151
|
El Naqa I, Karolak A, Luo Y, Folio L, Tarhini AA, Rollison D, Parodi K. Translation of AI into oncology clinical practice. Oncogene 2023; 42:3089-3097. [PMID: 37684407 DOI: 10.1038/s41388-023-02826-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023]
Abstract
Artificial intelligence (AI) is a transformative technology that is capturing popular imagination and can revolutionize biomedicine. AI and machine learning (ML) algorithms have the potential to break through existing barriers in oncology research and practice such as automating workflow processes, personalizing care, and reducing healthcare disparities. Emerging applications of AI/ML in the literature include screening and early detection of cancer, disease diagnosis, response prediction, prognosis, and accelerated drug discovery. Despite this excitement, only few AI/ML models have been properly validated and fewer have become regulated products for routine clinical use. In this review, we highlight the main challenges impeding AI/ML clinical translation. We present different clinical use cases from the domains of radiology, radiation oncology, immunotherapy, and drug discovery in oncology. We dissect the unique challenges and opportunities associated with each of these cases. Finally, we summarize the general requirements for successful AI/ML implementation in the clinic, highlighting specific examples and points of emphasis including the importance of multidisciplinary collaboration of stakeholders, role of domain experts in AI augmentation, transparency of AI/ML models, and the establishment of a comprehensive quality assurance program to mitigate risks of training bias and data drifts, all culminating toward safer and more beneficial AI/ML applications in oncology labs and clinics.
Collapse
Affiliation(s)
- Issam El Naqa
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, 33612, USA.
| | - Aleksandra Karolak
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Yi Luo
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Les Folio
- Diagnostic Imaging & Interventional Radiology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Ahmad A Tarhini
- Cutaneous Oncology and Immunology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Dana Rollison
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Katia Parodi
- Department of Medical Physics, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
152
|
Paliwal A, Faust K, Alshoumer A, Diamandis P. Standardizing analysis of intra-tumoral heterogeneity with computational pathology. Genes Chromosomes Cancer 2023; 62:526-539. [PMID: 37067005 DOI: 10.1002/gcc.23146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/18/2023] Open
Abstract
Many malignant cancers like glioblastoma are highly adaptive diseases that dynamically change their regional biology to survive and thrive under diverse microenvironmental and therapeutic pressures. While the concept of intra-tumoral heterogeneity has become a major paradigm in cancer research and care, systematic approaches to assess and document bio-variation in cancer are still in their infancy. Here we discuss existing approaches and challenges to documenting intra-tumoral heterogeneity and emerging computational approaches that leverage artificial intelligence to begin to overcome these limitations. We propose how these emerging techniques can be coupled with a diversity of molecular tools to address intra-tumoral heterogeneity more systematically in research and in practice, especially across larger specimens and longitudinal analyses. Systematic documentation and characterization of heterogeneity across entire tumor specimens and their longitudinal evolution has the potential to improve our understanding and treatment of cancer.
Collapse
Affiliation(s)
- Ameesha Paliwal
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Kevin Faust
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Azhar Alshoumer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine Program, Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - Phedias Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Laboratory Medicine Program, Department of Pathology, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
153
|
Isaksson LJ, Summers P, Mastroleo F, Marvaso G, Corrao G, Vincini MG, Zaffaroni M, Ceci F, Petralia G, Orecchia R, Jereczek-Fossa BA. Automatic Segmentation with Deep Learning in Radiotherapy. Cancers (Basel) 2023; 15:4389. [PMID: 37686665 PMCID: PMC10486603 DOI: 10.3390/cancers15174389] [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: 07/25/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
This review provides a formal overview of current automatic segmentation studies that use deep learning in radiotherapy. It covers 807 published papers and includes multiple cancer sites, image types (CT/MRI/PET), and segmentation methods. We collect key statistics about the papers to uncover commonalities, trends, and methods, and identify areas where more research might be needed. Moreover, we analyzed the corpus by posing explicit questions aimed at providing high-quality and actionable insights, including: "What should researchers think about when starting a segmentation study?", "How can research practices in medical image segmentation be improved?", "What is missing from the current corpus?", and more. This allowed us to provide practical guidelines on how to conduct a good segmentation study in today's competitive environment that will be useful for future research within the field, regardless of the specific radiotherapeutic subfield. To aid in our analysis, we used the large language model ChatGPT to condense information.
Collapse
Affiliation(s)
- Lars Johannes Isaksson
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (L.J.I.); (F.M.); (G.C.); (M.G.V.); (M.Z.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (F.C.); (G.P.)
| | - Paul Summers
- Division of Radiology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Federico Mastroleo
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (L.J.I.); (F.M.); (G.C.); (M.G.V.); (M.Z.); (B.A.J.-F.)
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 20188 Novara, Italy
| | - Giulia Marvaso
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (L.J.I.); (F.M.); (G.C.); (M.G.V.); (M.Z.); (B.A.J.-F.)
| | - Giulia Corrao
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (L.J.I.); (F.M.); (G.C.); (M.G.V.); (M.Z.); (B.A.J.-F.)
| | - Maria Giulia Vincini
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (L.J.I.); (F.M.); (G.C.); (M.G.V.); (M.Z.); (B.A.J.-F.)
| | - Mattia Zaffaroni
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (L.J.I.); (F.M.); (G.C.); (M.G.V.); (M.Z.); (B.A.J.-F.)
| | - Francesco Ceci
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (F.C.); (G.P.)
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Petralia
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (F.C.); (G.P.)
- Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Roberto Orecchia
- Scientific Directorate, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (L.J.I.); (F.M.); (G.C.); (M.G.V.); (M.Z.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (F.C.); (G.P.)
| |
Collapse
|
154
|
Cooper M, Ji Z, Krishnan RG. Machine learning in computational histopathology: Challenges and opportunities. Genes Chromosomes Cancer 2023; 62:540-556. [PMID: 37314068 DOI: 10.1002/gcc.23177] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/15/2023] Open
Abstract
Digital histopathological images, high-resolution images of stained tissue samples, are a vital tool for clinicians to diagnose and stage cancers. The visual analysis of patient state based on these images are an important part of oncology workflow. Although pathology workflows have historically been conducted in laboratories under a microscope, the increasing digitization of histopathological images has led to their analysis on computers in the clinic. The last decade has seen the emergence of machine learning, and deep learning in particular, a powerful set of tools for the analysis of histopathological images. Machine learning models trained on large datasets of digitized histopathology slides have resulted in automated models for prediction and stratification of patient risk. In this review, we provide context for the rise of such models in computational histopathology, highlight the clinical tasks they have found success in automating, discuss the various machine learning techniques that have been applied to this domain, and underscore open problems and opportunities.
Collapse
Affiliation(s)
- Michael Cooper
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- University Health Network, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Zongliang Ji
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Rahul G Krishnan
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
155
|
Shao J, Feng J, Li J, Liang S, Li W, Wang C. Novel tools for early diagnosis and precision treatment based on artificial intelligence. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:148-160. [PMID: 39171128 PMCID: PMC11332840 DOI: 10.1016/j.pccm.2023.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Indexed: 08/23/2024]
Abstract
Lung cancer has the highest mortality rate among all cancers in the world. Hence, early diagnosis and personalized treatment plans are crucial to improving its 5-year survival rate. Chest computed tomography (CT) serves as an essential tool for lung cancer screening, and pathology images are the gold standard for lung cancer diagnosis. However, medical image evaluation relies on manual labor and suffers from missed diagnosis or misdiagnosis, and physician heterogeneity. The rapid development of artificial intelligence (AI) has brought a whole novel opportunity for medical task processing, demonstrating the potential for clinical application in lung cancer diagnosis and treatment. AI technologies, including machine learning and deep learning, have been deployed extensively for lung nodule detection, benign and malignant classification, and subtype identification based on CT images. Furthermore, AI plays a role in the non-invasive prediction of genetic mutations and molecular status to provide the optimal treatment regimen, and applies to the assessment of therapeutic efficacy and prognosis of lung cancer patients, enabling precision medicine to become a reality. Meanwhile, histology-based AI models assist pathologists in typing, molecular characterization, and prognosis prediction to enhance the efficiency of diagnosis and treatment. However, the leap to extensive clinical application still faces various challenges, such as data sharing, standardized label acquisition, clinical application regulation, and multimodal integration. Nevertheless, AI holds promising potential in the field of lung cancer to improve cancer care.
Collapse
Affiliation(s)
- Jun Shao
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiaming Feng
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingwei Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shufan Liang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
156
|
Brummer O, Pölönen P, Mustjoki S, Brück O. Computational textural mapping harmonises sampling variation and reveals multidimensional histopathological fingerprints. Br J Cancer 2023; 129:683-695. [PMID: 37391505 PMCID: PMC10421901 DOI: 10.1038/s41416-023-02329-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/18/2023] [Accepted: 06/14/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Technical factors can bias H&E digital slides potentially compromising computational histopathology studies. Here, we hypothesised that sample quality and sampling variation can introduce even greater and undocumented technical fallacy. METHODS Using The Cancer Genome Atlas (TCGA) clear-cell renal cell carcinoma (ccRCC) as a model disease, we annotated ~78,000 image tiles and trained deep learning models to detect histological textures and lymphocyte infiltration at the tumour core and its surrounding margin and correlated these with clinical, immunological, genomic, and transcriptomic profiles. RESULTS The models reached 95% validation accuracy for classifying textures and 95% for lymphocyte infiltration enabling reliable profiling of ccRCC samples. We validated the lymphocyte-per-texture distributions in the Helsinki dataset (n = 64). Texture analysis indicated constitutive sampling bias by TCGA clinical centres and technically suboptimal samples. We demonstrate how computational texture mapping (CTM) can abrogate these issues by normalising textural variance. CTM-harmonised histopathological architecture resonated with both expected associations and novel molecular fingerprints. For instance, tumour fibrosis associated with histological grade, epithelial-to-mesenchymal transition, low mutation burden and metastasis. CONCLUSIONS This study highlights texture-based standardisation to resolve technical bias in computational histopathology and understand the molecular basis of tissue architecture. All code, data and models are released as a community resource.
Collapse
Affiliation(s)
- Otso Brummer
- Hematoscope Lab, Helsinki University Hospital, Comprehensive Cancer Center and Center of Diagnostics, Helsinki, Finland
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Petri Pölönen
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Oscar Brück
- Hematoscope Lab, Helsinki University Hospital, Comprehensive Cancer Center and Center of Diagnostics, Helsinki, Finland.
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
157
|
Wang Q, Jiang S, Wu Y, Zhang Y, Huang M, Qiu Y, Luo X. Prognostic and clinicopathological role of RACK1 for cancer patients: a systematic review and meta-analysis. PeerJ 2023; 11:e15873. [PMID: 37601269 PMCID: PMC10434108 DOI: 10.7717/peerj.15873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Background The receptor for activated C kinase 1 (RACK1) expression is associated with clinicopathological characteristics and the prognosis of various cancers; however, the conclusions are controversial. As a result, this study aimed to explore the clinicopathological and prognostic values of RACK1 expression in patients with cancer. Methodology PubMed, Embase, Web of Science, Cochrane Library, and Scopus were comprehensively explored from their inception to April 20, 2023, for selecting studies on the clinicopathological and prognostic role of RACK1 in patients with cancer that met the criteria for inclusion in this review. Pooled hazard ratios (HRs) and 95% confidence intervals (CIs) were used to assess the prognosis-predictive value of RACK1 expression, while pooled odds ratios (ORs) and 95% CIs were used to evaluate the correlation between RACK1 expression and the clinicopathological characteristics of patients with cancer. The quality of the included studies was evaluated using the Newcastle-Ottawa Scale. Results Twenty-two studies (13 on prognosis and 20 on clinicopathological characteristics) were included in this systematic review and meta-analysis. The findings indicated that high RACK1 expression was significantly associated with poor overall survival (HR = 1.62; 95% CI, 1.13-2.33; P = 0.009; I2 = 89%) and reversely correlated with disease-free survival/recurrence-free survival (HR = 1.87; 95% CI, 1.22-2.88; P = 0.004; I2 = 0%). Furthermore, increased RACK1 expression was significantly associated with lymphatic invasion/N+ stage (OR = 1.74; 95% CI, 1.04-2.90; P = 0.04; I2 = 79%) of tumors. Conclusions RACK1 may be a global predictive marker of poor prognosis in patients with cancer and unfavorable clinicopathological characteristics. However, further clinical studies are required to validate these findings.
Collapse
Affiliation(s)
- Qiuhao Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sixin Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuqi Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - You Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mei Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan Qiu
- Laboratory of Pathology, Clinical Research Center for Breast, Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaobo Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
158
|
Rösler W, Altenbuchinger M, Baeßler B, Beissbarth T, Beutel G, Bock R, von Bubnoff N, Eckardt JN, Foersch S, Loeffler CML, Middeke JM, Mueller ML, Oellerich T, Risse B, Scherag A, Schliemann C, Scholz M, Spang R, Thielscher C, Tsoukakis I, Kather JN. An overview and a roadmap for artificial intelligence in hematology and oncology. J Cancer Res Clin Oncol 2023; 149:7997-8006. [PMID: 36920563 PMCID: PMC10374829 DOI: 10.1007/s00432-023-04667-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 02/23/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Artificial intelligence (AI) is influencing our society on many levels and has broad implications for the future practice of hematology and oncology. However, for many medical professionals and researchers, it often remains unclear what AI can and cannot do, and what are promising areas for a sensible application of AI in hematology and oncology. Finally, the limits and perils of using AI in oncology are not obvious to many healthcare professionals. METHODS In this article, we provide an expert-based consensus statement by the joint Working Group on "Artificial Intelligence in Hematology and Oncology" by the German Society of Hematology and Oncology (DGHO), the German Association for Medical Informatics, Biometry and Epidemiology (GMDS), and the Special Interest Group Digital Health of the German Informatics Society (GI). We provide a conceptual framework for AI in hematology and oncology. RESULTS First, we propose a technological definition, which we deliberately set in a narrow frame to mainly include the technical developments of the last ten years. Second, we present a taxonomy of clinically relevant AI systems, structured according to the type of clinical data they are used to analyze. Third, we show an overview of potential applications, including clinical, research, and educational environments with a focus on hematology and oncology. CONCLUSION Thus, this article provides a point of reference for hematologists and oncologists, and at the same time sets forth a framework for the further development and clinical deployment of AI in hematology and oncology in the future.
Collapse
Affiliation(s)
- Wiebke Rösler
- Department for Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Altenbuchinger
- Department of Medical Bioinformatics, University Medical Center Göttingen, Göttingen, Germany
| | - Bettina Baeßler
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Würzburg, Germany
| | - Tim Beissbarth
- Department of Medical Bioinformatics, University Medical Center Göttingen, Göttingen, Germany
| | - Gernot Beutel
- Department for Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Robert Bock
- IMMS Institute for Microelectronics and Mechatronics Systems GmbH (NPO), Ilmenau, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, Medical Center, University of Schleswig Holstein, Campus Lübeck, Lübeck, Germany
| | - Jan-Niklas Eckardt
- Department of Medicine 1, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Else Kroener Fresenius Center for Digital Health (EFFZ), Technical University Dresden, Dresden, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Chiara M L Loeffler
- Department of Medicine 1, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Else Kroener Fresenius Center for Digital Health (EFFZ), Technical University Dresden, Dresden, Germany
| | - Jan Moritz Middeke
- Department of Medicine 1, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Else Kroener Fresenius Center for Digital Health (EFFZ), Technical University Dresden, Dresden, Germany
| | | | - Thomas Oellerich
- Medizinische Klinik 2-Haematology/Oncology, University Hospital, Frankfurt am Main, Germany
| | - Benjamin Risse
- Computer Vision and Machine Learning Systems Group, Institute for Geoinformatics, University of Münster, Münster, Germany
| | - André Scherag
- Institute of Medical Statistics, Computer and Data Sciences, Jena University Hospital - Friedrich Schiller University, Jena, Germany
| | | | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Rainer Spang
- Department of Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
| | | | - Ioannis Tsoukakis
- Department of Hematology and Oncology, Sana Klinikum Offenbach, Offenbach, Germany
| | - Jakob Nikolas Kather
- Department of Medicine 1, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
- Else Kroener Fresenius Center for Digital Health (EFFZ), Technical University Dresden, Dresden, Germany.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
159
|
Asif A, Rajpoot K, Graham S, Snead D, Minhas F, Rajpoot N. Unleashing the potential of AI for pathology: challenges and recommendations. J Pathol 2023; 260:564-577. [PMID: 37550878 PMCID: PMC10952719 DOI: 10.1002/path.6168] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 08/09/2023]
Abstract
Computational pathology is currently witnessing a surge in the development of AI techniques, offering promise for achieving breakthroughs and significantly impacting the practices of pathology and oncology. These AI methods bring with them the potential to revolutionize diagnostic pipelines as well as treatment planning and overall patient care. Numerous peer-reviewed studies reporting remarkable performance across diverse tasks serve as a testimony to the potential of AI in the field. However, widespread adoption of these methods in clinical and pre-clinical settings still remains a challenge. In this review article, we present a detailed analysis of the major obstacles encountered during the development of effective models and their deployment in practice. We aim to provide readers with an overview of the latest developments, assist them with insights into identifying some specific challenges that may require resolution, and suggest recommendations and potential future research directions. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Amina Asif
- Tissue Image Analytics Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Kashif Rajpoot
- School of Computer ScienceUniversity of BirminghamBirminghamUK
| | - Simon Graham
- Histofy Ltd, Birmingham Business ParkBirminghamUK
| | - David Snead
- Histofy Ltd, Birmingham Business ParkBirminghamUK
- Department of PathologyUniversity Hospitals Coventry & Warwickshire NHS TrustCoventryUK
| | - Fayyaz Minhas
- Tissue Image Analytics Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
- Cancer Research CentreUniversity of WarwickCoventryUK
| | - Nasir Rajpoot
- Tissue Image Analytics Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
- Histofy Ltd, Birmingham Business ParkBirminghamUK
- Cancer Research CentreUniversity of WarwickCoventryUK
- The Alan Turing InstituteLondonUK
| |
Collapse
|
160
|
Fu X, Sahai E, Wilkins A. Application of digital pathology-based advanced analytics of tumour microenvironment organisation to predict prognosis and therapeutic response. J Pathol 2023; 260:578-591. [PMID: 37551703 PMCID: PMC10952145 DOI: 10.1002/path.6153] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 08/09/2023]
Abstract
In recent years, the application of advanced analytics, especially artificial intelligence (AI), to digital H&E images, and other histological image types, has begun to radically change how histological images are used in the clinic. Alongside the recognition that the tumour microenvironment (TME) has a profound impact on tumour phenotype, the technical development of highly multiplexed immunofluorescence platforms has enhanced the biological complexity that can be captured in the TME with high precision. AI has an increasingly powerful role in the recognition and quantitation of image features and the association of such features with clinically important outcomes, as occurs in distinct stages in conventional machine learning. Deep-learning algorithms are able to elucidate TME patterns inherent in the input data with minimum levels of human intelligence and, hence, have the potential to achieve clinically relevant predictions and discovery of important TME features. Furthermore, the diverse repertoire of deep-learning algorithms able to interrogate TME patterns extends beyond convolutional neural networks to include attention-based models, graph neural networks, and multimodal models. To date, AI models have largely been evaluated retrospectively, outside the well-established rigour of prospective clinical trials, in part because traditional clinical trial methodology may not always be suitable for the assessment of AI technology. However, to enable digital pathology-based advanced analytics to meaningfully impact clinical care, specific measures of 'added benefit' to the current standard of care and validation in a prospective setting are important. This will need to be accompanied by adequate measures of explainability and interpretability. Despite such challenges, the combination of expanding datasets, increased computational power, and the possibility of integration of pre-clinical experimental insights into model development means there is exciting potential for the future progress of these AI applications. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Xiao Fu
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
- Biomolecular Modelling LaboratoryThe Francis Crick InstituteLondonUK
| | - Erik Sahai
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
| | - Anna Wilkins
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
- Division of Radiotherapy and ImagingInstitute of Cancer ResearchLondonUK
- Royal Marsden Hospitals NHS TrustLondonUK
| |
Collapse
|
161
|
Carrillo-Perez F, Pizurica M, Zheng Y, Nandi TN, Madduri R, Shen J, Gevaert O. RNA-to-image multi-cancer synthesis using cascaded diffusion models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523899. [PMID: 36711711 PMCID: PMC9882105 DOI: 10.1101/2023.01.13.523899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Data scarcity presents a significant obstacle in the field of biomedicine, where acquiring diverse and sufficient datasets can be costly and challenging. Synthetic data generation offers a potential solution to this problem by expanding dataset sizes, thereby enabling the training of more robust and generalizable machine learning models. Although previous studies have explored synthetic data generation for cancer diagnosis, they have predominantly focused on single modality settings, such as whole-slide image tiles or RNA-Seq data. To bridge this gap, we propose a novel approach, RNA-Cascaded-Diffusion-Model or RNA-CDM, for performing RNA-to-image synthesis in a multi-cancer context, drawing inspiration from successful text-to-image synthesis models used in natural images. In our approach, we employ a variational auto-encoder to reduce the dimensionality of a patient's gene expression profile, effectively distinguishing between different types of cancer. Subsequently, we employ a cascaded diffusion model to synthesize realistic whole-slide image tiles using the latent representation derived from the patient's RNA-Seq data. Our results demonstrate that the generated tiles accurately preserve the distribution of cell types observed in real-world data, with state-of-the-art cell identification models successfully detecting important cell types in the synthetic samples. Furthermore, we illustrate that the synthetic tiles maintain the cell fraction observed in bulk RNA-Seq data and that modifications in gene expression affect the composition of cell types in the synthetic tiles. Next, we utilize the synthetic data generated by RNA-CDM to pretrain machine learning models and observe improved performance compared to training from scratch. Our study emphasizes the potential usefulness of synthetic data in developing machine learning models in sarce-data settings, while also highlighting the possibility of imputing missing data modalities by leveraging the available information. In conclusion, our proposed RNA-CDM approach for synthetic data generation in biomedicine, particularly in the context of cancer diagnosis, offers a novel and promising solution to address data scarcity. By generating synthetic data that aligns with real-world distributions and leveraging it to pretrain machine learning models, we contribute to the development of robust clinical decision support systems and potential advancements in precision medicine.
Collapse
|
162
|
Hickman RA, Scholz SW. Precision diagnosis and staging of TDP-43 proteinopathies: harnessing the power of artificial intelligence. Brain 2023; 146:2666-2668. [PMID: 37224516 PMCID: PMC10316762 DOI: 10.1093/brain/awad175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 05/26/2023] Open
Abstract
This scientific commentary refers to ‘Data-driven neuropathological staging and subtyping of TDP-43 proteinopathies’ by Young et al. (https://doi.org/10.1093/brain/awad145).
Collapse
Affiliation(s)
- Richard A Hickman
- Department of Defense/Uniformed Services University Brain Tissue Repository, Uniformed Services University, Bethesda, MD 20817, USA
- Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD 20817, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
- Human Oncology and Pathogenesis Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA
| |
Collapse
|
163
|
Choi S, Kim S. Artificial Intelligence in the Pathology of Gastric Cancer. J Gastric Cancer 2023; 23:410-427. [PMID: 37553129 PMCID: PMC10412971 DOI: 10.5230/jgc.2023.23.e25] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/09/2023] [Accepted: 07/14/2023] [Indexed: 08/10/2023] Open
Abstract
Recent advances in artificial intelligence (AI) have provided novel tools for rapid and precise pathologic diagnosis. The introduction of digital pathology has enabled the acquisition of scanned slide images that are essential for the application of AI. The application of AI for improved pathologic diagnosis includes the error-free detection of potentially negligible lesions, such as a minute focus of metastatic tumor cells in lymph nodes, the accurate diagnosis of potentially controversial histologic findings, such as very well-differentiated carcinomas mimicking normal epithelial tissues, and the pathological subtyping of the cancers. Additionally, the utilization of AI algorithms enables the precise decision of the score of immunohistochemical markers for targeted therapies, such as human epidermal growth factor receptor 2 and programmed death-ligand 1. Studies have revealed that AI assistance can reduce the discordance of interpretation between pathologists and more accurately predict clinical outcomes. Several approaches have been employed to develop novel biomarkers from histologic images using AI. Moreover, AI-assisted analysis of the cancer microenvironment showed that the distribution of tumor-infiltrating lymphocytes was related to the response to the immune checkpoint inhibitor therapy, emphasizing its value as a biomarker. As numerous studies have demonstrated the significance of AI-assisted interpretation and biomarker development, the AI-based approach will advance diagnostic pathology.
Collapse
Affiliation(s)
- Sangjoon Choi
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.
| |
Collapse
|
164
|
Chen W, Sá RC, Bai Y, Napel S, Gevaert O, Lauderdale DS, Giger ML. Machine learning with multimodal data for COVID-19. Heliyon 2023; 9:e17934. [PMID: 37483733 PMCID: PMC10362086 DOI: 10.1016/j.heliyon.2023.e17934] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
In response to the unprecedented global healthcare crisis of the COVID-19 pandemic, the scientific community has joined forces to tackle the challenges and prepare for future pandemics. Multiple modalities of data have been investigated to understand the nature of COVID-19. In this paper, MIDRC investigators present an overview of the state-of-the-art development of multimodal machine learning for COVID-19 and model assessment considerations for future studies. We begin with a discussion of the lessons learned from radiogenomic studies for cancer diagnosis. We then summarize the multi-modality COVID-19 data investigated in the literature including symptoms and other clinical data, laboratory tests, imaging, pathology, physiology, and other omics data. Publicly available multimodal COVID-19 data provided by MIDRC and other sources are summarized. After an overview of machine learning developments using multimodal data for COVID-19, we present our perspectives on the future development of multimodal machine learning models for COVID-19.
Collapse
Affiliation(s)
- Weijie Chen
- Medical Imaging and Data Resource Center (MIDRC), USA
- Division of Imaging, Diagnostics, and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, USA
| | - Rui C. Sá
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Medicine, University of California, San Diego, USA
| | - Yuntong Bai
- Medical Imaging and Data Resource Center (MIDRC), USA
- Division of Imaging, Diagnostics, and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, USA
| | - Sandy Napel
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Radiology, Stanford University, USA
| | - Olivier Gevaert
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Medicine and Department of Biomedical Data Science, Stanford University, USA
| | - Diane S. Lauderdale
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Public Health Sciences, University of Chicago, USA
| | - Maryellen L. Giger
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Radiology, University of Chicago, USA
| |
Collapse
|
165
|
Zheng Q, Yang R, Xu H, Fan J, Jiao P, Ni X, Yuan J, Wang L, Chen Z, Liu X. A Weakly Supervised Deep Learning Model and Human-Machine Fusion for Accurate Grading of Renal Cell Carcinoma from Histopathology Slides. Cancers (Basel) 2023; 15:3198. [PMID: 37370808 DOI: 10.3390/cancers15123198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/23/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
(1) Background: The Fuhrman grading (FG) system is widely used in the management of clear cell renal cell carcinoma (ccRCC). However, it is affected by observer variability and irreproducibility in clinical practice. We aimed to use a deep learning multi-class model called SSL-CLAM to assist in diagnosing the FG status of ccRCC patients using digitized whole slide images (WSIs). (2) Methods: We recruited 504 eligible ccRCC patients from The Cancer Genome Atlas (TCGA) cohort and obtained 708 hematoxylin and eosin-stained WSIs for the development and internal validation of the SSL-CLAM model. Additionally, we obtained 445 WSIs from 188 ccRCC eligible patients in the Clinical Proteomic Tumor Analysis Consortium (CPTAC) cohort as an independent external validation set. A human-machine fusion approach was used to validate the added value of the SSL-CLAM model for pathologists. (3) Results: The SSL-CLAM model successfully diagnosed the five FG statuses (Grade-0, 1, 2, 3, and 4) of ccRCC, and achieved AUCs of 0.917 and 0.887 on the internal and external validation sets, respectively, outperforming a junior pathologist. For the normal/tumor classification (Grade-0, Grade-1/2/3/4) task, the SSL-CLAM model yielded AUCs close to 1 on both the internal and external validation sets. The SSL-CLAM model achieved a better performance for the two-tiered FG (Grade-0, Grade-1/2, and Grade-3/4) task, with AUCs of 0.936 and 0.915 on the internal and external validation sets, respectively. The human-machine diagnostic performance was superior to that of the SSL-CLAM model, showing promising prospects. In addition, the high-attention regions of the SSL-CLAM model showed that with an increasing FG status, the cell nuclei in the tumor region become larger, with irregular contours and increased cellular pleomorphism. (4) Conclusions: Our findings support the feasibility of using deep learning and human-machine fusion methods for FG classification on WSIs from ccRCC patients, which may assist pathologists in making diagnostic decisions.
Collapse
Affiliation(s)
- Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Huazhen Xu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Junjie Fan
- University of Chinese Academy of Sciences, Beijing 100049, China
- Trusted Computing and Information Assurance Laboratory, Institute of Software, Chinese Academy of Sciences, Beijing 100190, China
| | - Panpan Jiao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xinmiao Ni
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
166
|
Cheng J, Huang K, Xu J. Editorial: Computational pathology for precision diagnosis, treatment, and prognosis of cancer. Front Med (Lausanne) 2023; 10:1209666. [PMID: 37347090 PMCID: PMC10280156 DOI: 10.3389/fmed.2023.1209666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Affiliation(s)
- Jun Cheng
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Medical Ultrasound Image Computing (MUSIC) Laboratory, Shenzhen University, Shenzhen, China
- Marshall Laboratory of Biomedical Engineering, Shenzhen University, Shenzhen, China
| | - Kun Huang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, United States
- Regenstrief Institute, Indianapolis, IN, United States
| | - Jun Xu
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China
| |
Collapse
|
167
|
Jiang C, Hou X, Kondepudi A, Chowdury A, Freudiger CW, Orringer DA, Lee H, Hollon TC. Hierarchical discriminative learning improves visual representations of biomedical microscopy. PROCEEDINGS. IEEE COMPUTER SOCIETY CONFERENCE ON COMPUTER VISION AND PATTERN RECOGNITION 2023; 2023:19798-19808. [PMID: 37654477 PMCID: PMC10468966 DOI: 10.1109/cvpr52729.2023.01896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Learning high-quality, self-supervised, visual representations is essential to advance the role of computer vision in biomedical microscopy and clinical medicine. Previous work has focused on self-supervised representation learning (SSL) methods developed for instance discrimination and applied them directly to image patches, or fields-of-view, sampled from gigapixel whole-slide images (WSIs) used for cancer diagnosis. However, this strategy is limited because it (1) assumes patches from the same patient are independent, (2) neglects the patient-slide-patch hierarchy of clinical biomedical microscopy, and (3) requires strong data augmentations that can degrade downstream performance. Importantly, sampled patches from WSIs of a patient's tumor are a diverse set of image examples that capture the same underlying cancer diagnosis. This motivated HiDisc, a data-driven method that leverages the inherent patient-slide-patch hierarchy of clinical biomedical microscopy to define a hierarchical discriminative learning task that implicitly learns features of the underlying diagnosis. HiDisc uses a self-supervised contrastive learning framework in which positive patch pairs are defined based on a common ancestry in the data hierarchy, and a unified patch, slide, and patient discriminative learning objective is used for visual SSL. We benchmark HiDisc visual representations on two vision tasks using two biomedical microscopy datasets, and demonstrate that (1) HiDisc pretraining outperforms current state-of-the-art self-supervised pretraining methods for cancer diagnosis and genetic mutation prediction, and (2) HiDisc learns high-quality visual representations using natural patch diversity without strong data augmentations.
Collapse
|
168
|
Chen RJ, Wang JJ, Williamson DFK, Chen TY, Lipkova J, Lu MY, Sahai S, Mahmood F. Algorithmic fairness in artificial intelligence for medicine and healthcare. Nat Biomed Eng 2023; 7:719-742. [PMID: 37380750 PMCID: PMC10632090 DOI: 10.1038/s41551-023-01056-8] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 04/13/2023] [Indexed: 06/30/2023]
Abstract
In healthcare, the development and deployment of insufficiently fair systems of artificial intelligence (AI) can undermine the delivery of equitable care. Assessments of AI models stratified across subpopulations have revealed inequalities in how patients are diagnosed, treated and billed. In this Perspective, we outline fairness in machine learning through the lens of healthcare, and discuss how algorithmic biases (in data acquisition, genetic variation and intra-observer labelling variability, in particular) arise in clinical workflows and the resulting healthcare disparities. We also review emerging technology for mitigating biases via disentanglement, federated learning and model explainability, and their role in the development of AI-based software as a medical device.
Collapse
Affiliation(s)
- Richard J Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Judy J Wang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Boston University School of Medicine, Boston, MA, USA
| | - Drew F K Williamson
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tiffany Y Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jana Lipkova
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ming Y Lu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sharifa Sahai
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Cancer Program, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA.
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
169
|
Nikfar M, Mi H, Gong C, Kimko H, Popel AS. Quantifying Intratumoral Heterogeneity and Immunoarchitecture Generated In-Silico by a Spatial Quantitative Systems Pharmacology Model. Cancers (Basel) 2023; 15:2750. [PMID: 37345087 DOI: 10.3390/cancers15102750] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
Spatial heterogeneity is a hallmark of cancer. Tumor heterogeneity can vary with time and location. The tumor microenvironment (TME) encompasses various cell types and their interactions that impart response to therapies. Therefore, a quantitative evaluation of tumor heterogeneity is crucial for the development of effective treatments. Different approaches, such as multiregional sequencing, spatial transcriptomics, analysis of autopsy samples, and longitudinal analysis of biopsy samples, can be used to analyze the intratumoral heterogeneity (ITH) and temporal evolution and to reveal the mechanisms of therapeutic response. However, because of the limitations of these data and the uncertainty associated with the time points of sample collection, having a complete understanding of intratumoral heterogeneity role is challenging. Here, we used a hybrid model that integrates a whole-patient compartmental quantitative-systems-pharmacology (QSP) model with a spatial agent-based model (ABM) describing the TME; we applied four spatial metrics to quantify model-simulated intratumoral heterogeneity and classified the TME immunoarchitecture for representative cases of effective and ineffective anti-PD-1 therapy. The four metrics, adopted from computational digital pathology, included mixing score, average neighbor frequency, Shannon's entropy and area under the curve (AUC) of the G-cross function. A fifth non-spatial metric was used to supplement the analysis, which was the ratio of the number of cancer cells to immune cells. These metrics were utilized to classify the TME as "cold", "compartmentalized" and "mixed", which were related to treatment efficacy. The trends in these metrics for effective and ineffective treatments are in qualitative agreement with the clinical literature, indicating that compartmentalized immunoarchitecture is likely to result in more efficacious treatment outcomes.
Collapse
Affiliation(s)
- Mehdi Nikfar
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Haoyang Mi
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chang Gong
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, Waltham, MA 02451, USA
| | - Holly Kimko
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University, Baltimore, MD 21231, USA
| |
Collapse
|
170
|
Yan R, Shen Y, Zhang X, Xu P, Wang J, Li J, Ren F, Ye D, Zhou SK. Histopathological bladder cancer gene mutation prediction with hierarchical deep multiple-instance learning. Med Image Anal 2023; 87:102824. [PMID: 37126973 DOI: 10.1016/j.media.2023.102824] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/13/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
Gene mutation detection is usually carried out by molecular biological methods, which is expensive and has a long-time cycle. In contrast, pathological images are ubiquitous. If clinically significant gene mutations can be predicted only through pathological images, it will greatly promote the widespread use of gene mutation detection in clinical practice. However, current gene mutation prediction methods based on pathological images are ineffective because of the inability to identify mutated regions in gigapixel Whole Slide Image (WSI). To address this challenge, hereby we propose a carefully designed framework for WSI-based gene mutation prediction, which consists of three parts. (i) The first part of cancerous area segmentation, based on supervised learning, quickly filters out a large number of non-mutated regions; (ii) the second part of cancerous patch clustering, based on the representations derived from contrastive learning, ensures the comprehensiveness of patch selection; and (iii) the third part of mutation classification, based on the proposed hierarchical deep multi-instance learning method (HDMIL), ensures that sufficient patches are considered and inaccurate selections are ignored. In addition, benefiting from a two-stage attention mechanism in HDMIL, the patches that are highly correlated with gene mutations can be identified. This interpretability can help a pathologist to analyze the correlation between gene mutation and histopathological morphology. Experimental results demonstrate that the proposed gene mutation prediction framework significantly outperforms the state-of-the-art methods. In the TCGA bladder cancer dataset, five clinically relevant gene mutations are well predicted.
Collapse
Affiliation(s)
- Rui Yan
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yijun Shen
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xueyuan Zhang
- Zhijian Life Technology Co., Ltd., Beijing, 100036, China
| | - Peihang Xu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun Wang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jintao Li
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Fei Ren
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China; SKLP, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - S Kevin Zhou
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China; School of Biomedical Engineering & Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China.
| |
Collapse
|
171
|
Niehues JM, Quirke P, West NP, Grabsch HI, van Treeck M, Schirris Y, Veldhuizen GP, Hutchins GGA, Richman SD, Foersch S, Brinker TJ, Fukuoka J, Bychkov A, Uegami W, Truhn D, Brenner H, Brobeil A, Hoffmeister M, Kather JN. Generalizable biomarker prediction from cancer pathology slides with self-supervised deep learning: A retrospective multi-centric study. Cell Rep Med 2023; 4:100980. [PMID: 36958327 PMCID: PMC10140458 DOI: 10.1016/j.xcrm.2023.100980] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/28/2022] [Accepted: 02/24/2023] [Indexed: 03/25/2023]
Abstract
Deep learning (DL) can predict microsatellite instability (MSI) from routine histopathology slides of colorectal cancer (CRC). However, it is unclear whether DL can also predict other biomarkers with high performance and whether DL predictions generalize to external patient populations. Here, we acquire CRC tissue samples from two large multi-centric studies. We systematically compare six different state-of-the-art DL architectures to predict biomarkers from pathology slides, including MSI and mutations in BRAF, KRAS, NRAS, and PIK3CA. Using a large external validation cohort to provide a realistic evaluation setting, we show that models using self-supervised, attention-based multiple-instance learning consistently outperform previous approaches while offering explainable visualizations of the indicative regions and morphologies. While the prediction of MSI and BRAF mutations reaches a clinical-grade performance, mutation prediction of PIK3CA, KRAS, and NRAS was clinically insufficient.
Collapse
Affiliation(s)
- Jan Moritz Niehues
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 01307 Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Philip Quirke
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK
| | - Nicholas P West
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK
| | - Heike I Grabsch
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK; Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Center+, 6229 HX Maastricht, the Netherlands
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 01307 Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Yoni Schirris
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 01307 Dresden, Germany; Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; University of Amsterdam, 1012 WP Amsterdam, the Netherlands
| | - Gregory P Veldhuizen
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 01307 Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Gordon G A Hutchins
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK
| | - Susan D Richman
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Junya Fukuoka
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan; Department of Pathology, Kameda Medical Center, Kamogawa 296-8602, Chiba, Japan
| | - Andrey Bychkov
- Department of Pathology, Kameda Medical Center, Kamogawa 296-8602, Chiba, Japan
| | - Wataru Uegami
- Department of Pathology, Kameda Medical Center, Kamogawa 296-8602, Chiba, Japan
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Alexander Brobeil
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany; Tissue Bank, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 01307 Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK; Department of Medicine I, University Hospital Dresden, 01307 Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
172
|
Zhang W, Li E, Wang L, Lehmann BD, Chen XS. Transcriptome Meta-Analysis of Triple-Negative Breast Cancer Response to Neoadjuvant Chemotherapy. Cancers (Basel) 2023; 15:2194. [PMID: 37190123 PMCID: PMC10137141 DOI: 10.3390/cancers15082194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease with varying responses to neoadjuvant chemotherapy (NAC). The identification of biomarkers to predict NAC response and inform personalized treatment strategies is essential. In this study, we conducted large-scale gene expression meta-analyses to identify genes associated with NAC response and survival outcomes. The results showed that immune, cell cycle/mitotic, and RNA splicing-related pathways were significantly associated with favorable clinical outcomes. Furthermore, we integrated and divided the gene association results from NAC response and survival outcomes into four quadrants, which provided more insights into potential NAC response mechanisms and biomarker discovery.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Emma Li
- California Academy of Mathematics and Science, 1000 E Victoria St, Carson, CA 90747, USA
| | - Lily Wang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Brian D. Lehmann
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - X. Steven Chen
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
173
|
Mandair D, Reis-Filho JS, Ashworth A. Biological insights and novel biomarker discovery through deep learning approaches in breast cancer histopathology. NPJ Breast Cancer 2023; 9:21. [PMID: 37024522 PMCID: PMC10079681 DOI: 10.1038/s41523-023-00518-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/27/2023] [Indexed: 04/08/2023] Open
Abstract
Breast cancer remains a highly prevalent disease with considerable inter- and intra-tumoral heterogeneity complicating prognostication and treatment decisions. The utilization and depth of genomic, transcriptomic and proteomic data for cancer has exploded over recent times and the addition of spatial context to this information, by understanding the correlating morphologic and spatial patterns of cells in tissue samples, has created an exciting frontier of research, histo-genomics. At the same time, deep learning (DL), a class of machine learning algorithms employing artificial neural networks, has rapidly progressed in the last decade with a confluence of technical developments - including the advent of modern graphic processing units (GPU), allowing efficient implementation of increasingly complex architectures at scale; advances in the theoretical and practical design of network architectures; and access to larger datasets for training - all leading to sweeping advances in image classification and object detection. In this review, we examine recent developments in the application of DL in breast cancer histology with particular emphasis of those producing biologic insights or novel biomarkers, spanning the extraction of genomic information to the use of stroma to predict cancer recurrence, with the aim of suggesting avenues for further advancing this exciting field.
Collapse
Affiliation(s)
- Divneet Mandair
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94158, USA
| | | | - Alan Ashworth
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94158, USA.
| |
Collapse
|
174
|
Liang J, Zhang W, Yang J, Wu M, Dai Q, Yin H, Xiao Y, Kong L. Deep learning supported discovery of biomarkers for clinical prognosis of liver cancer. NAT MACH INTELL 2023. [DOI: 10.1038/s42256-023-00635-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
175
|
Liu P, Ji L, Ye F, Fu B. GraphLSurv: A scalable survival prediction network with adaptive and sparse structure learning for histopathological whole-slide images. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 231:107433. [PMID: 36841107 DOI: 10.1016/j.cmpb.2023.107433] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/17/2023] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND AND OBJECTIVE Predicting patients' survival from gigapixel Whole-Slide Images (WSIs) has always been a challenging task. To learn effective WSI representations for survival prediction, existing deep learning methods have explored utilizing graphs to describe the complex structure inner WSIs, where graph node is respective to WSI patch. However, these graphs are often densely-connected or static, leading to some redundant or missing patch correlations. Moreover, these methods cannot be directly scaled to the very-large WSI with more than 10,000 patches. To address these, this paper proposes a scalable graph convolution network, GraphLSurv, which can efficiently learn adaptive and sparse structures to better characterize WSIs for survival prediction. METHODS GraphLSurv has three highlights in methodology: (1) it generates adaptive and sparse structures for patches so that latent patch correlations could be captured and adjusted dynamically according to prediction tasks; (2) based on the generated structure and a given graph, GraphLSurv further aggregates local microenvironmental cues into a non-local embedding using the proposed hybrid message passing network; (3) to make this network suitable for very large-scale graphs, it adopts an anchor-based technique to reduce theorical computation complexity. RESULTS The experiments on 2268 WSIs show that GraphLSurv achieves a concordance-index of 0.66132 and 0.68348, with an improvement of 3.79% and 3.41% compared to existing methods, on NLST and TCGA-BRCA, respectively. CONCLUSIONS GraphLSurv could often perform better than previous methods, which suggests that GraphLSurv could provide an important and effective means for WSI survival prediction. Moreover, this work empirically shows that adaptive and sparse structures could be more suitable than static or dense ones for modeling WSIs.
Collapse
Affiliation(s)
- Pei Liu
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Xiyuan Ave, Chengdu 611731, Sichuan, China.
| | - Luping Ji
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Xiyuan Ave, Chengdu 611731, Sichuan, China.
| | - Feng Ye
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Guo Xue Xiang, Chengdu 610041, Sichuan, China.
| | - Bo Fu
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Xiyuan Ave, Chengdu 611731, Sichuan, China.
| |
Collapse
|
176
|
Steyaert S, Pizurica M, Nagaraj D, Khandelwal P, Hernandez-Boussard T, Gentles AJ, Gevaert O. Multimodal data fusion for cancer biomarker discovery with deep learning. NAT MACH INTELL 2023; 5:351-362. [PMID: 37693852 PMCID: PMC10484010 DOI: 10.1038/s42256-023-00633-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 02/17/2023] [Indexed: 09/12/2023]
Abstract
Technological advances now make it possible to study a patient from multiple angles with high-dimensional, high-throughput multi-scale biomedical data. In oncology, massive amounts of data are being generated ranging from molecular, histopathology, radiology to clinical records. The introduction of deep learning has significantly advanced the analysis of biomedical data. However, most approaches focus on single data modalities leading to slow progress in methods to integrate complementary data types. Development of effective multimodal fusion approaches is becoming increasingly important as a single modality might not be consistent and sufficient to capture the heterogeneity of complex diseases to tailor medical care and improve personalised medicine. Many initiatives now focus on integrating these disparate modalities to unravel the biological processes involved in multifactorial diseases such as cancer. However, many obstacles remain, including lack of usable data as well as methods for clinical validation and interpretation. Here, we cover these current challenges and reflect on opportunities through deep learning to tackle data sparsity and scarcity, multimodal interpretability, and standardisation of datasets.
Collapse
Affiliation(s)
- Sandra Steyaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
| | - Marija Pizurica
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
| | | | | | - Tina Hernandez-Boussard
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| | - Andrew J Gentles
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| |
Collapse
|
177
|
Saldanha OL, Loeffler CML, Niehues JM, van Treeck M, Seraphin TP, Hewitt KJ, Cifci D, Veldhuizen GP, Ramesh S, Pearson AT, Kather JN. Self-supervised attention-based deep learning for pan-cancer mutation prediction from histopathology. NPJ Precis Oncol 2023; 7:35. [PMID: 36977919 PMCID: PMC10050159 DOI: 10.1038/s41698-023-00365-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 02/17/2023] [Indexed: 03/30/2023] Open
Abstract
The histopathological phenotype of tumors reflects the underlying genetic makeup. Deep learning can predict genetic alterations from pathology slides, but it is unclear how well these predictions generalize to external datasets. We performed a systematic study on Deep-Learning-based prediction of genetic alterations from histology, using two large datasets of multiple tumor types. We show that an analysis pipeline that integrates self-supervised feature extraction and attention-based multiple instance learning achieves a robust predictability and generalizability.
Collapse
Affiliation(s)
- Oliver Lester Saldanha
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Jan Moritz Niehues
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Tobias P Seraphin
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty at Heinrich-Heine-University Duesseldorf, Düsseldorf, Germany
| | - Katherine Jane Hewitt
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Didem Cifci
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Gregory Patrick Veldhuizen
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Siddhi Ramesh
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Alexander T Pearson
- Biological Sciences Division, University of Chicago, Chicago, IL, USA
- University of Chicago Comprehensive Cancer Center, University of Chicago, Chicago, IL, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.
- Pathology & 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.
- Department of Medicine 1, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
178
|
Predicting gene mutation status via artificial intelligence technologies based on multimodal integration (MMI) to advance precision oncology. Semin Cancer Biol 2023; 91:1-15. [PMID: 36801447 DOI: 10.1016/j.semcancer.2023.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/30/2023] [Accepted: 02/15/2023] [Indexed: 02/21/2023]
Abstract
Personalized treatment strategies for cancer frequently rely on the detection of genetic alterations which are determined by molecular biology assays. Historically, these processes typically required single-gene sequencing, next-generation sequencing, or visual inspection of histopathology slides by experienced pathologists in a clinical context. In the past decade, advances in artificial intelligence (AI) technologies have demonstrated remarkable potential in assisting physicians with accurate diagnosis of oncology image-recognition tasks. Meanwhile, AI techniques make it possible to integrate multimodal data such as radiology, histology, and genomics, providing critical guidance for the stratification of patients in the context of precision therapy. Given that the mutation detection is unaffordable and time-consuming for a considerable number of patients, predicting gene mutations based on routine clinical radiological scans or whole-slide images of tissue with AI-based methods has become a hot issue in actual clinical practice. In this review, we synthesized the general framework of multimodal integration (MMI) for molecular intelligent diagnostics beyond standard techniques. Then we summarized the emerging applications of AI in the prediction of mutational and molecular profiles of common cancers (lung, brain, breast, and other tumor types) pertaining to radiology and histology imaging. Furthermore, we concluded that there truly exist multiple challenges of AI techniques in the way of its real-world application in the medical field, including data curation, feature fusion, model interpretability, and practice regulations. Despite these challenges, we still prospect the clinical implementation of AI as a highly potential decision-support tool to aid oncologists in future cancer treatment management.
Collapse
|
179
|
Toh MR, Wong EYT, Wong SH, Ng AWT, Loo LH, Chow PKH, Ngeow JYY. Global Epidemiology and Genetics of Hepatocellular Carcinoma. Gastroenterology 2023; 164:766-782. [PMID: 36738977 DOI: 10.1053/j.gastro.2023.01.033] [Citation(s) in RCA: 246] [Impact Index Per Article: 123.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading cancers worldwide. Classically, HCC develops in genetically susceptible individuals who are exposed to risk factors, especially in the presence of liver cirrhosis. Significant temporal and geographic variations exist for HCC and its etiologies. Over time, the burden of HCC has shifted from the low-moderate to the high sociodemographic index regions, reflecting the transition from viral to nonviral causes. Geographically, the hepatitis viruses predominate as the causes of HCC in Asia and Africa. Although there are genetic conditions that confer increased risk for HCC, these diagnoses are rarely recognized outside North America and Europe. In this review, we will evaluate the epidemiologic trends and risk factors of HCC, and discuss the genetics of HCC, including monogenic diseases, single-nucleotide polymorphisms, gut microbiome, and somatic mutations.
Collapse
Affiliation(s)
- Ming Ren Toh
- Cancer Genetics Service, National Cancer Centre Singapore, Singapore
| | | | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Alvin Wei Tian Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Lit-Hsin Loo
- Bioinformatics Institute, Agency for Science, Technology, and Research (A∗STAR), Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Pierce Kah-Hoe Chow
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, National Cancer Center Singapore and Singapore General Hospital, Singapore; Duke-NUS Medical School Singapore, Singapore
| | - Joanne Yuen Yie Ngeow
- Cancer Genetics Service, National Cancer Centre Singapore, Singapore; Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Duke-NUS Medical School Singapore, Singapore.
| |
Collapse
|
180
|
Foersch S, Glasner C, Woerl AC, Eckstein M, Wagner DC, Schulz S, Kellers F, Fernandez A, Tserea K, Kloth M, Hartmann A, Heintz A, Weichert W, Roth W, Geppert C, Kather JN, Jesinghaus M. Multistain deep learning for prediction of prognosis and therapy response in colorectal cancer. Nat Med 2023; 29:430-439. [PMID: 36624314 DOI: 10.1038/s41591-022-02134-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/17/2022] [Indexed: 01/11/2023]
Abstract
Although it has long been known that the immune cell composition has a strong prognostic and predictive value in colorectal cancer (CRC), scoring systems such as the immunoscore (IS) or quantification of intraepithelial lymphocytes are only slowly being adopted into clinical routine use and have their limitations. To address this we established and evaluated a multistain deep learning model (MSDLM) utilizing artificial intelligence (AI) to determine the AImmunoscore (AIS) in more than 1,000 patients with CRC. Our model had high prognostic capabilities and outperformed other clinical, molecular and immune cell-based parameters. It could also be used to predict the response to neoadjuvant therapy in patients with rectal cancer. Using an explainable AI approach, we confirmed that the MSDLM's decisions were based on established cellular patterns of anti-tumor immunity. Hence, the AIS could provide clinicians with a valuable decision-making tool based on the tumor immune microenvironment.
Collapse
Affiliation(s)
- Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany.
| | - Christina Glasner
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Ann-Christin Woerl
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
- Institute of Computer Science, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Eckstein
- Institute of Pathology and Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Stefan Schulz
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Franziska Kellers
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
- Department of Pathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Aurélie Fernandez
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | | | - Michael Kloth
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Arndt Hartmann
- Institute of Pathology and Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Achim Heintz
- Department of General Visceral and Vascular Surgery, Marien Hospital Mainz, Mainz, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Carol Geppert
- Institute of Pathology and Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 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
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Moritz Jesinghaus
- Institute of Pathology, Technical University Munich, Munich, Germany
- Institute of Pathology, University Hospital Marburg, Marburg, Germany
| |
Collapse
|
181
|
Howard FM, Kather JN, Pearson AT. Multimodal deep learning: An improvement in prognostication or a reflection of batch effect? Cancer Cell 2023; 41:5-6. [PMID: 36368319 DOI: 10.1016/j.ccell.2022.10.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany.
| | | |
Collapse
|
182
|
Wu M, Zhu C, Yang J, Cheng S, Yang X, Gu S, Xu S, Wu Y, Shen W, Huang S, Wang Y. Exploring prognostic indicators in the pathological images of ovarian cancer based on a deep survival network. Front Genet 2023; 13:1069673. [PMID: 36685892 PMCID: PMC9846244 DOI: 10.3389/fgene.2022.1069673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Background: Tumor pathology can assess patient prognosis based on a morphological deviation of tumor tissue from normal. Digitizing whole slide images (WSIs) of tissue enables the use of deep learning (DL) techniques in pathology, which may shed light on prognostic indicators of cancers, and avoid biases introduced by human experience. Purpose: We aim to explore new prognostic indicators of ovarian cancer (OC) patients using the DL framework on WSIs, and provide a valuable approach for OC risk stratification. Methods: We obtained the TCGA-OV dataset from the NIH Genomic Data Commons Data Portal database. The preprocessing of the dataset was comprised of three stages: 1) The WSIs and corresponding clinical data were paired and filtered based on a unique patient ID; 2) a weakly-supervised CLAM WSI-analysis tool was exploited to segment regions of interest; 3) the pre-trained model ResNet50 on ImageNet was employed to extract feature tensors. We proposed an attention-based network to predict a hazard score for each case. Furthermore, all cases were divided into a high-risk score group and a low-risk one according to the median as the threshold value. The multi-omics data of OC patients were used to assess the potential applications of the risk score. Finally, a nomogram based on risk scores and age features was established. Results: A total of 90 WSIs were processed, extracted, and fed into the attention-based network. The mean value of the resulting C-index was 0.5789 (0.5096-0.6053), and the resulting p-value was 0.00845. Moreover, the risk score showed a better prediction ability in the HRD + subgroup. Conclusion: Our deep learning framework is a promising method for searching WSIs, and providing a valuable clinical means for prognosis.
Collapse
Affiliation(s)
- Meixuan Wu
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China,Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chengguang Zhu
- MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jiani Yang
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shanshan Cheng
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaokang Yang
- MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Sijia Gu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shilin Xu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yongsong Wu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Shen
- MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Yu Wang, ; Shan Huang, ; Wei Shen,
| | - Shan Huang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China,*Correspondence: Yu Wang, ; Shan Huang, ; Wei Shen,
| | - Yu Wang
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China,*Correspondence: Yu Wang, ; Shan Huang, ; Wei Shen,
| |
Collapse
|
183
|
Lheureux S. Multi-omics Uncovering Different Faces of Clear Cell Ovarian Cancer. Clin Cancer Res 2022; 28:4838-4839. [PMID: 36094332 DOI: 10.1158/1078-0432.ccr-22-2365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/27/2022] [Accepted: 09/02/2022] [Indexed: 01/24/2023]
Abstract
The diagnosis of clear cell ovarian cancer relies on expert histopathology review. Further characterization from deep genomic and transcriptomic analyses can identify different subgroups. International collaboration is required to define the clinical impact and therapy opportunities in these specific subclassifications. See related article by Bolton et al., p. 4947.
Collapse
Affiliation(s)
- Stephanie Lheureux
- Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
184
|
Abstract
Meaningful integration of artificial intelligence (AI) will transform the application of "big data" for patient care, diagnosis, and research. In this issue of Cancer Cell, Chen et al. describe a transparent system to integrate histopathology and molecular data to predict outcomes and identify novel biomarkers in cancer.
Collapse
Affiliation(s)
- Alexander J Lazar
- Departments of Pathology, Genomic Medicine, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth G Demicco
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|