1
|
Bianco V, Valentino M, Pirone D, Miccio L, Memmolo P, Brancato V, Coppola L, Smaldone G, D’Aiuto M, Mossetti G, Salvatore M, Ferraro P. Classifying breast cancer and fibroadenoma tissue biopsies from paraffined stain-free slides by fractal biomarkers in Fourier Ptychographic Microscopy. Comput Struct Biotechnol J 2024; 24:225-236. [PMID: 38572166 PMCID: PMC10990711 DOI: 10.1016/j.csbj.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
Breast cancer is one of the most spread and monitored pathologies in high-income countries. After breast biopsy, histological tissue is stored in paraffin, sectioned and mounted. Conventional inspection of tissue slides under benchtop light microscopes involves paraffin removal and staining, typically with H&E. Then, expert pathologists are called to judge the stained slides. However, paraffin removal and staining are operator-dependent, time and resources consuming processes that can generate ambiguities due to non-uniform staining. Here we propose a novel method that can work directly on paraffined stain-free slides. We use Fourier Ptychography as a quantitative phase-contrast microscopy method, which allows accessing a very wide field of view (i.e., mm2) in one single image while guaranteeing high lateral resolution (i.e., 0.5 µm). This imaging method is multi-scale, since it enables looking at the big picture, i.e. the complex tissue structure and connections, with the possibility to zoom-in up to the single-cell level. To handle this informative image content, we introduce elements of fractal geometry as multi-scale analysis method. We show the effectiveness of fractal features in describing and classifying fibroadenoma and breast cancer tissue slides from ten patients with very high accuracy. We reach 94.0 ± 4.2% test accuracy in classifying single images. Above all, we show that combining the decisions of the single images, each patient's slide can be classified with no error. Besides, fractal geometry returns a guide map to help pathologist to judge the different tissue portions based on the likelihood these can be associated to a breast cancer or fibroadenoma biomarker. The proposed automatic method could significantly simplify the steps of tissue analysis and make it independent from the sample preparation, the skills of the lab operator and the pathologist.
Collapse
Affiliation(s)
- Vittorio Bianco
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems “E. Caianiello”, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| | - Marika Valentino
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems “E. Caianiello”, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
- DIETI, Department of Electrical Engineering and Information Technologies, University of Naples “Federico II”, via Claudio 21, 80125 Napoli, Italy
| | - Daniele Pirone
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems “E. Caianiello”, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| | - Lisa Miccio
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems “E. Caianiello”, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| | - Pasquale Memmolo
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems “E. Caianiello”, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| | | | - Luigi Coppola
- IRCCS SYNLAB SDN, Via E. Gianturco 113, Napoli 80143, Italy
| | | | | | - Gennaro Mossetti
- Pathological Anatomy Service, Casa di Cura Maria Rosaria, Via Colle San Bartolomeo 50, 80045 Pompei, Napoli, Italy
| | | | - Pietro Ferraro
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems “E. Caianiello”, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| |
Collapse
|
2
|
Stenman S, Bétrisey S, Vainio P, Huvila J, Lundin M, Linder N, Schmitt A, Perren A, Dettmer MS, Haglund C, Arola J, Lundin J. External validation of a deep learning-based algorithm for detection of tall cells in papillary thyroid carcinoma: A multicenter study. J Pathol Inform 2024; 15:100366. [PMID: 38425542 PMCID: PMC10901856 DOI: 10.1016/j.jpi.2024.100366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/03/2024] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
The tall cell subtype (TC-PTC) is an aggressive subtype of papillary thyroid carcinoma (PTC). The TC-PTC is defined as a PTC comprising at least 30% epithelial cells that are three times as tall as they are wide. In practice, this definition is difficult to adhere to, resulting in high inter-observer variability. In this multicenter study, we validated a previously trained deep learning (DL)-based algorithm for detection of tall cells on 160 externally collected hematoxylin and eosin (HE)-stained PTC whole-slide images. In a test set of 360 manual annotations of regions of interest from 18 separate tissue sections in the external dataset, the DL-based algorithm detected TCs with a sensitivity of 90.6% and a specificity of 88.5%. The DL algorithm detected non-TC areas with a sensitivity of 81.6% and a specificity of 92.9%. In the validation datasets, 20% and 30% TC thresholds correlated with a significantly shorter relapse-free survival. In conclusion, the DL algorithm detected TCs in unseen, external scanned HE tissue slides with high sensitivity and specificity without any retraining.
Collapse
Affiliation(s)
- Sebastian Stenman
- Institute for Molecular Medicine Finland – FIMM, University of Helsinki, Tukholmankatu 8, 00290 Helsinki, Finland
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 3C, 000290 HUS Helsinki, Finland
- Department of Surgery, Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
| | - Sylvain Bétrisey
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstrasse 31, 3008 Bern, Switzerland
| | - Paula Vainio
- Department of Pathology, University of Turku, Turku University Hospital, Kiinamyllykatu 10, 20520 Turku, Finland
| | - Jutta Huvila
- Department of Pathology, University of Turku, Turku University Hospital, Kiinamyllykatu 10, 20520 Turku, Finland
| | - Mikael Lundin
- Institute for Molecular Medicine Finland – FIMM, University of Helsinki, Tukholmankatu 8, 00290 Helsinki, Finland
| | - Nina Linder
- Institute for Molecular Medicine Finland – FIMM, University of Helsinki, Tukholmankatu 8, 00290 Helsinki, Finland
- Institute of Pathology, Klinikum Stuttgart, Kriegsbergstraße 60, 70174 Stuttgart, Germany
| | - Anja Schmitt
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstrasse 31, 3008 Bern, Switzerland
| | - Aurel Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstrasse 31, 3008 Bern, Switzerland
| | - Matthias S. Dettmer
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstrasse 31, 3008 Bern, Switzerland
- The Global Health & Migration Department of Women’s and Children’s Health, Uppsala University, 75185 Uppsala, Sweden
| | - Caj Haglund
- Department of Surgery, Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
- Research Programs Unit, Translational Cancer Medicine, University of Helsinki, Haartmaninkatu 4, 00290 Helsinki, Finland
| | - Johanna Arola
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 3C, 000290 HUS Helsinki, Finland
| | - Johan Lundin
- Institute for Molecular Medicine Finland – FIMM, University of Helsinki, Tukholmankatu 8, 00290 Helsinki, Finland
- Department of Global Public Health, Karolinska Institutet, Norrbackagatan 4, 17176 Stockholm, Sweden
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| |
Collapse
|
3
|
Hosseini MS, Bejnordi BE, Trinh VQH, Chan L, Hasan D, Li X, Yang S, Kim T, Zhang H, Wu T, Chinniah K, Maghsoudlou S, Zhang R, Zhu J, Khaki S, Buin A, Chaji F, Salehi A, Nguyen BN, Samaras D, Plataniotis KN. Computational pathology: A survey review and the way forward. J Pathol Inform 2024; 15:100357. [PMID: 38420608 PMCID: PMC10900832 DOI: 10.1016/j.jpi.2023.100357] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 03/02/2024] Open
Abstract
Computational Pathology (CPath) is an interdisciplinary science that augments developments of computational approaches to analyze and model medical histopathology images. The main objective for CPath is to develop infrastructure and workflows of digital diagnostics as an assistive CAD system for clinical pathology, facilitating transformational changes in the diagnosis and treatment of cancer that are mainly address by CPath tools. With evergrowing developments in deep learning and computer vision algorithms, and the ease of the data flow from digital pathology, currently CPath is witnessing a paradigm shift. Despite the sheer volume of engineering and scientific works being introduced for cancer image analysis, there is still a considerable gap of adopting and integrating these algorithms in clinical practice. This raises a significant question regarding the direction and trends that are undertaken in CPath. In this article we provide a comprehensive review of more than 800 papers to address the challenges faced in problem design all-the-way to the application and implementation viewpoints. We have catalogued each paper into a model-card by examining the key works and challenges faced to layout the current landscape in CPath. We hope this helps the community to locate relevant works and facilitate understanding of the field's future directions. In a nutshell, we oversee the CPath developments in cycle of stages which are required to be cohesively linked together to address the challenges associated with such multidisciplinary science. We overview this cycle from different perspectives of data-centric, model-centric, and application-centric problems. We finally sketch remaining challenges and provide directions for future technical developments and clinical integration of CPath. For updated information on this survey review paper and accessing to the original model cards repository, please refer to GitHub. Updated version of this draft can also be found from arXiv.
Collapse
Affiliation(s)
- Mahdi S. Hosseini
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | | | - Vincent Quoc-Huy Trinh
- Institute for Research in Immunology and Cancer of the University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Lyndon Chan
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Danial Hasan
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Xingwen Li
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Stephen Yang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Taehyo Kim
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Haochen Zhang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Theodore Wu
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Kajanan Chinniah
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Sina Maghsoudlou
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | - Ryan Zhang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Jiadai Zhu
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Samir Khaki
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Andrei Buin
- Huron Digitial Pathology, St. Jacobs, ON N0B 2N0, Canada
| | - Fatemeh Chaji
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | - Ala Salehi
- Department of Electrical and Computer Engineering, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| | - Bich Ngoc Nguyen
- University of Montreal Hospital Center, Montreal, QC H2X 0C2, Canada
| | - Dimitris Samaras
- Department of Computer Science, Stony Brook University, Stony Brook, NY 11794, United States
| | - Konstantinos N. Plataniotis
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| |
Collapse
|
4
|
Gonzalez R, Saha A, Campbell CJ, Nejat P, Lokker C, Norgan AP. Seeing the random forest through the decision trees. Supporting learning health systems from histopathology with machine learning models: Challenges and opportunities. J Pathol Inform 2024; 15:100347. [PMID: 38162950 PMCID: PMC10755052 DOI: 10.1016/j.jpi.2023.100347] [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: 08/21/2023] [Revised: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 01/03/2024] Open
Abstract
This paper discusses some overlooked challenges faced when working with machine learning models for histopathology and presents a novel opportunity to support "Learning Health Systems" with them. Initially, the authors elaborate on these challenges after separating them according to their mitigation strategies: those that need innovative approaches, time, or future technological capabilities and those that require a conceptual reappraisal from a critical perspective. Then, a novel opportunity to support "Learning Health Systems" by integrating hidden information extracted by ML models from digitalized histopathology slides with other healthcare big data is presented.
Collapse
Affiliation(s)
- Ricardo Gonzalez
- DeGroote School of Business, McMaster University, Hamilton, Ontario, Canada
- Division of Computational Pathology and Artificial Intelligence, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Ashirbani Saha
- Department of Oncology, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Escarpment Cancer Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Clinton J.V. Campbell
- William Osler Health System, Brampton, Ontario, Canada
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Peyman Nejat
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Cynthia Lokker
- Health Information Research Unit, Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Andrew P. Norgan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
5
|
Jacob M, Reddy RP, Garcia RI, Reddy AP, Khemka S, Roghani AK, Pattoor V, Sehar U, Reddy PH. Harnessing Artificial Intelligence for the Detection and Management of Colorectal Cancer Treatment. Cancer Prev Res (Phila) 2024; 17:499-515. [PMID: 39077801 PMCID: PMC11534518 DOI: 10.1158/1940-6207.capr-24-0178] [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: 04/08/2024] [Revised: 06/26/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
Currently, eight million people in the United States suffer from cancer and it is a major global health concern. Early detection and interventions are urgently needed for all cancers, including colorectal cancer. Colorectal cancer is the third most common type of cancer worldwide. Based on the diagnostic efforts to general awareness and lifestyle choices, it is understandable why colorectal cancer is so prevalent today. There is a notable lack of awareness concerning the impact of this cancer and its connection to lifestyle elements, as well as people sometimes mistaking symptoms for a different gastrointestinal condition. Artificial intelligence (AI) may assist in the early detection of all cancers, including colorectal cancer. The usage of AI has exponentially grown in healthcare through extensive research, and since clinical implementation, it has succeeded in improving patient lifestyles, modernizing diagnostic processes, and innovating current treatment strategies. Numerous challenges arise for patients with colorectal cancer and oncologists alike during treatment. For initial screening phases, conventional methods often result in misdiagnosis. Moreover, after detection, determining the course of which colorectal cancer can sometimes contribute to treatment delays. This article touches on recent advancements in AI and its clinical application while shedding light on why this disease is so common today.
Collapse
Affiliation(s)
- Michael Jacob
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas
| | - Ruhananhad P Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Lubbock High School, Lubbock, Texas
| | - Ricardo I Garcia
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Aananya P Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Lubbock High School, Lubbock, Texas
| | - Sachi Khemka
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Aryan Kia Roghani
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Frenship High School, Lubbock, Texas
| | - Vasanthkumar Pattoor
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- University of South Florida, Tampa, Florida
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Nutritional Sciences Department, College of Human Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, Texas
- Department of Speech, Language and Hearing Services, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, Texas
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| |
Collapse
|
6
|
Duo Y, Han L, Yang Y, Wang Z, Wang L, Chen J, Xiang Z, Yoon J, Luo G, Tang BZ. Aggregation-Induced Emission Luminogen: Role in Biopsy for Precision Medicine. Chem Rev 2024; 124:11242-11347. [PMID: 39380213 PMCID: PMC11503637 DOI: 10.1021/acs.chemrev.4c00244] [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: 04/03/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Biopsy, including tissue and liquid biopsy, offers comprehensive and real-time physiological and pathological information for disease detection, diagnosis, and monitoring. Fluorescent probes are frequently selected to obtain adequate information on pathological processes in a rapid and minimally invasive manner based on their advantages for biopsy. However, conventional fluorescent probes have been found to show aggregation-caused quenching (ACQ) properties, impeding greater progresses in this area. Since the discovery of aggregation-induced emission luminogen (AIEgen) have promoted rapid advancements in molecular bionanomaterials owing to their unique properties, including high quantum yield (QY) and signal-to-noise ratio (SNR), etc. This review seeks to present the latest advances in AIEgen-based biofluorescent probes for biopsy in real or artificial samples, and also the key properties of these AIE probes. This review is divided into: (i) tissue biopsy based on smart AIEgens, (ii) blood sample biopsy based on smart AIEgens, (iii) urine sample biopsy based on smart AIEgens, (iv) saliva sample biopsy based on smart AIEgens, (v) biopsy of other liquid samples based on smart AIEgens, and (vi) perspectives and conclusion. This review could provide additional guidance to motivate interest and bolster more innovative ideas for further exploring the applications of various smart AIEgens in precision medicine.
Collapse
Affiliation(s)
- Yanhong Duo
- Department
of Radiation Oncology, Shenzhen People’s Hospital, The Second
Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong China
- Wyss
Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02138, United States
| | - Lei Han
- College of
Chemistry and Pharmaceutical Sciences, Qingdao
Agricultural University, 700 Changcheng Road, Qingdao 266109, Shandong China
| | - Yaoqiang Yang
- Department
of Radiation Oncology, Shenzhen People’s Hospital, The Second
Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong China
| | - Zhifeng Wang
- Department
of Urology, Henan Provincial People’s Hospital, Zhengzhou University
People’s Hospital, Henan University
People’s Hospital, Zhengzhou, 450003, China
| | - Lirong Wang
- State
Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Jingyi Chen
- Wyss
Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02138, United States
| | - Zhongyuan Xiang
- Department
of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Juyoung Yoon
- Department
of Chemistry and Nanoscience, Ewha Womans
University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Guanghong Luo
- Department
of Radiation Oncology, Shenzhen People’s Hospital, The Second
Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong China
| | - Ben Zhong Tang
- School
of Science and Engineering, Shenzhen Institute of Aggregate Science
and Technology, The Chinese University of
Hong Kong, Shenzhen 518172, Guangdong China
| |
Collapse
|
7
|
Syrykh C, van den Brand M, Kather JN, Laurent C. Role of artificial intelligence in haematolymphoid diagnostics. Histopathology 2024. [PMID: 39435690 DOI: 10.1111/his.15327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
The advent of digital pathology and the deployment of high-throughput molecular techniques are generating an unprecedented mass of data. Thanks to advances in computational sciences, artificial intelligence (AI) approaches represent a promising avenue for extracting relevant information from complex data structures. From diagnostic assistance to powerful research tools, the potential fields of application of machine learning techniques in pathology are vast and constitute the subject of considerable research work. The aim of this article is to provide an overview of the potential applications of AI in the field of haematopathology and to define the role that these emerging technologies could play in our laboratories in the short to medium term.
Collapse
Affiliation(s)
- Charlotte Syrykh
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse CHU Toulouse, Toulouse, France
| | - Michiel van den Brand
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
- Pathology-DNA, Arnhem, The Netherlands
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Camille Laurent
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse CHU Toulouse, Toulouse, France
- INSERM UMR1037, CNRS UMR5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| |
Collapse
|
8
|
Zhang WY, Chang YJ, Shi RH. Artificial intelligence enhances the management of esophageal squamous cell carcinoma in the precision oncology era. World J Gastroenterol 2024; 30:4267-4280. [PMID: 39492825 PMCID: PMC11525855 DOI: 10.3748/wjg.v30.i39.4267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common histological type of esophageal cancer with a poor prognosis. Early diagnosis and prognosis assessment are crucial for improving the survival rate of ESCC patients. With the advancement of artificial intelligence (AI) technology and the proliferation of medical digital information, AI has demonstrated promising sensitivity and accuracy in assisting precise detection, treatment decision-making, and prognosis assessment of ESCC. It has become a unique opportunity to enhance comprehensive clinical management of ESCC in the era of precision oncology. This review examines how AI is applied to the diagnosis, treatment, and prognosis assessment of ESCC in the era of precision oncology, and analyzes the challenges and potential opportunities that AI faces in clinical translation. Through insights into future prospects, it is hoped that this review will contribute to the real-world application of AI in future clinical settings, ultimately alleviating the disease burden caused by ESCC.
Collapse
Affiliation(s)
- Wan-Yue Zhang
- School of Medicine, Southeast University, Nanjing 221000, Jiangsu Province, China
| | - Yong-Jian Chang
- School of Cyber Science and Engineering, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Rui-Hua Shi
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
9
|
Loeffler CML, El Nahhas OSM, Muti HS, Carrero ZI, Seibel T, van Treeck M, Cifci D, Gustav M, Bretz K, Gaisa NT, Lehmann KV, Leary A, Selenica P, Reis-Filho JS, Ortiz-Bruechle N, Kather JN. Prediction of homologous recombination deficiency from routine histology with attention-based multiple instance learning in nine different tumor types. BMC Biol 2024; 22:225. [PMID: 39379982 PMCID: PMC11462727 DOI: 10.1186/s12915-024-02022-9] [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: 01/08/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Homologous recombination deficiency (HRD) is recognized as a pan-cancer predictive biomarker that potentially indicates who could benefit from treatment with PARP inhibitors (PARPi). Despite its clinical significance, HRD testing is highly complex. Here, we investigated in a proof-of-concept study whether Deep Learning (DL) can predict HRD status solely based on routine hematoxylin & eosin (H&E) histology images across nine different cancer types. METHODS We developed a deep learning pipeline with attention-weighted multiple instance learning (attMIL) to predict HRD status from histology images. As part of our approach, we calculated a genomic scar HRD score by combining loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transitions (LST) from whole genome sequencing (WGS) data of n = 5209 patients across two independent cohorts. The model's effectiveness was evaluated using the area under the receiver operating characteristic curve (AUROC), focusing on its accuracy in predicting genomic HRD against a clinically recognized cutoff value. RESULTS Our study demonstrated the predictability of genomic HRD status in endometrial, pancreatic, and lung cancers reaching cross-validated AUROCs of 0.79, 0.58, and 0.66, respectively. These predictions generalized well to an external cohort, with AUROCs of 0.93, 0.81, and 0.73. Moreover, a breast cancer-trained image-based HRD classifier yielded an AUROC of 0.78 in the internal validation cohort and was able to predict HRD in endometrial, prostate, and pancreatic cancer with AUROCs of 0.87, 0.84, and 0.67, indicating that a shared HRD-like phenotype occurs across these tumor entities. CONCLUSIONS This study establishes that HRD can be directly predicted from H&E slides using attMIL, demonstrating its applicability across nine different tumor types.
Collapse
Affiliation(s)
- Chiara Maria Lavinia Loeffler
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine I, Faculty of Medicine Carl Gustav Carus, University Hospitaland, Technische Universität Dresden , Dresden, Germany
| | - Omar S M El Nahhas
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Hannah Sophie Muti
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department for Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tobias Seibel
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Marko van Treeck
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Didem Cifci
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Marco Gustav
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Kevin Bretz
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Nadine T Gaisa
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
- Joint Research Center Computational Biomedicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Kjong-Van Lehmann
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
- Joint Research Center Computational Biomedicine, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Duesseldorf, Germany
- Cancer Research Center Cologne-Essen, University Hospital Cologne, Cologne, Germany
| | - Alexandra Leary
- Gynecological Cancer Unit, Department of Medicine, Institut Gustave Roussy, Villejuif, France
| | - Pier Selenica
- Experimental Pathology, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge S Reis-Filho
- Experimental Pathology, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadina Ortiz-Bruechle
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Duesseldorf, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, Faculty of Medicine Carl Gustav Carus, University Hospitaland, Technische Universität Dresden , Dresden, 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.
| |
Collapse
|
10
|
Rao L, Yuan Y, Shen X, Yu G, Chen X. Designing nanotheranostics with machine learning. NATURE NANOTECHNOLOGY 2024:10.1038/s41565-024-01753-8. [PMID: 39362960 DOI: 10.1038/s41565-024-01753-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/08/2024] [Indexed: 10/05/2024]
Abstract
The inherent limits of traditional diagnoses and therapies have driven the development and application of emerging nanotechnologies for more effective and safer management of diseases, herein referred to as 'nanotheranostics'. Although many important technological successes have been achieved in this field, widespread adoption of nanotheranostics as a new paradigm is hindered by specific obstacles, including time-consuming synthesis of nanoparticles, incomplete understanding of nano-bio interactions, and challenges regarding chemistry, manufacturing and the controls required for clinical translation and commercialization. As a key branch of artificial intelligence, machine learning (ML) provides a set of tools capable of performing time-consuming and result-perception tasks, thus offering unique opportunities for nanotheranostics. This Review summarizes the progress and challenges in this emerging field of ML-aided nanotheranostics, and discusses the opportunities in developing next-generation nanotheranostics with reliable datasets and advanced ML models to offer better clinical benefits to patients.
Collapse
Affiliation(s)
- Lang Rao
- Shenzhen Bay Laboratory, Shenzhen, China.
| | - Yuan Yuan
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Computer Science, Boston College, Chestnut Hill, MA, USA
| | - Xi Shen
- Tencent AI Lab, Shenzhen, China
- Intellindust, Shenzhen, China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
11
|
Lu MY, Chen B, Williamson DFK, Chen RJ, Zhao M, Chow AK, Ikemura K, Kim A, Pouli D, Patel A, Soliman A, Chen C, Ding T, Wang JJ, Gerber G, Liang I, Le LP, Parwani AV, Weishaupt LL, Mahmood F. A multimodal generative AI copilot for human pathology. Nature 2024; 634:466-473. [PMID: 38866050 PMCID: PMC11464372 DOI: 10.1038/s41586-024-07618-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
Computational pathology1,2 has witnessed considerable progress in the development of both task-specific predictive models and task-agnostic self-supervised vision encoders3,4. However, despite the explosive growth of generative artificial intelligence (AI), there have been few studies on building general-purpose multimodal AI assistants and copilots5 tailored to pathology. Here we present PathChat, a vision-language generalist AI assistant for human pathology. We built PathChat by adapting a foundational vision encoder for pathology, combining it with a pretrained large language model and fine-tuning the whole system on over 456,000 diverse visual-language instructions consisting of 999,202 question and answer turns. We compare PathChat with several multimodal vision-language AI assistants and GPT-4V, which powers the commercially available multimodal general-purpose AI assistant ChatGPT-4 (ref. 6). PathChat achieved state-of-the-art performance on multiple-choice diagnostic questions from cases with diverse tissue origins and disease models. Furthermore, using open-ended questions and human expert evaluation, we found that overall PathChat produced more accurate and pathologist-preferable responses to diverse queries related to pathology. As an interactive vision-language AI copilot that can flexibly handle both visual and natural language inputs, PathChat may potentially find impactful applications in pathology education, research and human-in-the-loop clinical decision-making.
Collapse
Affiliation(s)
- Ming Y Lu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Bowen Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Drew F K Williamson
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Richard J Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Melissa Zhao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron K Chow
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Kenji Ikemura
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ahrong Kim
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Pusan National University, Busan, South Korea
| | - Dimitra Pouli
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ankush Patel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Amr Soliman
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Chengkuan Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tong Ding
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Judy J Wang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Georg Gerber
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ivy Liang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Long Phi Le
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anil V Parwani
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Luca L Weishaupt
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA, USA
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
12
|
Wang X, Zhao J, Marostica E, Yuan W, Jin J, Zhang J, Li R, Tang H, Wang K, Li Y, Wang F, Peng Y, Zhu J, Zhang J, Jackson CR, Zhang J, Dillon D, Lin NU, Sholl L, Denize T, Meredith D, Ligon KL, Signoretti S, Ogino S, Golden JA, Nasrallah MP, Han X, Yang S, Yu KH. A pathology foundation model for cancer diagnosis and prognosis prediction. Nature 2024; 634:970-978. [PMID: 39232164 DOI: 10.1038/s41586-024-07894-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 08/01/2024] [Indexed: 09/06/2024]
Abstract
Histopathology image evaluation is indispensable for cancer diagnoses and subtype classification. Standard artificial intelligence methods for histopathology image analyses have focused on optimizing specialized models for each diagnostic task1,2. Although such methods have achieved some success, they often have limited generalizability to images generated by different digitization protocols or samples collected from different populations3. Here, to address this challenge, we devised the Clinical Histopathology Imaging Evaluation Foundation (CHIEF) model, a general-purpose weakly supervised machine learning framework to extract pathology imaging features for systematic cancer evaluation. CHIEF leverages two complementary pretraining methods to extract diverse pathology representations: unsupervised pretraining for tile-level feature identification and weakly supervised pretraining for whole-slide pattern recognition. We developed CHIEF using 60,530 whole-slide images spanning 19 anatomical sites. Through pretraining on 44 terabytes of high-resolution pathology imaging datasets, CHIEF extracted microscopic representations useful for cancer cell detection, tumour origin identification, molecular profile characterization and prognostic prediction. We successfully validated CHIEF using 19,491 whole-slide images from 32 independent slide sets collected from 24 hospitals and cohorts internationally. Overall, CHIEF outperformed the state-of-the-art deep learning methods by up to 36.1%, showing its ability to address domain shifts observed in samples from diverse populations and processed by different slide preparation methods. CHIEF provides a generalizable foundation for efficient digital pathology evaluation for patients with cancer.
Collapse
Affiliation(s)
- Xiyue Wang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Junhan Zhao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eliana Marostica
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Boston, MA, USA
| | - Wei Yuan
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Jietian Jin
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiayu Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongping Tang
- Department of Pathology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Kanran Wang
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yu Li
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China
| | - Fang Wang
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yulong Peng
- Department of Pathology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Junyou Zhu
- Department of Burn, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Christopher R Jackson
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Pennsylvania State University, Hummelstown, PA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Deborah Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thomas Denize
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Meredith
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Keith L Ligon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shuji Ogino
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jeffrey A Golden
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - MacLean P Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Sen Yang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
13
|
Kresbach C, Hack K, Ricklefs F, Schüller U. Specifics of spinal neuropathology in the molecular age. Neurooncol Adv 2024; 6:iii3-iii12. [PMID: 39430396 PMCID: PMC11485660 DOI: 10.1093/noajnl/vdad127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024] Open
Abstract
Tumors located in the spinal cord and its coverings can be diagnostically challenging and require special consideration regarding treatment options. During the last decade, important advances regarding the molecular characterization of central and peripheral nervous system tumors were achieved, resulting in improved diagnostic precision, and understanding of the tumor spectrum of this compartment. In particular, array-based global DNA methylation profiling has emerged as a valuable tool to delineate biologically and clinically relevant tumor subgroups and has been incorporated in the current WHO classification for central nervous system tumors of 2021. In addition, several genetic drivers have been described, which may also help to define distinct tumor types and subtypes. Importantly, the current molecular understanding not only sharpens diagnostic precision but also provides the opportunity to investigate both targeted therapies as well as risk-adapted changes in treatment intensity. Here, we discuss the current knowledge and the clinical relevance of molecular neuropathology in spinal tumor entities.
Collapse
Affiliation(s)
- Catena Kresbach
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karoline Hack
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
| | - Franz Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
Shams A. Leveraging State-of-the-Art AI Algorithms in Personalized Oncology: From Transcriptomics to Treatment. Diagnostics (Basel) 2024; 14:2174. [PMID: 39410578 PMCID: PMC11476216 DOI: 10.3390/diagnostics14192174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Continuous breakthroughs in computational algorithms have positioned AI-based models as some of the most sophisticated technologies in the healthcare system. AI shows dynamic contributions in advancing various medical fields involving data interpretation and monitoring, imaging screening and diagnosis, and treatment response and survival prediction. Despite advances in clinical oncology, more effort must be employed to tailor therapeutic plans based on each patient's unique transcriptomic profile within the precision/personalized oncology frame. Furthermore, the standard analysis method is not compatible with the comprehensive deciphering of significant data streams, thus precluding the prediction of accurate treatment options. METHODOLOGY We proposed a novel approach that includes obtaining different tumour tissues and preparing RNA samples for comprehensive transcriptomic interpretation using specifically trained, programmed, and optimized AI-based models for extracting large data volumes, refining, and analyzing them. Next, the transcriptomic results will be scanned against an expansive drug library to predict the response of each target to the tested drugs. The obtained target-drug combination/s will be then validated using in vitro and in vivo experimental models. Finally, the best treatment combination option/s will be introduced to the patient. We also provided a comprehensive review discussing AI models' recent innovations and implementations to aid in molecular diagnosis and treatment planning. RESULTS The expected transcriptomic analysis generated by the AI-based algorithms will provide an inclusive genomic profile for each patient, containing statistical and bioinformatics analyses, identification of the dysregulated pathways, detection of the targeted genes, and recognition of molecular biomarkers. Subjecting these results to the prediction and pairing AI-based processes will result in statistical graphs presenting each target's likely response rate to various treatment options. Different in vitro and in vivo investigations will further validate the selection of the target drug/s pairs. CONCLUSIONS Leveraging AI models will provide more rigorous manipulation of large-scale datasets on specific cancer care paths. Such a strategy would shape treatment according to each patient's demand, thus fortifying the avenue of personalized/precision medicine. Undoubtedly, this will assist in improving the oncology domain and alleviate the burden of clinicians in the coming decade.
Collapse
Affiliation(s)
- Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; or ; Tel.: +00966-548638099
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
- High Altitude Research Center, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
15
|
Min W, Shi Z, Zhang J, Wan J, Wang C. Multimodal contrastive learning for spatial gene expression prediction using histology images. Brief Bioinform 2024; 25:bbae551. [PMID: 39471412 DOI: 10.1093/bib/bbae551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/23/2024] [Accepted: 10/14/2024] [Indexed: 11/01/2024] Open
Abstract
In recent years, the advent of spatial transcriptomics (ST) technology has unlocked unprecedented opportunities for delving into the complexities of gene expression patterns within intricate biological systems. Despite its transformative potential, the prohibitive cost of ST technology remains a significant barrier to its widespread adoption in large-scale studies. An alternative, more cost-effective strategy involves employing artificial intelligence to predict gene expression levels using readily accessible whole-slide images stained with Hematoxylin and Eosin (H&E). However, existing methods have yet to fully capitalize on multimodal information provided by H&E images and ST data with spatial location. In this paper, we propose mclSTExp, a multimodal contrastive learning with Transformer and Densenet-121 encoder for Spatial Transcriptomics Expression prediction. We conceptualize each spot as a "word", integrating its intrinsic features with spatial context through the self-attention mechanism of a Transformer encoder. This integration is further enriched by incorporating image features via contrastive learning, thereby enhancing the predictive capability of our model. We conducted an extensive evaluation of highly variable genes in two breast cancer datasets and a skin squamous cell carcinoma dataset, and the results demonstrate that mclSTExp exhibits superior performance in predicting spatial gene expression. Moreover, mclSTExp has shown promise in interpreting cancer-specific overexpressed genes, elucidating immune-related genes, and identifying specialized spatial domains annotated by pathologists. Our source code is available at https://github.com/shizhiceng/mclSTExp.
Collapse
Affiliation(s)
- Wenwen Min
- School of Information Science and Engineering, Yunnan University, East Outer Ring Road, Chenggong District, Kunming 650500, Yunnan, China
| | - Zhiceng Shi
- School of Information Science and Engineering, Yunnan University, East Outer Ring Road, Chenggong District, Kunming 650500, Yunnan, China
| | - Jun Zhang
- School of Information Science and Engineering, Yunnan University, East Outer Ring Road, Chenggong District, Kunming 650500, Yunnan, China
| | - Jun Wan
- School of Information and Engineering, Zhongnan University of Economics and Law, 182 South Lake Avenue, East Lake New Technology Development Zone, Wuhan 430073, Hubei, China
| | - Changmiao Wang
- Medical Big Data, Shenzhen Research Institute of Big Data, Longxiang Boulevard, Longgang District, Shenzhen 518172, Guangdong, China
| |
Collapse
|
16
|
Jiang B, Bao L, He S, Chen X, Jin Z, Ye Y. Deep learning applications in breast cancer histopathological imaging: diagnosis, treatment, and prognosis. Breast Cancer Res 2024; 26:137. [PMID: 39304962 DOI: 10.1186/s13058-024-01895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Breast cancer is the most common malignant tumor among women worldwide and remains one of the leading causes of death among women. Its incidence and mortality rates are continuously rising. In recent years, with the rapid advancement of deep learning (DL) technology, DL has demonstrated significant potential in breast cancer diagnosis, prognosis evaluation, and treatment response prediction. This paper reviews relevant research progress and applies DL models to image enhancement, segmentation, and classification based on large-scale datasets from TCGA and multiple centers. We employed foundational models such as ResNet50, Transformer, and Hover-net to investigate the performance of DL models in breast cancer diagnosis, treatment, and prognosis prediction. The results indicate that DL techniques have significantly improved diagnostic accuracy and efficiency, particularly in predicting breast cancer metastasis and clinical prognosis. Furthermore, the study emphasizes the crucial role of robust databases in developing highly generalizable models. Future research will focus on addressing challenges related to data management, model interpretability, and regulatory compliance, ultimately aiming to provide more precise clinical treatment and prognostic evaluation programs for breast cancer patients.
Collapse
Affiliation(s)
- Bitao Jiang
- Department of Hematology and Oncology, Beilun District People's Hospital, Ningbo, 315800, China.
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital of Zhejiang University, Ningbo, 315800, China.
| | - Lingling Bao
- Department of Hematology and Oncology, Beilun District People's Hospital, Ningbo, 315800, China
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital of Zhejiang University, Ningbo, 315800, China
| | - Songqin He
- Department of Oncology, The 906th Hospital of the Joint Logistics Force of the Chinese People's Liberation Army, Ningbo, 315100, China
| | - Xiao Chen
- Department of Oncology, The 906th Hospital of the Joint Logistics Force of the Chinese People's Liberation Army, Ningbo, 315100, China
| | - Zhihui Jin
- Department of Hematology and Oncology, Beilun District People's Hospital, Ningbo, 315800, China
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital of Zhejiang University, Ningbo, 315800, China
| | - Yingquan Ye
- Department of Oncology, The 906th Hospital of the Joint Logistics Force of the Chinese People's Liberation Army, Ningbo, 315100, China.
| |
Collapse
|
17
|
Wiest IC, Ferber D, Zhu J, van Treeck M, Meyer SK, Juglan R, Carrero ZI, Paech D, Kleesiek J, Ebert MP, Truhn D, Kather JN. Privacy-preserving large language models for structured medical information retrieval. NPJ Digit Med 2024; 7:257. [PMID: 39304709 DOI: 10.1038/s41746-024-01233-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Abstract
Most clinical information is encoded as free text, not accessible for quantitative analysis. This study presents an open-source pipeline using the local large language model (LLM) "Llama 2" to extract quantitative information from clinical text and evaluates its performance in identifying features of decompensated liver cirrhosis. The LLM identified five key clinical features in a zero- and one-shot manner from 500 patient medical histories in the MIMIC IV dataset. We compared LLMs of three sizes and various prompt engineering approaches, with predictions compared against ground truth from three blinded medical experts. Our pipeline achieved high accuracy, detecting liver cirrhosis with 100% sensitivity and 96% specificity. High sensitivities and specificities were also yielded for detecting ascites (95%, 95%), confusion (76%, 94%), abdominal pain (84%, 97%), and shortness of breath (87%, 97%) using the 70 billion parameter model, which outperformed smaller versions. Our study successfully demonstrates the capability of locally deployed LLMs to extract clinical information from free text with low hardware requirements.
Collapse
Affiliation(s)
- Isabella Catharina Wiest
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Dyke Ferber
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
| | - Jiefu Zhu
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Sonja K Meyer
- Department of Surgery I, University Hospital Würzburg, Würzburg, Germany
| | - Radhika Juglan
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Daniel Paech
- German Cancer Research Center, Division of Radiology, Heidelberg, Germany
- University Hospital Bonn, Clinic for Neuroradiology, Bonn, Germany
| | - Jens Kleesiek
- Institut für KI in der Medizin (IKIM), Universitätsmedizin Essen, Girardetstr. 2, 45131, Essen, Germany
- Cancer Research Center Cologne Essen (CCCE), West German Cancer Center Essen (WTZ), 45122, Essen, Germany
- TU Dortmund University, Department of Physics, Otto-Hahn-Straße 4, 44227, Dortmund, Germany
| | - Matthias P Ebert
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at the University Medical Center, Mannheim, Germany
- Molecular Medicine Partnership Unit, EMBL, Heidelberg, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital Aachen, Aachen, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.
- Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307, Dresden, Germany.
| |
Collapse
|
18
|
Kather JN, Abernethy AP. How AI will transform cancer care. Ann Oncol 2024:S0923-7534(24)03915-2. [PMID: 39288845 DOI: 10.1016/j.annonc.2024.08.2335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Affiliation(s)
- J N Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden; Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
19
|
Chen R, Liu Y, Xie J. Construction of a pathomics model for predicting mRNAsi in lung adenocarcinoma and exploration of biological mechanism. Heliyon 2024; 10:e37100. [PMID: 39286147 PMCID: PMC11402732 DOI: 10.1016/j.heliyon.2024.e37100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/04/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Objective This study aimed to predict the level of stemness index (mRNAsi) and survival prognosis of lung adenocarcinoma (LUAD) using pathomics model. Methods From The Cancer Genome Atlas (TCGA) database, 327 LUAD patients were randomly assigned to a training set (n = 229) and a validation set (n = 98) for pathomics model development and evaluation. PyRadiomics was used to extract pathomics features, followed by feature selection using the mRMR-RFE algorithm. In the training set, Gradient Boosting Machine (GBM) was utilized to establish a model for predicting mRNAsi in LUAD. The model's predictive performance was evaluated using ROC curves, calibration curves, and decision curve analysis (DCA). Prognostic analysis was conducted using Kaplan-Meier curves and cox regression. Additionally, gene enrichment analysis, tumor microenvironment analysis, and tumor mutational burden (TMB) analysis were performed to explore the biological mechanisms underlying the pathomics prediction model. Results Multivariable cox analysis (HR = 1.488, 95 % CI 1.012-2.187, P = 0.043) identified mRNAsi as a prognostic risk factor for LUAD. A total of 465 pathomics features were extracted from TCGA-LUAD histopathological images, and ultimately, the most representative 8 features were selected to construct the predictive model. ROC curves demonstrated the significant predictive value of the model for mRNAsi in both the training set (AUC = 0.769) and the validation set (AUC = 0.757). Calibration curves and Hosmer-Lemeshow goodness-of-fit test showed good consistency between the model's prediction of mRNAsi levels and the actual values. DCA indicated a good net benefit of the model. The prediction of mRNAsi levels by the pathomics model is represented using the pathomics score (PS). PS was strongly associated with the prognosis of LUAD (HR = 1.496, 95 % CI 1.008-2.222, P = 0.046). Signaling pathways related to DNA replication and damage repair were significantly enriched in the high PS group. Prediction of immune therapy response indicated significantly reduced Dysfunction in the high PS group (P < 0.001). The high PS group exhibited higher TMB values (P < 0.001). Conclusions The predictive model constructed based on pathomics features can forecast the mRNAsi and survival risk of LUAD. This model holds promise to aid clinical practitioners in identifying high-risk patients and devising more optimized treatment plans for patients by jointly employing therapeutic strategies targeting cancer stem cells (CSCs).
Collapse
Affiliation(s)
- Rui Chen
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Yuzhen Liu
- Department of Oncology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Junping Xie
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
20
|
Cheng H, Xu H, Peng B, Huang X, Hu Y, Zheng C, Zhang Z. Illuminating the future of precision cancer surgery with fluorescence imaging and artificial intelligence convergence. NPJ Precis Oncol 2024; 8:196. [PMID: 39251820 PMCID: PMC11385925 DOI: 10.1038/s41698-024-00699-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Real-time and accurate guidance for tumor resection has long been anticipated by surgeons. In the past decade, the flourishing material science has made impressive progress in near-infrared fluorophores that may fulfill this purpose. Fluorescence imaging-guided surgery shows great promise for clinical application and has undergone widespread evaluations, though it still requires continuous improvements to transition this technique from bench to bedside. Concurrently, the rapid progress of artificial intelligence (AI) has revolutionized medicine, aiding in the screening, diagnosis, and treatment of human doctors. Incorporating AI helps enhance fluorescence imaging and is poised to bring major innovations to surgical guidance, thereby realizing precision cancer surgery. This review provides an overview of the principles and clinical evaluations of fluorescence-guided surgery. Furthermore, recent endeavors to synergize AI with fluorescence imaging were presented, and the benefits of this interdisciplinary convergence were discussed. Finally, several implementation strategies to overcome technical hurdles were proposed to encourage and inspire future research to expedite the clinical application of these revolutionary technologies.
Collapse
Affiliation(s)
- Han Cheng
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China
| | - Hongtao Xu
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China
| | - Boyang Peng
- School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
| | - Xiaojuan Huang
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China
| | - Yongjie Hu
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China
| | - Chongyang Zheng
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China.
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China.
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China.
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China.
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China.
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China.
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China.
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China.
| |
Collapse
|
21
|
Li M, Xiong X, Xu B. Attitudes and perceptions of Chinese oncologists towards artificial intelligence in healthcare: a cross-sectional survey. Front Digit Health 2024; 6:1371302. [PMID: 39290363 PMCID: PMC11405309 DOI: 10.3389/fdgth.2024.1371302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
Background Artificial intelligence (AI) is transforming healthcare, yet little is known about Chinese oncologists' attitudes towards AI. This study investigated oncologists' knowledge, perceptions, and acceptance of AI in China. Methods A cross-sectional online survey was conducted among 228 oncologists across China. The survey examined demographics, AI exposure, knowledge and attitudes using 5-point Likert scales, and factors influencing AI adoption. Data were analyzed using descriptive statistics and chi-square tests. Results Respondents showed moderate understanding of AI concepts (mean 3.39/5), with higher knowledge among younger oncologists. Only 12.8% used ChatGPT. Most (74.13%) agreed AI is beneficial and could innovate healthcare, 52.19% respondents expressed trust in AI technology. Acceptance was cautiously optimistic (mean 3.57/5). Younger respondents (∼30) show significantly higher trust (p = 0.004) and acceptance (p = 0.009) of AI compared to older respondents, while trust is significantly higher among those with master's or doctorate vs. bachelor's degrees (p = 0.032), and acceptance is higher for those with prior IT experience (p = 0.035).Key drivers for AI adoption were improving efficiency (85.09%), quality (85.53%), reducing errors (84.65%), and enabling new approaches (73.25%). Conclusions Chinese oncologists are open to healthcare AI but remain prudently optimistic given limitations. Targeted education, especially for older oncologists, can facilitate AI implementation. AI is largely welcomed for its potential to augment human roles in enhancing efficiency, quality, safety, and innovations in oncology practice.
Collapse
Affiliation(s)
- Ming Li
- Department of Health Policy Management, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Xiaomin Xiong
- Department of Breast Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Institute of Intelligent Oncology, Chongqing University, Chongqing, China
| | - Bo Xu
- Department of Breast Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Institute of Intelligent Oncology, Chongqing University, Chongqing, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Department of Biochemistry and Molecular Biology, Ministry of Education, National Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
22
|
Sherry AD, Passy AH, McCaw ZR, Abi Jaoude J, Lin TA, Kouzy R, Miller AM, Kupferman GS, Beck EJ, Msaouel P, Ludmir EB. Increasing Power in Phase III Oncology Trials With Multivariable Regression: An Empirical Assessment of 535 Primary End Point Analyses. JCO Clin Cancer Inform 2024; 8:e2400102. [PMID: 39213473 PMCID: PMC11371366 DOI: 10.1200/cci.24.00102] [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: 04/26/2024] [Revised: 06/28/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
PURPOSE A previous study demonstrated that power against the (unobserved) true effect for the primary end point (PEP) of most phase III oncology trials is low, suggesting an increased risk of false-negative findings in the field of late-phase oncology. Fitting models with prognostic covariates is a potential solution to improve power; however, the extent to which trials leverage this approach, and its impact on trial interpretation at scale, is unknown. To that end, we hypothesized that phase III trials using multivariable PEP analyses are more likely to demonstrate superiority versus trials with univariable analyses. METHODS PEP analyses were reviewed from trials registered on ClinicalTrials.gov. Adjusted odds ratios (aORs) were calculated by logistic regressions. RESULTS Of the 535 trials enrolling 454,824 patients, 69% (n = 368) used a multivariable PEP analysis. Trials with multivariable PEP analyses were more likely to demonstrate PEP superiority (57% [209 of 368] v 42% [70 of 167]; aOR, 1.78 [95% CI, 1.18 to 2.72]; P = .007). Among trials with a multivariable PEP model, 16 conditioned on covariates and 352 stratified by covariates. However, 108 (35%) of 312 trials with stratified analyses lost power by categorizing a continuous variable, which was especially common among immunotherapy trials (aOR, 2.45 [95% CI, 1.23 to 4.92]; P = .01). CONCLUSION Trials increasing power by fitting multivariable models were more likely to demonstrate PEP superiority than trials with unadjusted analysis. Underutilization of conditioning models and empirical power loss associated with covariate categorization required by stratification were identified as barriers to power gains. These findings underscore the opportunity to increase power in phase III trials with conventional methodology and improve patient access to effective novel therapies.
Collapse
Affiliation(s)
- Alexander D Sherry
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Adina H Passy
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zachary R McCaw
- Insitro, South San Francisco, CA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Timothy A Lin
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ramez Kouzy
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Avital M Miller
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gabrielle S Kupferman
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Esther J Beck
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ethan B Ludmir
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
23
|
Braun Y, Friedmacher F, Theilen TM, Fiegel HC, Weber K, Harter PN, Rolle U. Diagnosis of Hirschsprung disease by analyzing acetylcholinesterase staining using artificial intelligence. J Pediatr Gastroenterol Nutr 2024; 79:729-737. [PMID: 39118474 DOI: 10.1002/jpn3.12339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/15/2024] [Accepted: 07/21/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVES Classical Hirschsprung disease (HD) is defined by the absence of ganglion cells in the rectosigmoid colon. The diagnosis is made from rectal biopsy, which reveals the aganglionosis and the presence of cholinergic hyperinnervation. However, depending on the method of rectal biopsy, the quality of the specimens and the related diagnostic accuracy varies substantially. To facilitate and objectify the diagnosis of HD, we investigated whether software-based identification of cholinergic hyperinnervation in digitalized histopathology slides is suitable for distinguishing healthy individuals from affected individuals. METHODS N = 190 samples of 112 patients who underwent open surgical rectal biopsy at our pediatric surgery center between 2009 and 2019 were included in this study. Acetylcholinesterase (AChE) stained slides of these samples were collected and digitalized via slide scanning and analyzed using two digital imaging software programs (HALO, QuPath). The AChE-positive staining area in the mucosal layers of the intestinal wall was determined. In the next step machine learning was employed to identify patterns of cholinergic hyperinnervation. RESULTS The area of AChE-positive staining was greater in HD patients compared to healthy individuals (p < 0.0001). Artificial intelligence-based assessment of parasympathetic hyperinnervation identified HD with a high precision (area under the curve [AUC] 0.96). The accuracy of the prediction model increased when nonrectal samples were excluded (AUC 0.993). CONCLUSIONS Software-assisted machine-learning analysis of AChE staining is suitable to improve the diagnostic accuracy of HD.
Collapse
Affiliation(s)
- Yannick Braun
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Florian Friedmacher
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Till-Martin Theilen
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Henning C Fiegel
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Katharina Weber
- Neurological Institute, Edinger Institute, Neuropathology, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Center for Tumor Diseases, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Patrick N Harter
- Neurological Institute, Edinger Institute, Neuropathology, Goethe University, Frankfurt am Main, Germany
- Centre for Neuropathology and Prion-Research, Ludwig-Maximilians-Universität München, München, Germany
| | - Udo Rolle
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
24
|
Zhu M, Sali R, Baba F, Khasawneh H, Ryndin M, Leveillee RJ, Hurwitz MD, Lui K, Dixon C, Zhang DY. Artificial intelligence in pathologic diagnosis, prognosis and prediction of prostate cancer. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2024; 12:200-215. [PMID: 39308594 PMCID: PMC11411179 DOI: 10.62347/jsae9732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024]
Abstract
Histopathology, which is the gold-standard for prostate cancer diagnosis, faces significant challenges. With prostate cancer ranking among the most common cancers in the United States and worldwide, pathologists experience an increased number for prostate biopsies. At the same time, precise pathological assessment and classification are necessary for risk stratification and treatment decisions in prostate cancer care, adding to the challenge to pathologists. Recent advancement in digital pathology makes artificial intelligence and learning tools adopted in histopathology feasible. In this review, we introduce the concept of AI and its various techniques in the field of histopathology. We summarize the clinical applications of AI pathology for prostate cancer, including pathological diagnosis, grading, prognosis evaluation, and treatment options. We also discuss how AI applications can be integrated into the routine pathology workflow. With these rapid advancements, it is evident that AI applications in prostate cancer go beyond the initial goal of being tools for diagnosis and grading. Instead, pathologists can provide additional information to improve long-term patient outcomes by assessing detailed histopathologic features at pixel level using digital pathology and AI. Our review not only provides a comprehensive summary of the existing research but also offers insights for future advancements.
Collapse
Affiliation(s)
- Min Zhu
- Department of Computational Pathology, NovinoAI1443 NE 4th Ave, Fort Lauderdale, FL 33304, USA
| | - Rasoul Sali
- Department of Computational Pathology, NovinoAI1443 NE 4th Ave, Fort Lauderdale, FL 33304, USA
- Department of Radiation Oncology, Stanford University School of MedicineStanford, CA 94305, USA
| | - Firas Baba
- Department of Computational Pathology, NovinoAI1443 NE 4th Ave, Fort Lauderdale, FL 33304, USA
| | - Hamdi Khasawneh
- King Hussein School of Computing Sciences, Princess Sumaya University for TechnologyAmman 11855, Jordan
| | - Michelle Ryndin
- College of Agriculture and Life Sciences, Cornell University616 Thurston Ave, Ithaca, NY 14853, USA
| | - Raymond J Leveillee
- Department of Surgery, Florida Atlantic University, Division of Urology, Bethesda Hospital East, Baptist Health South Florida2800 S. Seacrest Drive, Boynton Beach, FL 33435, USA
| | - Mark D Hurwitz
- Department of Radiation Medicine, New York Medical College and Westchester Medical CenterValhalla, NY 10595, USA
| | - Kin Lui
- Department of Urology, Mount Sinai HospitalNew York, NY 10029, USA
| | - Christopher Dixon
- Department of Urology, Good Samaritan Hospital, Westchester Medical Center Health NetworkSuffern, NY 10901, USA
| | - David Y Zhang
- Department of Computational Pathology, NovinoAI1443 NE 4th Ave, Fort Lauderdale, FL 33304, USA
- Pathology and Laboratory Services, Department of Veterans Affairs New York Harbor Healthcare SystemNew York, NY 10010, USA
| |
Collapse
|
25
|
Guffanti F, Mengoli I, Damia G. Current HRD assays in ovarian cancer: differences, pitfalls, limitations, and novel approaches. Front Oncol 2024; 14:1405361. [PMID: 39220639 PMCID: PMC11361952 DOI: 10.3389/fonc.2024.1405361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Ovarian carcinoma (OC) still represents an insidious and fatal malignancy, and few significant results have been obtained in the last two decades to improve patient survival. Novel targeted therapies such as poly (ADP-ribose) polymerase inhibitors (PARPi) have been successfully introduced in the clinical management of OC, but not all patients will benefit, and drug resistance almost inevitably occurs. The identification of patients who are likely to respond to PARPi-based therapies relies on homologous recombination deficiency (HRD) tests, as this condition is associated with response to these treatments. This review summarizes the genomic and functional HRD assays currently used in clinical practice and those under evaluation, the clinical implications of HRD testing in OC, and their current pitfalls and limitations. Special emphasis will be placed on the functional HRD assays under development and the use of machine learning and artificial intelligence technologies as novel strategies to overcome the current limitations of HRD tests for a better-personalized treatment to improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Giovanna Damia
- Laboratory of Preclinical Gynaecological Oncology, Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
26
|
Maurya SP, Sisodia PS, Mishra R, Singh DP. Performance of machine learning algorithms for lung cancer prediction: a comparative approach. Sci Rep 2024; 14:18562. [PMID: 39122762 PMCID: PMC11316115 DOI: 10.1038/s41598-024-58345-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/27/2024] [Indexed: 08/12/2024] Open
Abstract
Due to the excessive growth of PM 2.5 in aerosol, the cases of lung cancer are increasing rapidly and are most severe among other types as the highest mortality rate. In most of the cases, lung cancer is detected with least symptoms at its later stage. Hence, clinical records may play a vital role to diagnose this disease at the correct stage for suitable medication to cure it. To detect lung cancer an accurate prediction method is needed which is significantly reliable. In the digital clinical record era with advancement in computing algorithms including machine learning techniques opens an opportunity to ease the process. Various machine learning algorithms may be applied over realistic clinical data but the predictive power is yet to be comprehended for accurate results. This paper envisages to compare twelve potential machine learning algorithms over clinical data with eleven symptoms of lung cancer along with two major habits of patients to predict a positive case accurately. The result has been found based on classification and heat map correlation. K-Nearest Neighbor Model and Bernoulli Naive Bayes Model are found most significant methods for early lung cancer prediction.
Collapse
Affiliation(s)
- Satya Prakash Maurya
- Department of Computer Science and Engineering, Graphic Era (Deemed to be University), Dehradun, India
| | | | - Rahul Mishra
- Department of Electronics and Computer Engineering, National Institute of Advanced Manufacturing Technology (NIAMT), Ranchi, India.
| | - Devesh Pratap Singh
- Department of Computer Science and Engineering, Graphic Era (Deemed to be University), Dehradun, India
| |
Collapse
|
27
|
Hölscher DL, Bülow RD. Decoding pathology: the role of computational pathology in research and diagnostics. Pflugers Arch 2024:10.1007/s00424-024-03002-2. [PMID: 39095655 DOI: 10.1007/s00424-024-03002-2] [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: 04/18/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
Traditional histopathology, characterized by manual quantifications and assessments, faces challenges such as low-throughput and inter-observer variability that hinder the introduction of precision medicine in pathology diagnostics and research. The advent of digital pathology allowed the introduction of computational pathology, a discipline that leverages computational methods, especially based on deep learning (DL) techniques, to analyze histopathology specimens. A growing body of research shows impressive performances of DL-based models in pathology for a multitude of tasks, such as mutation prediction, large-scale pathomics analyses, or prognosis prediction. New approaches integrate multimodal data sources and increasingly rely on multi-purpose foundation models. This review provides an introductory overview of advancements in computational pathology and discusses their implications for the future of histopathology in research and diagnostics.
Collapse
Affiliation(s)
- David L Hölscher
- Department for Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
- Institute for Pathology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Roman D Bülow
- Institute for Pathology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany.
| |
Collapse
|
28
|
Calderaro J, Žigutytė L, Truhn D, Jaffe A, Kather JN. Artificial intelligence in liver cancer - new tools for research and patient management. Nat Rev Gastroenterol Hepatol 2024; 21:585-599. [PMID: 38627537 DOI: 10.1038/s41575-024-00919-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 07/31/2024]
Abstract
Liver cancer has high incidence and mortality globally. Artificial intelligence (AI) has advanced rapidly, influencing cancer care. AI systems are already approved for clinical use in some tumour types (for example, colorectal cancer screening). Crucially, research demonstrates that AI can analyse histopathology, radiology and natural language in liver cancer, and can replace manual tasks and access hidden information in routinely available clinical data. However, for liver cancer, few of these applications have translated into large-scale clinical trials or clinically approved products. Here, we advocate for the incorporation of AI in all stages of liver cancer management. We present a taxonomy of AI approaches in liver cancer, highlighting areas with academic and commercial potential, and outline a policy for AI-based liver cancer management, including interdisciplinary training of researchers, clinicians and patients. The potential of AI in liver cancer is immense, but effort is required to ensure that AI can fulfil expectations.
Collapse
Affiliation(s)
- Julien Calderaro
- Département de Pathologie, Assistance Publique Hôpitaux de Paris, Groupe Hospitalier Henri Mondor, Créteil, France
- Institut Mondor de Recherche Biomédicale, MINT-HEP Mondor Integrative Hepatology, Université Paris Est Créteil, Créteil, France
| | - Laura Žigutytė
- Else Kroener Fresenius Center for Digital Health (EKFZ), Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Ariel Jaffe
- Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health (EKFZ), Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Dresden, Germany.
- Medical Oncology, National Center for Tumour Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
29
|
Zheng Q, Wang X, Yang R, Fan J, Yuan J, Liu X, Wang L, Xiao Z, Chen Z. Predicting tumor mutation burden and VHL mutation from renal cancer pathology slides with self-supervised deep learning. Cancer Med 2024; 13:e70112. [PMID: 39166457 PMCID: PMC11336896 DOI: 10.1002/cam4.70112] [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: 01/05/2024] [Revised: 05/28/2024] [Accepted: 08/04/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Tumor mutation burden (TMB) and VHL mutation play a crucial role in the management of patients with clear cell renal cell carcinoma (ccRCC), such as guiding adjuvant chemotherapy and improving clinical outcomes. However, the time-consuming and expensive high-throughput sequencing methods severely limit their clinical applicability. Predicting intratumoral heterogeneity poses significant challenges in biology and clinical settings. Our aimed to develop a self-supervised attention-based multiple instance learning (SSL-ABMIL) model to predict TMB and VHL mutation status from hematoxylin and eosin-stained histopathological images. METHODS We obtained whole slide images (WSIs) and somatic mutation data of 350 ccRCC patients from The Cancer Genome Atlas for developing SSL-ABMIL model. In parallel, 163 ccRCC patients from Clinical Proteomic Tumor Analysis Consortium cohort was used as independent external validation set. We systematically compared three different models (Wang-ABMIL, Ciga-ABMIL, and ImageNet-MIL) for their ability to predict TMB and VHL alterations. RESULTS We first identified two groups of populations with high- and low-TMB (cut-off point = 0.9). In two independent cohorts, the Wang-ABMIL model achieved the highest performance with decent generalization performance (AUROC = 0.83 ± 0.02 and 0.8 ± 0.04 in predicting TMB and VHL, respectively). Attention heatmaps revealed that the Wang-ABMIL model paid the highest attention to tumor regions in high-TMB patients, while in VHL mutation prediction, non-tumor regions were also assigned high attention, particularly the stromal regions infiltrated by lymphocytes. CONCLUSIONS Our results indicated that SSL-ABMIL can effectively extract histological features for predicting TMB and VHL mutation, demonstrating promising results in linking tumor morphology and molecular biology.
Collapse
Affiliation(s)
- Qingyuan Zheng
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Xinyu Wang
- Centre for Reproductive ScienceRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Rui Yang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Junjie Fan
- University of Chinese Academy of SciencesBeijingChina
- Trusted Computing and Information Assurance LaboratoryInstitute of Software, Chinese Academy of SciencesBeijingChina
| | - Jingping Yuan
- Department of PathologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Xiuheng Liu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Lei Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zhuoni Xiao
- Centre for Reproductive ScienceRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zhiyuan Chen
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| |
Collapse
|
30
|
Jaume G, de Brot S, Song AH, Williamson DFK, Oldenburg L, Zhang A, Chen RJ, Asin J, Blatter S, Dettwiler M, Goepfert C, Grau-Roma L, Soto S, Keller SM, Rottenberg S, del-Pozo J, Pettit R, Le LP, Mahmood F. Deep Learning-based Modeling for Preclinical Drug Safety Assessment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.20.604430. [PMID: 39091793 PMCID: PMC11291027 DOI: 10.1101/2024.07.20.604430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
In drug development, assessing the toxicity of candidate compounds is crucial for successfully transitioning from preclinical research to early-stage clinical trials. Drug safety is typically assessed using animal models with a manual histopathological examination of tissue sections to characterize the dose-response relationship of the compound - a time-intensive process prone to inter-observer variability and predominantly involving tedious review of cases without abnormalities. Artificial intelligence (AI) methods in pathology hold promise to accelerate this assessment and enhance reproducibility and objectivity. Here, we introduce TRACE, a model designed for toxicologic liver histopathology assessment capable of tackling a range of diagnostic tasks across multiple scales, including situations where labeled data is limited. TRACE was trained on 15 million histopathology images extracted from 46,734 digitized tissue sections from 157 preclinical studies conducted on Rattus norvegicus. We show that TRACE can perform various downstream toxicology tasks spanning histopathological response assessment, lesion severity scoring, morphological retrieval, and automatic dose-response characterization. In an independent reader study, TRACE was evaluated alongside ten board-certified veterinary pathologists and achieved higher concordance with the consensus opinion than the average of the pathologists. Our study represents a substantial leap over existing computational models in toxicology by offering the first framework for accelerating and automating toxicological pathology assessment, promoting significant progress with faster, more consistent, and reliable diagnostic processes.
Collapse
Affiliation(s)
- Guillaume Jaume
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | - Simone de Brot
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Switzerland
| | - Andrew H. Song
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | - Drew F. K. Williamson
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Lukas Oldenburg
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Andrew Zhang
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA
| | - Richard J. Chen
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | - Javier Asin
- California Animal Health and Food Safety Laboratory, University of California-Davis, San Bernardino, CA
- School of Veterinary Medicine, Department of Pathology, University of California-Davis, Davis, CA
| | - Sohvi Blatter
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
| | | | - Christine Goepfert
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
| | - Llorenç Grau-Roma
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
| | - Sara Soto
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
| | | | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Switzerland
- Department for BioMedical Research, University of Bern, Switzerland
| | - Jorge del-Pozo
- Royal (Dick) School of Veterinary Studies, Roslin, United-Kingdom
| | - Rowland Pettit
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Long Phi Le
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA
| |
Collapse
|
31
|
Li L, Sun M, Wang J, Wan S. Multi-omics based artificial intelligence for cancer research. Adv Cancer Res 2024; 163:303-356. [PMID: 39271266 DOI: 10.1016/bs.acr.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
With significant advancements of next generation sequencing technologies, large amounts of multi-omics data, including genomics, epigenomics, transcriptomics, proteomics, and metabolomics, have been accumulated, offering an unprecedented opportunity to explore the heterogeneity and complexity of cancer across various molecular levels and scales. One of the promising aspects of multi-omics lies in its capacity to offer a holistic view of the biological networks and pathways underpinning cancer, facilitating a deeper understanding of its development, progression, and response to treatment. However, the exponential growth of data generated by multi-omics studies present significant analytical challenges. Processing, analyzing, integrating, and interpreting these multi-omics datasets to extract meaningful insights is an ambitious task that stands at the forefront of current cancer research. The application of artificial intelligence (AI) has emerged as a powerful solution to these challenges, demonstrating exceptional capabilities in deciphering complex patterns and extracting valuable information from large-scale, intricate omics datasets. This review delves into the synergy of AI and multi-omics, highlighting its revolutionary impact on oncology. We dissect how this confluence is reshaping the landscape of cancer research and clinical practice, particularly in the realms of early detection, diagnosis, prognosis, treatment and pathology. Additionally, we elaborate the latest AI methods for multi-omics integration to provide a comprehensive insight of the complex biological mechanisms and inherent heterogeneity of cancer. Finally, we discuss the current challenges of data harmonization, algorithm interpretability, and ethical considerations. Addressing these challenges necessitates a multidisciplinary collaboration, paving the promising way for more precise, personalized, and effective treatments for cancer patients.
Collapse
Affiliation(s)
- Lusheng Li
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mengtao Sun
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jieqiong Wang
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shibiao Wan
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
32
|
Hamada K, Murakami R, Ueda A, Kashima Y, Miyagawa C, Taki M, Yamanoi K, Yamaguchi K, Hamanishi J, Minamiguchi S, Matsumura N, Mandai M. A Deep Learning-Based Assessment Pipeline for Intraepithelial and Stromal Tumor-Infiltrating Lymphocytes in High-Grade Serous Ovarian Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1272-1284. [PMID: 38537936 DOI: 10.1016/j.ajpath.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/22/2024] [Accepted: 02/21/2024] [Indexed: 04/07/2024]
Abstract
Tumor-infiltrating lymphocytes (TILs) are associated with improved survival in patients with epithelial ovarian cancer. However, TIL evaluation has not been used in routine clinical practice because of reproducibility issues. The current study developed two convolutional neural network models to detect TILs and to determine their spatial location in whole slide images, and established a spatial assessment pipeline to objectively quantify intraepithelial and stromal TILs in patients with high-grade serous ovarian carcinoma. The predictions of the established models showed a significant positive correlation with the number of CD8+ T cells and immune gene expressions. Patients with a higher density of intraepithelial TILs had a significantly prolonged overall survival and progression-free survival in multiple cohorts. On the basis of the density of intraepithelial and stromal TILs, patients were classified into three immunophenotypes: immune inflamed, excluded, and desert. The immune-desert subgroup showed the worst prognosis. Gene expression analysis showed that the immune-desert subgroup had lower immune cytolytic activity and T-cell-inflamed gene-expression profile scores, whereas the immune-excluded subgroup had higher expression of interferon-γ and programmed death 1 receptor signaling pathway. The established evaluation method provided detailed and comprehensive quantification of intraepithelial and stromal TILs throughout hematoxylin and eosin-stained slides. It has potential for clinical application for personalized treatment of patients with ovarian cancer.
Collapse
Affiliation(s)
- Kohei Hamada
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Akihiko Ueda
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoko Kashima
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Chiho Miyagawa
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Mana Taki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sachiko Minamiguchi
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
33
|
Darbandsari A, Farahani H, Asadi M, Wiens M, Cochrane D, Khajegili Mirabadi A, Jamieson A, Farnell D, Ahmadvand P, Douglas M, Leung S, Abolmaesumi P, Jones SJM, Talhouk A, Kommoss S, Gilks CB, Huntsman DG, Singh N, McAlpine JN, Bashashati A. AI-based histopathology image analysis reveals a distinct subset of endometrial cancers. Nat Commun 2024; 15:4973. [PMID: 38926357 PMCID: PMC11208496 DOI: 10.1038/s41467-024-49017-2] [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: 07/11/2023] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Endometrial cancer (EC) has four molecular subtypes with strong prognostic value and therapeutic implications. The most common subtype (NSMP; No Specific Molecular Profile) is assigned after exclusion of the defining features of the other three molecular subtypes and includes patients with heterogeneous clinical outcomes. In this study, we employ artificial intelligence (AI)-powered histopathology image analysis to differentiate between p53abn and NSMP EC subtypes and consequently identify a sub-group of NSMP EC patients that has markedly inferior progression-free and disease-specific survival (termed 'p53abn-like NSMP'), in a discovery cohort of 368 patients and two independent validation cohorts of 290 and 614 from other centers. Shallow whole genome sequencing reveals a higher burden of copy number abnormalities in the 'p53abn-like NSMP' group compared to NSMP, suggesting that this group is biologically distinct compared to other NSMP ECs. Our work demonstrates the power of AI to detect prognostically different and otherwise unrecognizable subsets of EC where conventional and standard molecular or pathologic criteria fall short, refining image-based tumor classification. This study's findings are applicable exclusively to females.
Collapse
Affiliation(s)
- Amirali Darbandsari
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Hossein Farahani
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Maryam Asadi
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Matthew Wiens
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Dawn Cochrane
- Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | | | - Amy Jamieson
- Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada
| | - David Farnell
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver General Hospital, Vancouver, BC, Canada
| | - Pouya Ahmadvand
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Maxwell Douglas
- Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Samuel Leung
- Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Purang Abolmaesumi
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Steven J M Jones
- Michael Smith Genome Sciences Center, British Columbia Cancer Research Center, Vancouver, BC, Canada
| | - Aline Talhouk
- Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada
| | - Stefan Kommoss
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver General Hospital, Vancouver, BC, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Naveena Singh
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver General Hospital, Vancouver, BC, Canada
| | - Jessica N McAlpine
- Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada
| | - Ali Bashashati
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
34
|
Morel D, Robert C, Paragios N, Grégoire V, Deutsch E. Translational Frontiers and Clinical Opportunities of Immunologically Fitted Radiotherapy. Clin Cancer Res 2024; 30:2317-2332. [PMID: 38477824 PMCID: PMC11145173 DOI: 10.1158/1078-0432.ccr-23-3632] [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: 11/21/2023] [Revised: 01/09/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024]
Abstract
Ionizing radiation can have a wide range of impacts on tumor-immune interactions, which are being studied with the greatest interest and at an accelerating pace by the medical community. Despite its undeniable immunostimulatory potential, it clearly appears that radiotherapy as it is prescribed and delivered nowadays often alters the host's immunity toward a suboptimal state. This may impair the full recovery of a sustained and efficient antitumor immunosurveillance posttreatment. An emerging concept is arising from this awareness and consists of reconsidering the way of designing radiation treatment planning, notably by taking into account the individualized risks of deleterious radio-induced immune alteration that can be deciphered from the planned beam trajectory through lymphocyte-rich organs. In this review, we critically appraise key aspects to consider while planning immunologically fitted radiotherapy, including the challenges linked to the identification of new dose constraints to immune-rich structures. We also discuss how pharmacologic immunomodulation could be advantageously used in combination with radiotherapy to compensate for the radio-induced loss, for example, with (i) agonists of interleukin (IL)2, IL4, IL7, IL9, IL15, or IL21, similarly to G-CSF being used for the prophylaxis of severe chemo-induced neutropenia, or with (ii) myeloid-derived suppressive cell blockers.
Collapse
Affiliation(s)
- Daphné Morel
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- INSERM U1030, Molecular Radiotherapy, Villejuif, France
| | - Charlotte Robert
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- INSERM U1030, Molecular Radiotherapy, Villejuif, France
- Paris-Saclay University, School of Medicine, Le Kremlin Bicêtre, France
| | - Nikos Paragios
- Therapanacea, Paris, France
- CentraleSupélec, Gif-sur-Yvette, France
| | - Vincent Grégoire
- Department of Radiation Oncology, Centre Léon Bérard, Lyon, France
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- INSERM U1030, Molecular Radiotherapy, Villejuif, France
- Paris-Saclay University, School of Medicine, Le Kremlin Bicêtre, France
| |
Collapse
|
35
|
Perez-Lopez R, Ghaffari Laleh N, Mahmood F, Kather JN. A guide to artificial intelligence for cancer researchers. Nat Rev Cancer 2024; 24:427-441. [PMID: 38755439 DOI: 10.1038/s41568-024-00694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 05/18/2024]
Abstract
Artificial intelligence (AI) has been commoditized. It has evolved from a specialty resource to a readily accessible tool for cancer researchers. AI-based tools can boost research productivity in daily workflows, but can also extract hidden information from existing data, thereby enabling new scientific discoveries. Building a basic literacy in these tools is useful for every cancer researcher. Researchers with a traditional biological science focus can use AI-based tools through off-the-shelf software, whereas those who are more computationally inclined can develop their own AI-based software pipelines. In this article, we provide a practical guide for non-computational cancer researchers to understand how AI-based tools can benefit them. We convey general principles of AI for applications in image analysis, natural language processing and drug discovery. In addition, we give examples of how non-computational researchers can get started on the journey to productively use AI in their own work.
Collapse
Affiliation(s)
- Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Narmin Ghaffari Laleh
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Dresden, Germany.
- Medical Oncology, National Center for Tumour Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
36
|
Davies E, Chetwynd A, McDowell G, Rao A, Oni L. The current use of proteomics and metabolomics in glomerulonephritis: a systematic literature review. J Nephrol 2024; 37:1209-1225. [PMID: 38689160 PMCID: PMC11405440 DOI: 10.1007/s40620-024-01923-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 02/24/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Glomerulonephritis inherently leads to the development of chronic kidney disease. It is the second most common diagnosis in patients requiring renal replacement therapy in the United Kingdom. Metabolomics and proteomics can characterise, identify and quantify an individual's protein and metabolite make-up. These techniques have been optimised and can be performed on samples including kidney tissue, blood and urine. Utilising omic techniques in nephrology can uncover disease pathophysiology and transform the diagnostics and treatment options for glomerulonephritis. OBJECTIVES To evaluate the utility of metabolomics and proteomics using mass spectrometry and nuclear magnetic resonance in glomerulonephritis. METHODS The systematic review was registered on PROSPERO (CRD42023442092). Standard and extensive Cochrane search methods were used. The latest search date was March 2023. Participants were of any age with a histological diagnosis of glomerulonephritis. Descriptive analysis was performed, and data presented in tabular form. An area under the curve or p-value was presented for potential biomarkers discovered. RESULTS Twenty-seven studies were included (metabolomics (n = 9)), and (proteomics (n = 18)) with 1818 participants. The samples analysed were urine (n = 19) blood (n = 4) and biopsy (n = 6). The typical outcome themes were potential biomarkers, disease phenotype, risk of progression and treatment response. CONCLUSION This review shows the potential of metabolomic and proteomic analysis to discover new disease biomarkers that may influence diagnostics and disease management. Further larger-scale research is required to establish the validity of the study outcomes, including the several proposed biomarkers.
Collapse
Affiliation(s)
- Elin Davies
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.
- Department of Nephrology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| | - Andrew Chetwynd
- Centre for Proteome Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Garry McDowell
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, UK
- Clinical Directorate, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
- Research Laboratory, Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Anirudh Rao
- Department of Nephrology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, UK
- Clinical Directorate, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Louise Oni
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Paediatric Nephrology, Alder Hey Children's, NHS Foundation Trust Hospital, Eaton Road, Liverpool, UK
| |
Collapse
|
37
|
Xu H, Usuyama N, Bagga J, Zhang S, Rao R, Naumann T, Wong C, Gero Z, González J, Gu Y, Xu Y, Wei M, Wang W, Ma S, Wei F, Yang J, Li C, Gao J, Rosemon J, Bower T, Lee S, Weerasinghe R, Wright BJ, Robicsek A, Piening B, Bifulco C, Wang S, Poon H. A whole-slide foundation model for digital pathology from real-world data. Nature 2024; 630:181-188. [PMID: 38778098 PMCID: PMC11153137 DOI: 10.1038/s41586-024-07441-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/19/2024] [Indexed: 05/25/2024]
Abstract
Digital pathology poses unique computational challenges, as a standard gigapixel slide may comprise tens of thousands of image tiles1-3. Prior models have often resorted to subsampling a small portion of tiles for each slide, thus missing the important slide-level context4. Here we present Prov-GigaPath, a whole-slide pathology foundation model pretrained on 1.3 billion 256 × 256 pathology image tiles in 171,189 whole slides from Providence, a large US health network comprising 28 cancer centres. The slides originated from more than 30,000 patients covering 31 major tissue types. To pretrain Prov-GigaPath, we propose GigaPath, a novel vision transformer architecture for pretraining gigapixel pathology slides. To scale GigaPath for slide-level learning with tens of thousands of image tiles, GigaPath adapts the newly developed LongNet5 method to digital pathology. To evaluate Prov-GigaPath, we construct a digital pathology benchmark comprising 9 cancer subtyping tasks and 17 pathomics tasks, using both Providence and TCGA data6. With large-scale pretraining and ultra-large-context modelling, Prov-GigaPath attains state-of-the-art performance on 25 out of 26 tasks, with significant improvement over the second-best method on 18 tasks. We further demonstrate the potential of Prov-GigaPath on vision-language pretraining for pathology7,8 by incorporating the pathology reports. In sum, Prov-GigaPath is an open-weight foundation model that achieves state-of-the-art performance on various digital pathology tasks, demonstrating the importance of real-world data and whole-slide modelling.
Collapse
Affiliation(s)
- Hanwen Xu
- Microsoft Research, Redmond, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | - Yu Gu
- Microsoft Research, Redmond, WA, USA
| | - Yanbo Xu
- Microsoft Research, Redmond, WA, USA
| | - Mu Wei
- Microsoft Research, Redmond, WA, USA
| | | | | | - Furu Wei
- Microsoft Research, Redmond, WA, USA
| | | | | | | | | | | | - Soohee Lee
- Providence Research Network, Renton, WA, USA
| | | | | | | | - Brian Piening
- Providence Genomics, Portland, OR, USA
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Carlo Bifulco
- Providence Genomics, Portland, OR, USA.
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA.
| | - Sheng Wang
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA.
- Department of Surgery, University of Washington, Seattle, WA, USA.
| | | |
Collapse
|
38
|
Shao W, Shi H, Liu J, Zuo Y, Sun L, Xia T, Chen W, Wan P, Sheng J, Zhu Q, Zhang D. Multi-Instance Multi-Task Learning for Joint Clinical Outcome and Genomic Profile Predictions From the Histopathological Images. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:2266-2278. [PMID: 38319755 DOI: 10.1109/tmi.2024.3362852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
With the remarkable success of digital histopathology and the deep learning technology, many whole-slide pathological images (WSIs) based deep learning models are designed to help pathologists diagnose human cancers. Recently, rather than predicting categorical variables as in cancer diagnosis, several deep learning studies are also proposed to estimate the continuous variables such as the patients' survival or their transcriptional profile. However, most of the existing studies focus on conducting these predicting tasks separately, which overlooks the useful intrinsic correlation among them that can boost the prediction performance of each individual task. In addition, it is sill challenge to design the WSI-based deep learning models, since a WSI is with huge size but annotated with coarse label. In this study, we propose a general multi-instance multi-task learning framework (HistMIMT) for multi-purpose prediction from WSIs. Specifically, we firstly propose a novel multi-instance learning module (TMICS) considering both common and specific task information across different tasks to generate bag representation for each individual task. Then, a soft-mask based fusion module with channel attention (SFCA) is developed to leverage useful information from the related tasks to help improve the prediction performance on target task. We evaluate our method on three cancer cohorts derived from the Cancer Genome Atlas (TCGA). For each cohort, our multi-purpose prediction tasks range from cancer diagnosis, survival prediction and estimating the transcriptional profile of gene TP53. The experimental results demonstrated that HistMIMT can yield better outcome on all clinical prediction tasks than its competitors.
Collapse
|
39
|
Hilgers L, Ghaffari Laleh N, West NP, Westwood A, Hewitt KJ, Quirke P, Grabsch HI, Carrero ZI, Matthaei E, Loeffler CML, Brinker TJ, Yuan T, Brenner H, Brobeil A, Hoffmeister M, Kather JN. Automated curation of large-scale cancer histopathology image datasets using deep learning. Histopathology 2024; 84:1139-1153. [PMID: 38409878 DOI: 10.1111/his.15159] [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: 09/19/2023] [Revised: 12/29/2023] [Accepted: 02/09/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND Artificial intelligence (AI) has numerous applications in pathology, supporting diagnosis and prognostication in cancer. However, most AI models are trained on highly selected data, typically one tissue slide per patient. In reality, especially for large surgical resection specimens, dozens of slides can be available for each patient. Manually sorting and labelling whole-slide images (WSIs) is a very time-consuming process, hindering the direct application of AI on the collected tissue samples from large cohorts. In this study we addressed this issue by developing a deep-learning (DL)-based method for automatic curation of large pathology datasets with several slides per patient. METHODS We collected multiple large multicentric datasets of colorectal cancer histopathological slides from the United Kingdom (FOXTROT, N = 21,384 slides; CR07, N = 7985 slides) and Germany (DACHS, N = 3606 slides). These datasets contained multiple types of tissue slides, including bowel resection specimens, endoscopic biopsies, lymph node resections, immunohistochemistry-stained slides, and tissue microarrays. We developed, trained, and tested a deep convolutional neural network model to predict the type of slide from the slide overview (thumbnail) image. The primary statistical endpoint was the macro-averaged area under the receiver operating curve (AUROCs) for detection of the type of slide. RESULTS In the primary dataset (FOXTROT), with an AUROC of 0.995 [95% confidence interval [CI]: 0.994-0.996] the algorithm achieved a high classification performance and was able to accurately predict the type of slide from the thumbnail image alone. In the two external test cohorts (CR07, DACHS) AUROCs of 0.982 [95% CI: 0.979-0.985] and 0.875 [95% CI: 0.864-0.887] were observed, which indicates the generalizability of the trained model on unseen datasets. With a confidence threshold of 0.95, the model reached an accuracy of 94.6% (7331 classified cases) in CR07 and 85.1% (2752 classified cases) for the DACHS cohort. CONCLUSION Our findings show that using the low-resolution thumbnail image is sufficient to accurately classify the type of slide in digital pathology. This can support researchers to make the vast resource of existing pathology archives accessible to modern AI models with only minimal manual annotations.
Collapse
Affiliation(s)
- Lars Hilgers
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Narmin Ghaffari Laleh
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Nicholas P West
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Alice Westwood
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Katherine J Hewitt
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Philip Quirke
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Heike I Grabsch
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Emylou Matthaei
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tanwei Yuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Brobeil
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Tissue Bank, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, 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
| |
Collapse
|
40
|
Herdiantoputri RR, Komura D, Ochi M, Fukawa Y, Kayamori K, Tsuchiya M, Kikuchi Y, Ushiku T, Ikeda T, Ishikawa S. Benchmarking Deep Learning-Based Image Retrieval of Oral Tumor Histology. Cureus 2024; 16:e62264. [PMID: 39011227 PMCID: PMC11247249 DOI: 10.7759/cureus.62264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2024] [Indexed: 07/17/2024] Open
Abstract
INTRODUCTION Oral tumors necessitate a dependable computer-assisted pathological diagnosis system considering their rarity and diversity. A content-based image retrieval (CBIR) system using deep neural networks has been successfully devised for digital pathology. No CBIR system for oral pathology has been investigated because of the lack of an extensive image database and feature extractors tailored to oral pathology. MATERIALS AND METHODS This study uses a large CBIR database constructed from 30 categories of oral tumors to compare deep learning methods as feature extractors. RESULTS The highest average area under the receiver operating characteristic curve (AUC) was achieved by models trained on database images using self-supervised learning (SSL) methods (0.900 with SimCLR and 0.897 with TiCo). The generalizability of the models was validated using query images from the same cases taken with smartphones. When smartphone images were tested as queries, both models yielded the highest mean AUC (0.871 with SimCLR and 0.857 with TiCo). We ensured the retrieved image result would be easily observed by evaluating the top 10 mean accuracies and checking for an exact diagnostic category and its differential diagnostic categories. CONCLUSION Training deep learning models with SSL methods using image data specific to the target site is beneficial for CBIR tasks in oral tumor histology to obtain histologically meaningful results and high performance. This result provides insight into the effective development of a CBIR system to help improve the accuracy and speed of histopathology diagnosis and advance oral tumor research in the future.
Collapse
Affiliation(s)
| | - Daisuke Komura
- Department of Preventive Medicine, The University of Tokyo, Tokyo, JPN
| | - Mieko Ochi
- Department of Preventive Medicine, The University of Tokyo, Tokyo, JPN
| | - Yuki Fukawa
- Department of Oral Pathology, Tokyo Medical and Dental University, Tokyo, JPN
| | - Kou Kayamori
- Department of Oral Pathology, Tokyo Medical and Dental University, Tokyo, JPN
| | - Maiko Tsuchiya
- Department of Pathology, Teikyo University School of Medicine, Tokyo, JPN
| | - Yoshinao Kikuchi
- Department of Pathology, Teikyo University School of Medicine, Tokyo, JPN
| | - Tetsuo Ushiku
- Department of Pathology, The University of Tokyo, Tokyo, JPN
| | - Tohru Ikeda
- Department of Oral Pathology, Tokyo Medical and Dental University, Tokyo, JPN
| | - Shumpei Ishikawa
- Department of Preventive Medicine, The University of Tokyo, Tokyo, JPN
| |
Collapse
|
41
|
Wang P, Wei X, Qu X, Zhu Y. Potential clinical application of microRNAs in bladder cancer. J Biomed Res 2024; 38:289-306. [PMID: 38808545 PMCID: PMC11300522 DOI: 10.7555/jbr.37.20230245] [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: 10/08/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 05/30/2024] Open
Abstract
Bladder cancer (BC) is the tenth most prevalent malignancy globally, presenting significant clinical and societal challenges because of its high incidence, rapid progression, and frequent recurrence. Presently, cystoscopy and urine cytology serve as the established diagnostic methods for BC. However, their efficacy is limited by their invasive nature and low sensitivity. Therefore, the development of highly specific biomarkers and effective non-invasive detection strategies is imperative for achieving a precise and timely diagnosis of BC, as well as for facilitating an optimal tumor treatment and an improved prognosis. microRNAs (miRNAs), short noncoding RNA molecules spanning around 20-25 nucleotides, are implicated in the regulation of diverse carcinogenic pathways. Substantially altered miRNAs form robust functional regulatory networks that exert a notable influence on the tumorigenesis and progression of BC. Investigations into aberrant miRNAs derived from blood, urine, or extracellular vesicles indicate their potential roles as diagnostic biomarkers and prognostic indicators in BC, enabling miRNAs to monitor the progression and predict the recurrence of the disease. Simultaneously, the investigation centered on miRNA as a potential therapeutic agent presents a novel approach for the treatment of BC. This review comprehensively analyzes biological roles of miRNAs in tumorigenesis and progression, and systematically summarizes their potential as diagnostic and prognostic biomarkers, as well as therapeutic targets for BC. Additionally, we evaluate the progress made in laboratory techniques within this field and discuss the prospects.
Collapse
Affiliation(s)
- Pei Wang
- Laboratory Medicine Center, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Xiaowei Wei
- Laboratory Medicine Center, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Xiaojun Qu
- Laboratory Medicine Center, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Yefei Zhu
- Laboratory Medicine Center, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, China
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|
42
|
Song AH, Williams M, Williamson DFK, Chow SSL, Jaume G, Gao G, Zhang A, Chen B, Baras AS, Serafin R, Colling R, Downes MR, Farré X, Humphrey P, Verrill C, True LD, Parwani AV, Liu JTC, Mahmood F. Analysis of 3D pathology samples using weakly supervised AI. Cell 2024; 187:2502-2520.e17. [PMID: 38729110 PMCID: PMC11168832 DOI: 10.1016/j.cell.2024.03.035] [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: 08/01/2023] [Revised: 01/15/2024] [Accepted: 03/25/2024] [Indexed: 05/12/2024]
Abstract
Human tissue, which is inherently three-dimensional (3D), is traditionally examined through standard-of-care histopathology as limited two-dimensional (2D) cross-sections that can insufficiently represent the tissue due to sampling bias. To holistically characterize histomorphology, 3D imaging modalities have been developed, but clinical translation is hampered by complex manual evaluation and lack of computational platforms to distill clinical insights from large, high-resolution datasets. We present TriPath, a deep-learning platform for processing tissue volumes and efficiently predicting clinical outcomes based on 3D morphological features. Recurrence risk-stratification models were trained on prostate cancer specimens imaged with open-top light-sheet microscopy or microcomputed tomography. By comprehensively capturing 3D morphologies, 3D volume-based prognostication achieves superior performance to traditional 2D slice-based approaches, including clinical/histopathological baselines from six certified genitourinary pathologists. Incorporating greater tissue volume improves prognostic performance and mitigates risk prediction variability from sampling bias, further emphasizing the value of capturing larger extents of heterogeneous morphology.
Collapse
Affiliation(s)
- Andrew H Song
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mane Williams
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Drew F K Williamson
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sarah S L Chow
- Department of Mechanical Engineering, Bioengineering, and Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Guillaume Jaume
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gan Gao
- Department of Mechanical Engineering, Bioengineering, and Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Andrew Zhang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bowen Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexander S Baras
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Serafin
- Department of Mechanical Engineering, Bioengineering, and Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Richard Colling
- Nuffield Department of Surgical Sciences, University of Oxford, UK; Department of Cellular Pathology, Oxford University Hospitals NHS Foundations Trust, John Radcliffe Hospital, Oxford, UK
| | - Michelle R Downes
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Xavier Farré
- Public Health Agency of Catalonia, Lleida, Spain
| | - Peter Humphrey
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Clare Verrill
- Nuffield Department of Surgical Sciences, University of Oxford, UK; Department of Cellular Pathology, Oxford University Hospitals NHS Foundations Trust, John Radcliffe Hospital, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Lawrence D True
- Department of Laboratory Medicine & Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Anil V Parwani
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Jonathan T C Liu
- Department of Mechanical Engineering, Bioengineering, and Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA.
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
43
|
Chakraborty D, Gutierrez‐Chakraborty E, Rodriguez‐Aguayo C, Başağaoğlu H, Lopez‐Berestein G, Amero P. Discovering genetic biomarkers for targeted cancer therapeutics with eXplainable Artificial Intelligence. Cancer Commun (Lond) 2024; 44:584-588. [PMID: 38566430 PMCID: PMC11110951 DOI: 10.1002/cac2.12530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/12/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Affiliation(s)
- Debaditya Chakraborty
- School of Civil and Environmental Engineering and Construction ManagementThe University of Texas at San AntonioSan AntonioTexasUSA
| | | | - Cristian Rodriguez‐Aguayo
- Department of Experimental TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | | | - Gabriel Lopez‐Berestein
- Department of Experimental TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Paola Amero
- Department of Experimental TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
44
|
Erdat EC, Yalciner M, Urun Y. Accuracy and usability of artificial intelligence chatbot generated chemotherapy protocols. Future Oncol 2024:1-6. [PMID: 38646965 DOI: 10.2217/fon-2023-0950] [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: 11/06/2023] [Accepted: 03/20/2024] [Indexed: 04/25/2024] Open
Abstract
Background: Medical practitioners are increasingly using artificial intelligence (AI) chatbots for easier and faster access to information. To our knowledge, the accuracy and availability of AI-generated chemotherapy protocols has not yet been studied. Methods: Nine simulated cancer patient cases were designed and AI chatbots, ChatGPT version 3.5 (OpenAI) and Bing (Microsoft), were used to generate chemotherapy protocols for each case. Results: Generated chemotherapy protocols were compared with the original protocols for nine simulated cancer patients. ChatGPT's overall performance was 5 out of 9 on protocol generation, and Bing's was 4 out of 9; this was statistically nonsignificant (p = 1). Conclusion: AI chatbots show both potential and limitations in generating chemotherapy protocols. The overall performance is low, and they should be used carefully in oncological practice.
Collapse
Affiliation(s)
- Efe Cem Erdat
- Ankara University Department of Medical Oncology, Ankara, Turkey
| | - Merih Yalciner
- Ankara University Department of Medical Oncology, Ankara, Turkey
| | - Yuksel Urun
- Ankara University Department of Medical Oncology, Ankara, Turkey
| |
Collapse
|
45
|
Zhu L, Pan J, Mou W, Deng L, Zhu Y, Wang Y, Pareek G, Hyams E, Carneiro BA, Hadfield MJ, El-Deiry WS, Yang T, Tan T, Tong T, Ta N, Zhu Y, Gao Y, Lai Y, Cheng L, Chen R, Xue W. Harnessing artificial intelligence for prostate cancer management. Cell Rep Med 2024; 5:101506. [PMID: 38593808 PMCID: PMC11031422 DOI: 10.1016/j.xcrm.2024.101506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/05/2024] [Accepted: 03/19/2024] [Indexed: 04/11/2024]
Abstract
Prostate cancer (PCa) is a common malignancy in males. The pathology review of PCa is crucial for clinical decision-making, but traditional pathology review is labor intensive and subjective to some extent. Digital pathology and whole-slide imaging enable the application of artificial intelligence (AI) in pathology. This review highlights the success of AI in detecting and grading PCa, predicting patient outcomes, and identifying molecular subtypes. We propose that AI-based methods could collaborate with pathologists to reduce workload and assist clinicians in formulating treatment recommendations. We also introduce the general process and challenges in developing AI pathology models for PCa. Importantly, we summarize publicly available datasets and open-source codes to facilitate the utilization of existing data and the comparison of the performance of different models to improve future studies.
Collapse
Affiliation(s)
- Lingxuan Zhu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Changping Laboratory, Beijing, China
| | - Jiahua Pan
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Weiming Mou
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Longxin Deng
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yinjie Zhu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yanqing Wang
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Gyan Pareek
- Department of Surgery (Urology), Brown University Warren Alpert Medical School, Providence, RI, USA; Minimally Invasive Urology Institute, Providence, RI, USA
| | - Elias Hyams
- Department of Surgery (Urology), Brown University Warren Alpert Medical School, Providence, RI, USA; Minimally Invasive Urology Institute, Providence, RI, USA
| | - Benedito A Carneiro
- The Legorreta Cancer Center at Brown University, Lifespan Cancer Institute, Providence, RI, USA
| | - Matthew J Hadfield
- The Legorreta Cancer Center at Brown University, Lifespan Cancer Institute, Providence, RI, USA
| | - Wafik S El-Deiry
- The Legorreta Cancer Center at Brown University, Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology & Laboratory Medicine, The Warren Alpert Medical School of Brown University, The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Division of Hematology/Oncology, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Tao Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Tan
- Faculty of Applied Sciences, Macao Polytechnic University, Address: R. de Luís Gonzaga Gomes, Macao, China
| | - Tong Tong
- College of Physics and Information Engineering, Fuzhou University, Fujian 350108, China
| | - Na Ta
- Department of Pathology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yan Zhu
- Department of Pathology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yisha Gao
- Department of Pathology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yancheng Lai
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Liang Cheng
- Department of Surgery (Urology), Brown University Warren Alpert Medical School, Providence, RI, USA; Department of Pathology and Laboratory Medicine, Department of Surgery (Urology), Brown University Warren Alpert Medical School, Lifespan Health, and the Legorreta Cancer Center at Brown University, Providence, RI, USA.
| | - Rui Chen
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Wei Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
46
|
Jin D, Liang S, Shmatko A, Arnold A, Horst D, Grünewald TGP, Gerstung M, Bai X. Teacher-student collaborated multiple instance learning for pan-cancer PDL1 expression prediction from histopathology slides. Nat Commun 2024; 15:3063. [PMID: 38594278 PMCID: PMC11004138 DOI: 10.1038/s41467-024-46764-0] [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: 07/30/2023] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Programmed cell death ligand 1 (PDL1), as an important biomarker, is quantified by immunohistochemistry (IHC) with few established histopathological patterns. Deep learning aids in histopathological assessment, yet heterogeneity and lacking spatially resolved annotations challenge precise analysis. Here, we present a weakly supervised learning approach using bulk RNA sequencing for PDL1 expression prediction from hematoxylin and eosin (H&E) slides. Our method extends the multiple instance learning paradigm with the teacher-student framework, which assigns dynamic pseudo-labels for intra-slide heterogeneity and retrieves unlabeled instances using temporal ensemble model distillation. The approach, evaluated on 12,299 slides across 20 solid tumor types, achieves a weighted average area under the curve of 0.83 on fresh-frozen and 0.74 on formalin-fixed specimens for 9 tumors with PDL1 as an established biomarker. Our method predicts PDL1 expression patterns, validated by IHC on 20 slides, offering insights into histologies relevant to PDL1. This demonstrates the potential of deep learning in identifying diverse histological patterns for molecular changes from H&E images.
Collapse
Affiliation(s)
- Darui Jin
- Image Processing Center, Beihang University, Beijing, 102206, China
- Division of AI in Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Shen Yuan Honors College, Beihang University, Beijing, 100191, China
| | - Shangying Liang
- Image Processing Center, Beihang University, Beijing, 102206, China
| | - Artem Shmatko
- Division of AI in Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Arnold
- Charité - Universitätsmedizin Berlin, Institute of Pathology, 10117, Berlin, Germany
| | - David Horst
- Charité - Universitätsmedizin Berlin, Institute of Pathology, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas G P Grünewald
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany.
- Hopp Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany.
| | - Moritz Gerstung
- Division of AI in Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Xiangzhi Bai
- Image Processing Center, Beihang University, Beijing, 102206, China.
- State Key Laboratory of Virtual Reality Technology and Systems, Beihang University, Beijing, 100191, China.
- Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100083, China.
| |
Collapse
|
47
|
Sali R, Jiang Y, Attaranzadeh A, Holmes B, Li R. Morphological diversity of cancer cells predicts prognosis across tumor types. J Natl Cancer Inst 2024; 116:555-564. [PMID: 37982756 PMCID: PMC10995848 DOI: 10.1093/jnci/djad243] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/23/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Intratumor heterogeneity drives disease progression and treatment resistance, which can lead to poor patient outcomes. Here, we present a computational approach for quantification of cancer cell diversity in routine hematoxylin-eosin-stained histopathology images. METHODS We analyzed publicly available digitized whole-slide hematoxylin-eosin images for 2000 patients. Four tumor types were included: lung, head and neck, colon, and rectal cancers, representing major histology subtypes (adenocarcinomas and squamous cell carcinomas). We performed single-cell analysis on hematoxylin-eosin images and trained a deep convolutional autoencoder to automatically learn feature representations of individual cancer nuclei. We then computed features of intranuclear variability and internuclear diversity to quantify tumor heterogeneity. Finally, we used these features to build a machine-learning model to predict patient prognosis. RESULTS A total of 68 million cancer cells were segmented and analyzed for nuclear image features. We discovered multiple morphological subtypes of cancer cells (range = 15-20) that co-exist within the same tumor, each with distinct phenotypic characteristics. Moreover, we showed that a higher morphological diversity is associated with chromosome instability and genomic aneuploidy. A machine-learning model based on morphological diversity demonstrated independent prognostic values across tumor types (hazard ratio range = 1.62-3.23, P < .035) in validation cohorts and further improved prognostication when combined with clinical risk factors. CONCLUSIONS Our study provides a practical approach for quantifying intratumor heterogeneity based on routine histopathology images. The cancer cell diversity score can be used to refine risk stratification and inform personalized treatment strategies.
Collapse
Affiliation(s)
- Rasoul Sali
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuming Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Armin Attaranzadeh
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Brittany Holmes
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
48
|
Goodwin RJA, Platz SJ, Reis-Filho JS, Barry ST. Accelerating Drug Development Using Spatial Multi-omics. Cancer Discov 2024; 14:620-624. [PMID: 38571424 DOI: 10.1158/2159-8290.cd-24-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
SUMMARY Spatial biology approaches enabled by innovations in imaging biomarker platforms and artificial intelligence-enabled data integration and analysis provide an assessment of patient and disease heterogeneity at ever-increasing resolution. The utility of spatial biology data in accelerating drug programs, however, requires balancing exploratory discovery investigations against scalable and clinically applicable spatial biomarker analysis.
Collapse
Affiliation(s)
- Richard J A Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Stefan J Platz
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Jorge S Reis-Filho
- Cancer Biomarker Development, Early Oncology, AstraZeneca, Gaithersburg, Maryland
| | - Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
49
|
Omar M, Xu Z, Rand SB, Alexanderani MK, Salles DC, Valencia I, Schaeffer EM, Robinson BD, Lotan TL, Loda M, Marchionni L. Semi-Supervised, Attention-Based Deep Learning for Predicting TMPRSS2:ERG Fusion Status in Prostate Cancer Using Whole Slide Images. Mol Cancer Res 2024; 22:347-359. [PMID: 38284821 PMCID: PMC10985477 DOI: 10.1158/1541-7786.mcr-23-0639] [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: 10/02/2023] [Revised: 12/26/2023] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
IMPLICATIONS Our study illuminates the potential of deep learning in effectively inferring key prostate cancer genetic alterations from the tissue morphology depicted in routinely available histology slides, offering a cost-effective method that could revolutionize diagnostic strategies in oncology.
Collapse
Affiliation(s)
- Mohamed Omar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Zhuoran Xu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sophie B. Rand
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Daniela C. Salles
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Itzel Valencia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | | | - Brian D. Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
50
|
Rigamonti A, Viatore M, Polidori R, Rahal D, Erreni M, Fumagalli MR, Zanini D, Doni A, Putignano AR, Bossi P, Voulaz E, Alloisio M, Rossi S, Zucali PA, Santoro A, Balzano V, Nisticò P, Feuerhake F, Mantovani A, Locati M, Marchesi F. Integrating AI-Powered Digital Pathology and Imaging Mass Cytometry Identifies Key Classifiers of Tumor Cells, Stroma, and Immune Cells in Non-Small Cell Lung Cancer. Cancer Res 2024; 84:1165-1177. [PMID: 38315789 PMCID: PMC10982643 DOI: 10.1158/0008-5472.can-23-1698] [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: 06/12/2023] [Revised: 11/13/2023] [Accepted: 02/01/2024] [Indexed: 02/07/2024]
Abstract
Artificial intelligence (AI)-powered approaches are becoming increasingly used as histopathologic tools to extract subvisual features and improve diagnostic workflows. On the other hand, hi-plex approaches are widely adopted to analyze the immune ecosystem in tumor specimens. Here, we aimed at combining AI-aided histopathology and imaging mass cytometry (IMC) to analyze the ecosystem of non-small cell lung cancer (NSCLC). An AI-based approach was used on hematoxylin and eosin (H&E) sections from 158 NSCLC specimens to accurately identify tumor cells, both adenocarcinoma and squamous carcinoma cells, and to generate a classifier of tumor cell spatial clustering. Consecutive tissue sections were stained with metal-labeled antibodies and processed through the IMC workflow, allowing quantitative detection of 24 markers related to tumor cells, tissue architecture, CD45+ myeloid and lymphoid cells, and immune activation. IMC identified 11 macrophage clusters that mainly localized in the stroma, except for S100A8+ cells, which infiltrated tumor nests. T cells were preferentially localized in peritumor areas or in tumor nests, the latter being associated with better prognosis, and they were more abundant in highly clustered tumors. Integrated tumor and immune classifiers were validated as prognostic on whole slides. In conclusion, integration of AI-powered H&E and multiparametric IMC allows investigation of spatial patterns and reveals tissue relevant features with clinical relevance. SIGNIFICANCE Leveraging artificial intelligence-powered H&E analysis integrated with hi-plex imaging mass cytometry provides insights into the tumor ecosystem and can translate tumor features into classifiers to predict prognosis, genotype, and therapy response.
Collapse
Affiliation(s)
- Alessandra Rigamonti
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan; Milan, Italy
| | - Marika Viatore
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan; Milan, Italy
| | - Rebecca Polidori
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan; Milan, Italy
| | - Daoud Rahal
- Department of Pathology, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
| | - Marco Erreni
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Maria Rita Fumagalli
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Damiano Zanini
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Andrea Doni
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Anna Rita Putignano
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
| | - Paola Bossi
- Department of Pathology, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
| | - Emanuele Voulaz
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Thoracic Surgery, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Marco Alloisio
- Division of Thoracic Surgery, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Sabrina Rossi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Paolo Andrea Zucali
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Armando Santoro
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Vittoria Balzano
- Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Nisticò
- Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Alberto Mantovani
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Massimo Locati
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan; Milan, Italy
| | - Federica Marchesi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan; Milan, Italy
| |
Collapse
|