27:775-784. [PMID: 33990804 DOI: 10.1038/s41591-021-01343-4] [Citation(s) in RCA: 311] [Impact Index Per Article: 103.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/31/2021] [Indexed: 02/08/2023]
Abstract
Machine learning techniques have great potential to improve medical diagnostics, offering ways to improve accuracy, reproducibility and speed, and to ease workloads for clinicians. In the field of histopathology, deep learning algorithms have been developed that perform similarly to trained pathologists for tasks such as tumor detection and grading. However, despite these promising results, very few algorithms have reached clinical implementation, challenging the balance between hope and hype for these new techniques. This Review provides an overview of the current state of the field, as well as describing the challenges that still need to be addressed before artificial intelligence in histopathology can achieve clinical value.
Collapse
Affiliation(s)
- Jeroen van der Laak
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands. .,Center for Medical Image Science and Visualization, Linköping University, Linköping, Sweden.
| | - Geert Litjens
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Francesco Ciompi
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|