151
|
Pathology of Combined Hepatocellular Carcinoma-Cholangiocarcinoma: An Update. Cancers (Basel) 2023; 15:cancers15020494. [PMID: 36672443 PMCID: PMC9856551 DOI: 10.3390/cancers15020494] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Combined hepatocellular carcinoma-cholangiocarcinoma (cHCC-CCA) is a rare primary liver cancer that is composed of both hepatocellular and cholangiocellular differentiated cells. It is slightly more common in men and among Asian and Pacific islanders. Overall, risk factors are similar to classic risk factors of hepatocellular carcinoma (HCC). The classification has significantly evolved over time. The last WHO classification (2019) mainly emphasized diagnosis on morphological basis with routine stainings, discarded previously recognized classifications with carcinomas with stem cell features, introduced intermediate cell carcinoma as a specific subtype and considered cholangiolocarcinoma as a subtype of cholangiocellular carcinoma. Immunohistochemical markers may be applied for further specification but have limited value for diagnosis. Recent discoveries in molecular pathway regulation may pioneer new therapeutic approaches for this poor prognostic and challenging diagnosis.
Collapse
|
152
|
Ogier du Terrail J, Leopold A, Joly C, Béguier C, Andreux M, Maussion C, Schmauch B, Tramel EW, Bendjebbar E, Zaslavskiy M, Wainrib G, Milder M, Gervasoni J, Guerin J, Durand T, Livartowski A, Moutet K, Gautier C, Djafar I, Moisson AL, Marini C, Galtier M, Balazard F, Dubois R, Moreira J, Simon A, Drubay D, Lacroix-Triki M, Franchet C, Bataillon G, Heudel PE. Federated learning for predicting histological response to neoadjuvant chemotherapy in triple-negative breast cancer. Nat Med 2023; 29:135-146. [PMID: 36658418 DOI: 10.1038/s41591-022-02155-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 11/23/2022] [Indexed: 01/21/2023]
Abstract
Triple-negative breast cancer (TNBC) is a rare cancer, characterized by high metastatic potential and poor prognosis, and has limited treatment options. The current standard of care in nonmetastatic settings is neoadjuvant chemotherapy (NACT), but treatment efficacy varies substantially across patients. This heterogeneity is still poorly understood, partly due to the paucity of curated TNBC data. Here we investigate the use of machine learning (ML) leveraging whole-slide images and clinical information to predict, at diagnosis, the histological response to NACT for early TNBC women patients. To overcome the biases of small-scale studies while respecting data privacy, we conducted a multicentric TNBC study using federated learning, in which patient data remain secured behind hospitals' firewalls. We show that local ML models relying on whole-slide images can predict response to NACT but that collaborative training of ML models further improves performance, on par with the best current approaches in which ML models are trained using time-consuming expert annotations. Our ML model is interpretable and is sensitive to specific histological patterns. This proof of concept study, in which federated learning is applied to real-world datasets, paves the way for future biomarker discovery using unprecedentedly large datasets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Camille Franchet
- Institut Universitaire du Cancer de Toulouse (IUCT) Oncopole, Toulouse, France
| | | | | |
Collapse
|
153
|
Role of AI and digital pathology for colorectal immuno-oncology. Br J Cancer 2023; 128:3-11. [PMID: 36183010 DOI: 10.1038/s41416-022-01986-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 01/27/2023] Open
Abstract
Immunotherapy deals with therapeutic interventions to arrest the progression of tumours using the immune system. These include checkpoint inhibitors, T-cell manipulation, cytokines, oncolytic viruses and tumour vaccines. In this paper, we present a survey of the latest developments on immunotherapy in colorectal cancer (CRC) and the role of artificial intelligence (AI) in this context. Among these, microsatellite instability (MSI) is perhaps the most popular IO biomarker globally. We first discuss the MSI status of tumours, its implications for patient management, and its relationship to immune response. In recent years, several aspiring studies have used AI to predict the MSI status of patients from digital whole-slide images (WSIs) of routine diagnostic slides. We present a survey of AI literature on the prediction of MSI and tumour mutation burden from digitised WSIs of haematoxylin and eosin-stained diagnostic slides. We discuss AI approaches in detail and elaborate their contributions, limitations and key takeaways to drive future research. We further expand this survey to other IO-related biomarkers like immune cell infiltrates and alternate data modalities like immunohistochemistry and gene expression. Finally, we underline possible future directions in immunotherapy for CRC and promise of AI to accelerate this exploration for patient benefits.
Collapse
|
154
|
Yavuz A, Alpsoy A, Gedik EO, Celik MY, Bassorgun CI, Unal B, Elpek GO. Artificial intelligence applications in predicting the behavior of gastrointestinal cancers in pathology. Artif Intell Gastroenterol 2022; 3:142-162. [DOI: 10.35712/aig.v3.i5.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/25/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
Recent research has provided a wealth of data supporting the application of artificial intelligence (AI)-based applications in routine pathology practice. Indeed, it is clear that these methods can significantly support an accurate and rapid diagnosis by eliminating errors, increasing reliability, and improving workflow. In addition, the effectiveness of AI in the pathological evaluation of prognostic parameters associated with behavior, course, and treatment in many types of tumors has also been noted. Regarding gastrointestinal system (GIS) cancers, the contribution of AI methods to pathological diagnosis has been investigated in many studies. On the other hand, studies focusing on AI applications in evaluating parameters to determine tumor behavior are relatively few. For this purpose, the potential of AI models has been studied over a broad spectrum, from tumor subtyping to the identification of new digital biomarkers. The capacity of AI to infer genetic alterations of cancer tissues from digital slides has been demonstrated. Although current data suggest the merit of AI-based approaches in assessing tumor behavior in GIS cancers, a wide range of challenges still need to be solved, from laboratory infrastructure to improving the robustness of algorithms, before incorporating AI applications into real-life GIS pathology practice. This review aims to present data from AI applications in evaluating pathological parameters related to the behavior of GIS cancer with an overview of the opportunities and challenges encountered in implementing AI in pathology.
Collapse
Affiliation(s)
- Aysen Yavuz
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Anil Alpsoy
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Elif Ocak Gedik
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | | | | | - Betul Unal
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Gulsum Ozlem Elpek
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| |
Collapse
|
155
|
Lazard T, Bataillon G, Naylor P, Popova T, Bidard FC, Stoppa-Lyonnet D, Stern MH, Decencière E, Walter T, Vincent-Salomon A. Deep learning identifies morphological patterns of homologous recombination deficiency in luminal breast cancers from whole slide images. Cell Rep Med 2022; 3:100872. [PMID: 36516847 PMCID: PMC9798078 DOI: 10.1016/j.xcrm.2022.100872] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/04/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022]
Abstract
Homologous recombination DNA-repair deficiency (HRD) is becoming a well-recognized marker of platinum salt and polyADP-ribose polymerase inhibitor chemotherapies in ovarian and breast cancers. While large-scale screening for HRD using genomic markers is logistically and economically challenging, stained tissue slides are routinely acquired in clinical practice. With the objectives of providing a robust deep-learning method for HRD prediction from tissue slides and identifying related morphological phenotypes, we first show that digital pathology workflows are sensitive to potential biases in the training set, then we propose a method to overcome the influence of these biases, and we develop an interpretation method capable of identifying complex phenotypes. Application to our carefully curated in-house dataset allows us to predict HRD with high accuracy (area under the receiver-operator characteristics curve 0.86) and to identify morphological phenotypes related to HRD. In particular, the presence of laminated fibrosis and clear tumor cells associated with HRD open new hypotheses regarding its phenotypic impact.
Collapse
Affiliation(s)
- Tristan Lazard
- Center for Computational Biology (CBIO), Mines Paris, PSL University, 60 Boulevard Saint Michel, 75006 Paris, France,Institut Curie, PSL University, 75005 Paris, France,INSERM U900, 75005 Paris, France
| | - Guillaume Bataillon
- Institut Curie, PSL University, 75005 Paris, France,INSERM U900, 75005 Paris, France,Diagnostic and Theranostic Medicine Division, Institut Curie, PSL University, Paris, France
| | - Peter Naylor
- Center for Computational Biology (CBIO), Mines Paris, PSL University, 60 Boulevard Saint Michel, 75006 Paris, France,Institut Curie, PSL University, 75005 Paris, France,INSERM U900, 75005 Paris, France
| | - Tatiana Popova
- INSERM U830, DNA Repair and Uveal Melanoma (DRUM), Equipe Labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, 75005 Paris, France
| | - François-Clément Bidard
- Department of Medical Oncology, Institut Curie, Université de Versailles Saint-Quentin, Saint-Cloud, France,INSERM CIC-BT 1428, Institut Curie, Paris, France
| | - Dominique Stoppa-Lyonnet
- INSERM U830, DNA Repair and Uveal Melanoma (DRUM), Equipe Labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, 75005 Paris, France,Université Paris Cité, 75006 Paris, France
| | - Marc-Henri Stern
- Diagnostic and Theranostic Medicine Division, Institut Curie, PSL University, Paris, France,INSERM U830, DNA Repair and Uveal Melanoma (DRUM), Equipe Labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, 75005 Paris, France
| | - Etienne Decencière
- Center for Mathematical Morphology (CMM), Mines Paris, PSL University, 77300 Fontainebleau, France
| | - Thomas Walter
- Center for Computational Biology (CBIO), Mines Paris, PSL University, 60 Boulevard Saint Michel, 75006 Paris, France,Institut Curie, PSL University, 75005 Paris, France,INSERM U900, 75005 Paris, France,Corresponding author
| | - Anne Vincent-Salomon
- Diagnostic and Theranostic Medicine Division, Institut Curie, PSL University, Paris, France,INSERM U934, CNRS UMR 3215, Paris, France,Corresponding author
| |
Collapse
|
156
|
Couture HD. Deep Learning-Based Prediction of Molecular Tumor Biomarkers from H&E: A Practical Review. J Pers Med 2022; 12:2022. [PMID: 36556243 PMCID: PMC9784641 DOI: 10.3390/jpm12122022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/26/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Molecular and genomic properties are critical in selecting cancer treatments to target individual tumors, particularly for immunotherapy. However, the methods to assess such properties are expensive, time-consuming, and often not routinely performed. Applying machine learning to H&E images can provide a more cost-effective screening method. Dozens of studies over the last few years have demonstrated that a variety of molecular biomarkers can be predicted from H&E alone using the advancements of deep learning: molecular alterations, genomic subtypes, protein biomarkers, and even the presence of viruses. This article reviews the diverse applications across cancer types and the methodology to train and validate these models on whole slide images. From bottom-up to pathologist-driven to hybrid approaches, the leading trends include a variety of weakly supervised deep learning-based approaches, as well as mechanisms for training strongly supervised models in select situations. While results of these algorithms look promising, some challenges still persist, including small training sets, rigorous validation, and model explainability. Biomarker prediction models may yield a screening method to determine when to run molecular tests or an alternative when molecular tests are not possible. They also create new opportunities in quantifying intratumoral heterogeneity and predicting patient outcomes.
Collapse
|
157
|
Iqbal MS, Ahmad W, Alizadehsani R, Hussain S, Rehman R. Breast Cancer Dataset, Classification and Detection Using Deep Learning. Healthcare (Basel) 2022; 10:2395. [PMID: 36553919 PMCID: PMC9778593 DOI: 10.3390/healthcare10122395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Incorporating scientific research into clinical practice via clinical informatics, which includes genomics, proteomics, bioinformatics, and biostatistics, improves patients' treatment. Computational pathology is a growing subspecialty with the potential to integrate whole slide images, multi-omics data, and health informatics. Pathology and laboratory medicine are critical to diagnosing cancer. This work will review existing computational and digital pathology methods for breast cancer diagnosis with a special focus on deep learning. The paper starts by reviewing public datasets related to breast cancer diagnosis. Additionally, existing deep learning methods for breast cancer diagnosis are reviewed. The publicly available code repositories are introduced as well. The paper is closed by highlighting challenges and future works for deep learning-based diagnosis.
Collapse
Affiliation(s)
- Muhammad Shahid Iqbal
- Department of Computer Science and Information Technology, Women University AJK, Bagh 12500, Pakistan
| | - Waqas Ahmad
- Higher Education Department Govt, AJK, Mirpur 10250, Pakistan
| | - Roohallah Alizadehsani
- Institute for Intelligent Systems Research and Innovation (IISRI), Deakin University, Geelong, VIC 3216, Australia
| | - Sadiq Hussain
- Examination Branch, Dibrugarh University, Dibrugarh 786004, India
| | - Rizwan Rehman
- Centre for Computer Science and Applications, Dibrugarh University, Dibrugarh 786004, India
| |
Collapse
|
158
|
Deep learning model to predict Epstein-Barr virus associated gastric cancer in histology. Sci Rep 2022; 12:18466. [PMID: 36323712 PMCID: PMC9630260 DOI: 10.1038/s41598-022-22731-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/18/2022] [Indexed: 11/20/2022] Open
Abstract
The detection of Epstein-Barr virus (EBV) in gastric cancer patients is crucial for clinical decision making, as it is related with specific treatment responses and prognoses. Despite its importance, the limited medical resources preclude universal EBV testing. Herein, we propose a deep learning-based EBV prediction method from H&E-stained whole-slide images (WSI). Our model was developed using 319 H&E stained WSI (26 EBV positive; TCGA dataset) from the Cancer Genome Atlas, and 108 WSI (8 EBV positive; ISH dataset) from an independent institution. Our deep learning model, EBVNet consists of two sequential components: a tumor classifier and an EBV classifier. We visualized the learned representation by the classifiers using UMAP. We externally validated the model using 60 additional WSI (7 being EBV positive; HGH dataset). We compared the model's performance with those of four pathologists. EBVNet achieved an AUPRC of 0.65, whereas the four pathologists yielded a mean AUPRC of 0.41. Moreover, EBVNet achieved an negative predictive value, sensitivity, specificity, precision, and F1-score of 0.98, 0.86, 0.92, 0.60, and 0.71, respectively. Our proposed model is expected to contribute to prescreen patients for confirmatory testing, potentially to save test-related cost and labor.
Collapse
|
159
|
Deng J, Yang J, Hou L, Wu J, He Y, Zhao M, Ni B, Wei D, Pfister H, Zhou C, Jiang T, She Y, Wu C, Chen C. Genopathomic profiling identifies signatures for immunotherapy response of lung adenocarcinoma via confounder-aware representation learning. iScience 2022; 25:105382. [PMCID: PMC9636035 DOI: 10.1016/j.isci.2022.105382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/18/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Jiajun Deng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Jiancheng Yang
- Shanghai Jiao Tong University, Shanghai, P.R. China
- MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, P.R. China
- Dianei Technology, Shanghai, P.R. China
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Yi He
- Dianei Technology, Shanghai, P.R. China
| | - Mengmeng Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Bingbing Ni
- Shanghai Jiao Tong University, Shanghai, P.R. China
- MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, P.R. China
- Huawei Hisilicon, Shanghai, P.R. China
| | | | | | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
- Corresponding author
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
- Corresponding author
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
- The First Hospital of Lanzhou University, Gansu, P.R. China
- The International Science and Technology Cooperation Base for Development and Application of Key Technologies in Thoracic Surgery, Gansu, P.R. China
- Corresponding author
| |
Collapse
|
160
|
Kolmar L, Autour A, Ma X, Vergier B, Eduati F, Merten CA. Technological and computational advances driving high-throughput oncology. Trends Cell Biol 2022; 32:947-961. [PMID: 35577671 DOI: 10.1016/j.tcb.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 01/21/2023]
Abstract
Engineering and computational advances have opened many new avenues in cancer research, particularly when being exploited in interdisciplinary approaches. For example, the combination of microfluidics, novel sequencing technologies, and computational analyses has been crucial to enable single-cell assays, giving a detailed picture of tumor heterogeneity for the very first time. In a similar way, these 'tech' disciplines have been elementary for generating large data sets in multidimensional cancer 'omics' approaches, cell-cell interaction screens, 3D tumor models, and tissue level analyses. In this review we summarize the most important technology and computational developments that have been or will be instrumental for transitioning classical cancer research to a large data-driven, high-throughput, high-content discipline across all biological scales.
Collapse
Affiliation(s)
- Leonie Kolmar
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alexis Autour
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Xiaoli Ma
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Blandine Vergier
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Federica Eduati
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands.
| | - Christoph A Merten
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
161
|
An Analysis of Transcriptomic Burden Identifies Biological Progression Roadmaps for Hematological Malignancies and Solid Tumors. Biomedicines 2022; 10:biomedicines10112720. [DOI: 10.3390/biomedicines10112720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Biological paths of tumor progression are difficult to predict without time-series data. Using median shift and abacus transformation in the analysis of RNA sequencing data sets, natural patient stratifications were found based on their transcriptomic burden (TcB). Using gene-behavior analysis, TcB groups were evaluated further to discover biological courses of tumor progression. We found that solid tumors and hematological malignancies (n = 4179) share conserved biological patterns, and biological network complexity decreases at increasing TcB levels. An analysis of gene expression datasets including pediatric leukemia patients revealed TcB patterns with biological directionality and survival implications. A prospective interventional study with PI3K targeted therapy in canine lymphomas proved that directional biological responses are dynamic. To conclude, TcB-enriched biological mechanisms detected the existence of biological trajectories within tumors. Using this prognostic informative novel informatics method, which can be applied to tumor transcriptomes and progressive diseases inspires the design of progression-specific therapeutic approaches.
Collapse
|
162
|
Lipkova J, Chen RJ, Chen B, Lu MY, Barbieri M, Shao D, Vaidya AJ, Chen C, Zhuang L, Williamson DFK, Shaban M, Chen TY, Mahmood F. Artificial intelligence for multimodal data integration in oncology. Cancer Cell 2022; 40:1095-1110. [PMID: 36220072 PMCID: PMC10655164 DOI: 10.1016/j.ccell.2022.09.012] [Citation(s) in RCA: 226] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/12/2022] [Accepted: 09/15/2022] [Indexed: 02/07/2023]
Abstract
In oncology, the patient state is characterized by a whole spectrum of modalities, ranging from radiology, histology, and genomics to electronic health records. Current artificial intelligence (AI) models operate mainly in the realm of a single modality, neglecting the broader clinical context, which inevitably diminishes their potential. Integration of different data modalities provides opportunities to increase robustness and accuracy of diagnostic and prognostic models, bringing AI closer to clinical practice. AI models are also capable of discovering novel patterns within and across modalities suitable for explaining differences in patient outcomes or treatment resistance. The insights gleaned from such models can guide exploration studies and contribute to the discovery of novel biomarkers and therapeutic targets. To support these advances, here we present a synopsis of AI methods and strategies for multimodal data fusion and association discovery. We outline approaches for AI interpretability and directions for AI-driven exploration through multimodal data interconnections. We examine challenges in clinical adoption and discuss emerging solutions.
Collapse
Affiliation(s)
- Jana Lipkova
- 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
| | - 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; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, 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; Department of Computer Science, Harvard University, Cambridge, MA, USA
| | - 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; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Matteo Barbieri
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel Shao
- 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; Harvard-MIT Health Sciences and Technology (HST), Cambridge, MA, USA
| | - Anurag J Vaidya
- 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; Harvard-MIT Health Sciences and Technology (HST), Cambridge, MA, USA
| | - Chengkuan 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
| | - Luoting Zhuang
- 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
| | - 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
| | - Muhammad Shaban
- 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
| | - Tiffany Y 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
| | - 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; Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
163
|
Lee SH, Jang HJ. Deep learning-based prediction of molecular cancer biomarkers from tissue slides: A new tool for precision oncology. Clin Mol Hepatol 2022; 28:754-772. [PMID: 35443570 PMCID: PMC9597228 DOI: 10.3350/cmh.2021.0394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/17/2022] [Indexed: 02/06/2023] Open
Abstract
Molecular tests are necessary to stratify cancer patients for targeted therapy. However, high cost and technical barriers limit the application of these tests, hindering optimal treatment. Recently, deep learning (DL) has been applied to predict molecular test results from digitized images of tissue slides. Furthermore, treatment response and prognosis can be predicted from tissue slides using DL. In this review, we summarized DL-based studies regarding the prediction of genetic mutation, microsatellite instability, tumor mutational burden, molecular subtypes, gene expression, treatment response, and prognosis directly from hematoxylin- and eosin-stained tissue slides. Although performance needs to be improved, these studies clearly demonstrated the feasibility of DL-based prediction of key molecular features in cancer tissues. With the accumulation of data and technical advances, the performance of the DL system could be improved in the near future. Therefore, we expect that DL could provide cost- and time-effective alternative tools for patient stratification in the era of precision oncology.
Collapse
Affiliation(s)
- Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyun-Jong Jang
- Catholic Big Data Integration Center, Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, Korea,Corresponding author : Hyun-Jong Jang Department of Physiology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-7274, Fax: +82-2-532-9575, E-mail:
| |
Collapse
|
164
|
Qiao Y, Zhao L, Luo C, Luo Y, Wu Y, Li S, Bu D, Zhao Y. Multi-modality artificial intelligence in digital pathology. Brief Bioinform 2022; 23:6702380. [PMID: 36124675 PMCID: PMC9677480 DOI: 10.1093/bib/bbac367] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/27/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022] Open
Abstract
In common medical procedures, the time-consuming and expensive nature of obtaining test results plagues doctors and patients. Digital pathology research allows using computational technologies to manage data, presenting an opportunity to improve the efficiency of diagnosis and treatment. Artificial intelligence (AI) has a great advantage in the data analytics phase. Extensive research has shown that AI algorithms can produce more up-to-date and standardized conclusions for whole slide images. In conjunction with the development of high-throughput sequencing technologies, algorithms can integrate and analyze data from multiple modalities to explore the correspondence between morphological features and gene expression. This review investigates using the most popular image data, hematoxylin-eosin stained tissue slide images, to find a strategic solution for the imbalance of healthcare resources. The article focuses on the role that the development of deep learning technology has in assisting doctors' work and discusses the opportunities and challenges of AI.
Collapse
Affiliation(s)
- Yixuan Qiao
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lianhe Zhao
- Corresponding authors: Yi Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences; Shandong First Medical University & Shandong Academy of Medical Sciences. Tel.: +86 10 6260 0822; Fax: +86 10 6260 1356; E-mail: ; Lianhe Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences. Tel.: +86 18513983324; E-mail:
| | - Chunlong Luo
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yufan Luo
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Wu
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Shengtong Li
- Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dechao Bu
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Yi Zhao
- Corresponding authors: Yi Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences; Shandong First Medical University & Shandong Academy of Medical Sciences. Tel.: +86 10 6260 0822; Fax: +86 10 6260 1356; E-mail: ; Lianhe Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences. Tel.: +86 18513983324; E-mail:
| |
Collapse
|
165
|
Wang K, Ren Y, Ma L, Fan Y, Yang Z, Yang Q, Shi J, Sun Y. Deep Learning-Based Prediction of Treatment Prognosis from Nasal Polyp Histology Slides. Int Forum Allergy Rhinol 2022; 13:886-898. [PMID: 36066094 DOI: 10.1002/alr.23083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Histopathology of nasal polyps contains rich prognostic information, which is difficult to objectively extract. In the present study, we aimed to develop a prognostic indicator of patient outcomes by analyzing scanned conventional haematoxylin and eosin (H&E) -stained slides alone using deep learning. METHODS An interpretable supervised deep learning model was developed using 185 H&E-stained whole-slide images (WSIs) of nasal polyps, each from a patient randomly selected from the pool of 232 patients who underwent endoscopic sinus surgery at the First Affiliated Hospital of Sun Yat-sen University (internal cohort). We internally validated the model on a holdout dataset from the internal cohort (47 H&E-stained WSIs) and externally validated the model on 122 H&E-stained WSIs from the Seventh Affiliated Hospital of Sun Yat-sen University and the University of Hong Kong-Shenzhen Hospital (external cohort). A poor prognosis score (PPS) was established to evaluate patient outcomes, and then risk activation mapping was applied to visualize the histopathological features underlying PPS. RESULTS The model yielded a patient-level sensitivity of 79.5%, and specificity of 92.3%, with areas under the receiver operating characteristic curve of 0.943, on the multi-center external cohort. The predictive ability of PPS was superior to that of conventional tissue eosinophil number. Notably, eosinophil infiltration, goblet cell hyperplasia, glandular hyperplasia, squamous metaplasia, and fibrin deposition were identified as the main underlying features of PPS. CONCLUSIONS Our deep learning model is an effective method for decoding pathological images of nasal polyps, providing a valuable solution for disease prognosis prediction and precise patient treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kanghua Wang
- Department of Otolaryngology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.,Department of Otolaryngology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yong Ren
- Center for Digestive Disease, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.,Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Ling Ma
- Department of Otorhinolaryngology, the University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Yunping Fan
- Department of Otolaryngology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zheng Yang
- Department of Pathology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Qintai Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianbo Shi
- Department of Otolaryngology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yueqi Sun
- Department of Otolaryngology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.,Department of Otolaryngology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
166
|
Song JH, Hong Y, Kim ER, Kim SH, Sohn I. Utility of artificial intelligence with deep learning of hematoxylin and eosin-stained whole slide images to predict lymph node metastasis in T1 colorectal cancer using endoscopically resected specimens; prediction of lymph node metastasis in T1 colorectal cancer. J Gastroenterol 2022; 57:654-666. [PMID: 35802259 DOI: 10.1007/s00535-022-01894-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/09/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND When endoscopically resected specimens of early colorectal cancer (CRC) show high-risk features, surgery should be performed based on current guidelines because of the high-risk of lymph node metastasis (LNM). The aim of this study was to determine the utility of an artificial intelligence (AI) with deep learning (DL) of hematoxylin and eosin (H&E)-stained endoscopic resection specimens without manual-pixel-level annotation for predicting LNM in T1 CRC. In addition, we assessed AI performance for patients with only submucosal (SM) invasion depth of 1000 to 2000 μm known to be difficult to predict LNM in clinical practice. METHODS H&E-stained whole slide images (WSIs) were scanned for endoscopic resection specimens of 400 patients who underwent endoscopic treatment for newly diagnosed T1 CRC with additional surgery. The area under the curve (AUC) of the receiver operating characteristic curve was used to determine the accuracy of AI for predicting LNM with a fivefold cross-validation in the training set and in a held-out test set. RESULTS We developed an AI model using a two-step attention-based DL approach without clinical features (AUC, 0.764). Incorporating clinical features into the model did not improve its prediction accuracy for LNM. Our model reduced unnecessary additional surgery by 15.1% more than using the current guidelines (67.4% vs. 82.5%). In patients with SM invasion depth of 1000 to 2000 μm, the AI avoided 16.1% of unnecessary additional surgery than using the JSCCR guidelines. CONCLUSIONS Our study is the first to show that AI trained with DL of H&E-stained WSIs has the potential to predict LNM in T1 CRC using only endoscopically resected specimens with conventional histologic risk factors.
Collapse
Affiliation(s)
- Joo Hye Song
- Department of Medicine, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Yiyu Hong
- Department of R&D Center, Arontier Co., Ltd, Seoul, Republic of Korea
| | - Eun Ran Kim
- Department of Medicine, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| | - Seok-Hyung Kim
- Department of Pathology, Samsung Medical Center, Seoul, Republic of Korea
| | - Insuk Sohn
- Department of R&D Center, Arontier Co., Ltd, Seoul, Republic of Korea
| |
Collapse
|
167
|
Shmatko A, Ghaffari Laleh N, Gerstung M, Kather JN. Artificial intelligence in histopathology: enhancing cancer research and clinical oncology. NATURE CANCER 2022; 3:1026-1038. [PMID: 36138135 DOI: 10.1038/s43018-022-00436-4] [Citation(s) in RCA: 185] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 08/03/2022] [Indexed: 06/16/2023]
Abstract
Artificial intelligence (AI) methods have multiplied our capabilities to extract quantitative information from digital histopathology images. AI is expected to reduce workload for human experts, improve the objectivity and consistency of pathology reports, and have a clinical impact by extracting hidden information from routinely available data. Here, we describe how AI can be used to predict cancer outcome, treatment response, genetic alterations and gene expression from digitized histopathology slides. We summarize the underlying technologies and emerging approaches, noting limitations, including the need for data sharing and standards. Finally, we discuss the broader implications of AI in cancer research and oncology.
Collapse
Affiliation(s)
- Artem Shmatko
- Division of AI in Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
| | | | - Moritz Gerstung
- Division of AI in Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK.
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.
- Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
168
|
Fremond S, Koelzer VH, Horeweg N, Bosse T. The evolving role of morphology in endometrial cancer diagnostics: From histopathology and molecular testing towards integrative data analysis by deep learning. Front Oncol 2022; 12:928977. [PMID: 36059702 PMCID: PMC9433878 DOI: 10.3389/fonc.2022.928977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Endometrial cancer (EC) diagnostics is evolving into a system in which molecular aspects are increasingly important. The traditional histological subtype-driven classification has shifted to a molecular-based classification that stratifies EC into DNA polymerase epsilon mutated (POLEmut), mismatch repair deficient (MMRd), and p53 abnormal (p53abn), and the remaining EC as no specific molecular profile (NSMP). The molecular EC classification has been implemented in the World Health Organization 2020 classification and the 2021 European treatment guidelines, as it serves as a better basis for patient management. As a result, the integration of the molecular class with histopathological variables has become a critical focus of recent EC research. Pathologists have observed and described several morphological characteristics in association with specific genomic alterations, but these appear insufficient to accurately classify patients according to molecular subgroups. This requires pathologists to rely on molecular ancillary tests in routine workup. In this new era, it has become increasingly challenging to assign clinically relevant weights to histological and molecular features on an individual patient basis. Deep learning (DL) technology opens new options for the integrative analysis of multi-modal image and molecular datasets with clinical outcomes. Proof-of-concept studies in other cancers showed promising accuracy in predicting molecular alterations from H&E-stained tumor slide images. This suggests that some morphological characteristics that are associated with molecular alterations could be identified in EC, too, expanding the current understanding of the molecular-driven EC classification. Here in this review, we report the morphological characteristics of the molecular EC classification currently identified in the literature. Given the new challenges in EC diagnostics, this review discusses, therefore, the potential supportive role that DL could have, by providing an outlook on all relevant studies using DL on histopathology images in various cancer types with a focus on EC. Finally, we touch upon how DL might shape the management of future EC patients.
Collapse
Affiliation(s)
- Sarah Fremond
- Department of Pathology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Viktor Hendrik Koelzer
- Department of Pathology and Molecular Pathology, University Hospital and University of Zürich, Zürich, Switzerland
| | - Nanda Horeweg
- Department of Radiotherapy, Leiden University Medical Center, Leiden, Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center (LUMC), Leiden, Netherlands
- *Correspondence: Tjalling Bosse,
| |
Collapse
|
169
|
Da Q, Huang X, Li Z, Zuo Y, Zhang C, Liu J, Chen W, Li J, Xu D, Hu Z, Yi H, Guo Y, Wang Z, Chen L, Zhang L, He X, Zhang X, Mei K, Zhu C, Lu W, Shen L, Shi J, Li J, S S, Krishnamurthi G, Yang J, Lin T, Song Q, Liu X, Graham S, Bashir RMS, Yang C, Qin S, Tian X, Yin B, Zhao J, Metaxas DN, Li H, Wang C, Zhang S. DigestPath: A benchmark dataset with challenge review for the pathological detection and segmentation of digestive-system. Med Image Anal 2022; 80:102485. [DOI: 10.1016/j.media.2022.102485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 04/08/2022] [Accepted: 05/20/2022] [Indexed: 12/19/2022]
|
170
|
Orsulic S, John J, Walts AE, Gertych A. Computational pathology in ovarian cancer. Front Oncol 2022; 12:924945. [PMID: 35965569 PMCID: PMC9372445 DOI: 10.3389/fonc.2022.924945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
Histopathologic evaluations of tissue sections are key to diagnosing and managing ovarian cancer. Pathologists empirically assess and integrate visual information, such as cellular density, nuclear atypia, mitotic figures, architectural growth patterns, and higher-order patterns, to determine the tumor type and grade, which guides oncologists in selecting appropriate treatment options. Latent data embedded in pathology slides can be extracted using computational imaging. Computers can analyze digital slide images to simultaneously quantify thousands of features, some of which are visible with a manual microscope, such as nuclear size and shape, while others, such as entropy, eccentricity, and fractal dimensions, are quantitatively beyond the grasp of the human mind. Applications of artificial intelligence and machine learning tools to interpret digital image data provide new opportunities to explore and quantify the spatial organization of tissues, cells, and subcellular structures. In comparison to genomic, epigenomic, transcriptomic, and proteomic patterns, morphologic and spatial patterns are expected to be more informative as quantitative biomarkers of complex and dynamic tumor biology. As computational pathology is not limited to visual data, nuanced subvisual alterations that occur in the seemingly “normal” pre-cancer microenvironment could facilitate research in early cancer detection and prevention. Currently, efforts to maximize the utility of computational pathology are focused on integrating image data with other -omics platforms that lack spatial information, thereby providing a new way to relate the molecular, spatial, and microenvironmental characteristics of cancer. Despite a dire need for improvements in ovarian cancer prevention, early detection, and treatment, the ovarian cancer field has lagged behind other cancers in the application of computational pathology. The intent of this review is to encourage ovarian cancer research teams to apply existing and/or develop additional tools in computational pathology for ovarian cancer and actively contribute to advancing this important field.
Collapse
Affiliation(s)
- Sandra Orsulic
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, United States
- *Correspondence: Sandra Orsulic,
| | - Joshi John
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Arkadiusz Gertych
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Faculty of Biomedical Engineering, Silesian University of Technology, Zabrze, Poland
| |
Collapse
|
171
|
Combined Hepatocellular-Cholangiocarcinoma: An Update on Pathology and Diagnostic Approach. Biomedicines 2022; 10:biomedicines10081826. [PMID: 36009374 PMCID: PMC9405224 DOI: 10.3390/biomedicines10081826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 12/04/2022] Open
Abstract
Combined hepatocellular-cholangiocarcinoma (cHCC-CCA) is a rare primary liver carcinoma displaying both hepatocytic and cholangiocytic differentiation within the same tumor. Relative to classic hepatocellular carcinoma (HCC), cHCC-CCA has more aggressive behavior and a poorer prognosis. Though recent advances have improved our understanding of the biology underlying cHCC-CCAs, they remain diagnostically challenging for pathologists because of their morphologic and phenotypic diversity. Accurate diagnosis of cHCC-CCA is important for patient management and prognostication. Herein, we review recent updates on cHCC-CCA, focusing on tumor classification, pathology, and diagnostic approach.
Collapse
|
172
|
Huang H, Zhou G, Liu X, Deng L, Wu C, Zhang D, Liu H. Contrastive learning-based computational histopathology predict differential expression of cancer driver genes. Brief Bioinform 2022; 23:6651307. [PMID: 35901472 DOI: 10.1093/bib/bbac294] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/16/2022] [Accepted: 06/29/2022] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Digital pathological analysis is run as the main examination used for cancer diagnosis. Recently, deep learning-driven feature extraction from pathology images is able to detect genetic variations and tumor environment, but few studies focus on differential gene expression in tumor cells. RESULTS In this paper, we propose a self-supervised contrastive learning framework, HistCode, to infer differential gene expression from whole slide images (WSIs). We leveraged contrastive learning on large-scale unannotated WSIs to derive slide-level histopathological features in latent space, and then transfer it to tumor diagnosis and prediction of differentially expressed cancer driver genes. Our experiments showed that our method outperformed other state-of-the-art models in tumor diagnosis tasks, and also effectively predicted differential gene expression. Interestingly, we found the genes with higher fold change can be more precisely predicted. To intuitively illustrate the ability to extract informative features from pathological images, we spatially visualized the WSIs colored by the attention scores of image tiles. We found that the tumor and necrosis areas were highly consistent with the annotations of experienced pathologists. Moreover, the spatial heatmap generated by lymphocyte-specific gene expression patterns was also consistent with the manually labeled WSIs.
Collapse
Affiliation(s)
- Haojie Huang
- School of Computer Science and Engineering, Central South University, 410075, Changsha, China
| | - Gongming Zhou
- School of Computer Science and Engineering, Central South University, 410075, Changsha, China
| | - Xuejun Liu
- School of Computer Science and Technology, Nanjing Tech University, 211816, Nanjing, China
| | - Lei Deng
- School of Computer Science and Engineering, Central South University, 410075, Changsha, China
| | - Chen Wu
- The third affiliated hospital of Soochow University, 213100, Changzhou, China
| | - Dachuan Zhang
- The third affiliated hospital of Soochow University, 213100, Changzhou, China
| | - Hui Liu
- School of Computer Science and Technology, Nanjing Tech University, 211816, Nanjing, China
| |
Collapse
|
173
|
Abedi V, Kawamura Y, Li J, Phan TG, Zand R. Editorial: Machine Learning in Action: Stroke Diagnosis and Outcome Prediction. Front Neurol 2022; 13:984467. [PMID: 35937051 PMCID: PMC9346061 DOI: 10.3389/fneur.2022.984467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Vida Abedi
- Department of Public Health Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
- *Correspondence: Vida Abedi
| | - Yuki Kawamura
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Jiang Li
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Health System, Danville, PA, United States
| | - Thanh G. Phan
- Stroke and Aging Research Group, Clinical Trials, Imaging and Informatics Division, School of Clinical Sciences at Monash Health, Melbourne, VIC, Australia
- Department of Neurology, Monash Health, Melbourne, VIC, Australia
| | - Ramin Zand
- Department of Neurology, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
174
|
Zeng Y, Wei Z, Yu W, Yin R, Yuan Y, Li B, Tang Z, Lu Y, Yang Y. Spatial transcriptomics prediction from histology jointly through Transformer and graph neural networks. Brief Bioinform 2022; 23:6645485. [PMID: 35849101 DOI: 10.1093/bib/bbac297] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/12/2022] [Accepted: 06/29/2022] [Indexed: 12/16/2022] Open
Abstract
The rapid development of spatial transcriptomics allows the measurement of RNA abundance at a high spatial resolution, making it possible to simultaneously profile gene expression, spatial locations of cells or spots, and the corresponding hematoxylin and eosin-stained histology images. It turns promising to predict gene expression from histology images that are relatively easy and cheap to obtain. For this purpose, several methods are devised, but they have not fully captured the internal relations of the 2D vision features or spatial dependency between spots. Here, we developed Hist2ST, a deep learning-based model to predict RNA-seq expression from histology images. Around each sequenced spot, the corresponding histology image is cropped into an image patch and fed into a convolutional module to extract 2D vision features. Meanwhile, the spatial relations with the whole image and neighbored patches are captured through Transformer and graph neural network modules, respectively. These learned features are then used to predict the gene expression by following the zero-inflated negative binomial distribution. To alleviate the impact by the small spatial transcriptomics data, a self-distillation mechanism is employed for efficient learning of the model. By comprehensive tests on cancer and normal datasets, Hist2ST was shown to outperform existing methods in terms of both gene expression prediction and spatial region identification. Further pathway analyses indicated that our model could reserve biological information. Thus, Hist2ST enables generating spatial transcriptomics data from histology images for elucidating molecular signatures of tissues.
Collapse
Affiliation(s)
- Yuansong Zeng
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Zhuoyi Wei
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Weijiang Yu
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Rui Yin
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Yuchen Yuan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Bingling Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhonghui Tang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yutong Lu
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Yuedong Yang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China.,Key Laboratory of Machine Intelligence and Advanced Computing (MOE), Guangzhou 510000, China
| |
Collapse
|
175
|
Fu X, Bates PA. Application of deep learning methods: From molecular modelling to patient classification. Exp Cell Res 2022; 418:113278. [PMID: 35810775 DOI: 10.1016/j.yexcr.2022.113278] [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: 01/10/2022] [Revised: 06/16/2022] [Accepted: 07/05/2022] [Indexed: 11/28/2022]
Abstract
We are now well into the information driven age with complex, heterogeneous, datasets in the biological sciences continuing to grow at a rapid pace. Moreover, distilling of such datasets, to find new governing principles, are underway. Leading the surge are new and exciting algorithmic developments in computer simulation and machine learning, most notably for the latter, those centred on deep learning. However, practical applications of cell centric computations within the biological sciences, even when carefully benchmarked against existing experimental datasets, remain challenging. Here we discuss the application of deep learning methodologies to support our understanding of cell functionality and as an aid to patient classification. Whilst comprehensive end-to-end deep learning approaches that utilise knowledge of the cell and its molecular components to aid human disease classification are yet to be implemented, important for opening the door to more effective molecular and cell-based therapies, we illustrate that many deep learning applications have been developed to tackle components of such an ambitious pipeline. We end our discussion on what the future may hold, especially how an integrated framework of computer simulations and deep learning, in conjunction with wet-bench experimentation, could enable to reveal the governing principles underlying cell functionalities within the tissue environments cells operate.
Collapse
Affiliation(s)
- Xiao Fu
- Biomolecular Modelling Laboratory, The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK.
| | - Paul A Bates
- Biomolecular Modelling Laboratory, The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK.
| |
Collapse
|
176
|
Li X, Cen M, Xu J, Zhang H, Xu XS. Improving feature extraction from histopathological images through a fine-tuning ImageNet model. J Pathol Inform 2022; 13:100115. [PMID: 36268072 PMCID: PMC9577036 DOI: 10.1016/j.jpi.2022.100115] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/05/2022] [Accepted: 06/24/2022] [Indexed: 11/04/2022] Open
Abstract
Background Due to lack of annotated pathological images, transfer learning has been the predominant approach in the field of digital pathology. Pre-trained neural networks based on ImageNet database are often used to extract "off-the-shelf" features, achieving great success in predicting tissue types, molecular features, and clinical outcomes, etc. We hypothesize that fine-tuning the pre-trained models using histopathological images could further improve feature extraction, and downstream prediction performance. Methods We used 100 000 annotated H&E image patches for colorectal cancer (CRC) to fine-tune a pre-trained Xception model via a 2-step approach. The features extracted from fine-tuned Xception (FTX-2048) model and Image-pretrained (IMGNET-2048) model were compared through: (1) tissue classification for H&E images from CRC, same image type that was used for fine-tuning; (2) prediction of immune-related gene expression, and (3) gene mutations for lung adenocarcinoma (LUAD). Five-fold cross validation was used for model performance evaluation. Each experiment was repeated 50 times. Findings The extracted features from the fine-tuned FTX-2048 exhibited significantly higher accuracy (98.4%) for predicting tissue types of CRC compared to the "off-the-shelf" features directly from Xception based on ImageNet database (96.4%) (P value = 2.2 × 10-6). Particularly, FTX-2048 markedly improved the accuracy for stroma from 87% to 94%. Similarly, features from FTX-2048 boosted the prediction of transcriptomic expression of immune-related genes in LUAD. For the genes that had significant relationships with image features (P < 0.05, n = 171), the features from the fine-tuned model improved the prediction for the majority of the genes (139; 81%). In addition, features from FTX-2048 improved prediction of mutation for 5 out of 9 most frequently mutated genes (STK11, TP53, LRP1B, NF1, and FAT1) in LUAD. Conclusions We proved the concept that fine-tuning the pretrained ImageNet neural networks with histopathology images can produce higher quality features and better prediction performance for not only the same-cancer tissue classification where similar images from the same cancer are used for fine-tuning, but also cross-cancer prediction for gene expression and mutation at patient level.
Collapse
Affiliation(s)
- Xingyu Li
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Min Cen
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jinfeng Xu
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong
| | - Hong Zhang
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xu Steven Xu
- Clinical Pharmacology and Quantitative Science, Genmab Inc., Princeton, New Jersey, USA
| |
Collapse
|
177
|
Foroughi pour A, White BS, Park J, Sheridan TB, Chuang JH. Deep learning features encode interpretable morphologies within histological images. Sci Rep 2022; 12:9428. [PMID: 35676395 PMCID: PMC9177767 DOI: 10.1038/s41598-022-13541-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
Convolutional neural networks (CNNs) are revolutionizing digital pathology by enabling machine learning-based classification of a variety of phenotypes from hematoxylin and eosin (H&E) whole slide images (WSIs), but the interpretation of CNNs remains difficult. Most studies have considered interpretability in a post hoc fashion, e.g. by presenting example regions with strongly predicted class labels. However, such an approach does not explain the biological features that contribute to correct predictions. To address this problem, here we investigate the interpretability of H&E-derived CNN features (the feature weights in the final layer of a transfer-learning-based architecture). While many studies have incorporated CNN features into predictive models, there has been little empirical study of their properties. We show such features can be construed as abstract morphological genes (“mones”) with strong independent associations to biological phenotypes. Many mones are specific to individual cancer types, while others are found in multiple cancers especially from related tissue types. We also observe that mone-mone correlations are strong and robustly preserved across related cancers. Importantly, linear mone-based classifiers can very accurately separate 38 distinct classes (19 tumor types and their adjacent normals, AUC = \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$97.1\% \pm 2.8\%$$\end{document}97.1%±2.8% for each class prediction), and linear classifiers are also highly effective for universal tumor detection (AUC = \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$99.2\% \pm 0.12\%$$\end{document}99.2%±0.12%). This linearity provides evidence that individual mones or correlated mone clusters may be associated with interpretable histopathological features or other patient characteristics. In particular, the statistical similarity of mones to gene expression values allows integrative mone analysis via expression-based bioinformatics approaches. We observe strong correlations between individual mones and individual gene expression values, notably mones associated with collagen gene expression in ovarian cancer. Mone-expression comparisons also indicate that immunoglobulin expression can be identified using mones in colon adenocarcinoma and that immune activity can be identified across multiple cancer types, and we verify these findings by expert histopathological review. Our work demonstrates that mones provide a morphological H&E decomposition that can be effectively associated with diverse phenotypes, analogous to the interpretability of transcription via gene expression values. Our work also demonstrates mones can be interpreted without using a classifier as a proxy.
Collapse
|
178
|
Ye Y, Lui VCH, Tam PKH. Pathogenesis of Choledochal Cyst: Insights from Genomics and Transcriptomics. Genes (Basel) 2022; 13:genes13061030. [PMID: 35741793 PMCID: PMC9223186 DOI: 10.3390/genes13061030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 12/10/2022] Open
Abstract
Choledochal cysts (CC) is characterized by extra- and/or intra-hepatic b\ile duct dilations. There are two main theories, “pancreaticobiliary maljunction” and “congenital stenosis of bile ducts” proposed for the pathogenesis of CC. Although family cases or CC associated with other anomalies have been reported, the molecular pathogenesis of CC is still poorly understood. Recent advances in transcriptomics and genomics analysis platforms have unveiled key expression signatures/genes/signaling pathways in the pathogenesis of human diseases including CC. This review summarizes insights from genomics and transcriptomics studies into the pathogenesis of CC, with the aim to improve (i) our understanding of its underlying complex pathomechanisms, and (ii) clinical management of different subtypes of CC, in particular their associated hepatic fibrotic change and their risk of malignancy transformation.
Collapse
Affiliation(s)
- Yongqin Ye
- Faculty of Medicine, Macau University of Science and Technology, Macau, China;
- Department of Surgery, School of Clinical Medicine, University of Hong Kong, Hong Kong, China;
| | - Vincent Chi Hang Lui
- Department of Surgery, School of Clinical Medicine, University of Hong Kong, Hong Kong, China;
| | - Paul Kwong Hang Tam
- Faculty of Medicine, Macau University of Science and Technology, Macau, China;
- Correspondence:
| |
Collapse
|
179
|
Weakly-supervised tumor purity prediction from frozen H&E stained slides. EBioMedicine 2022; 80:104067. [PMID: 35644123 PMCID: PMC9157012 DOI: 10.1016/j.ebiom.2022.104067] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background Estimating tumor purity is especially important in the age of precision medicine. Purity estimates have been shown to be critical for correction of tumor sequencing results, and higher purity samples allow for more accurate interpretations from next-generation sequencing results. Molecular-based purity estimates using computational approaches require sequencing of tumors, which is both time-consuming and expensive. Methods Here we propose an approach, weakly-supervised purity (wsPurity), which can accurately quantify tumor purity within a digitally captured hematoxylin and eosin (H&E) stained histological slide, using several types of cancer from The Cancer Genome Atlas (TCGA) as a proof-of-concept. Findings Our model predicts cancer type with high accuracy on unseen cancer slides from TCGA and shows promising generalizability to unseen data from an external cohort (F1-score of 0.83 for prostate adenocarcinoma). In addition we compare performance of our model on tumor purity prediction with a comparable fully-supervised approach on our TCGA held-out cohort and show our model has improved performance, as well as generalizability to unseen frozen slides (0.1543 MAE on an independent test cohort). In addition to tumor purity prediction, our approach identified high resolution tumor regions within a slide, and can also be used to stratify tumors into high and low tumor purity, using different cancer-dependent thresholds. Interpretation Overall, we demonstrate our deep learning model's different capabilities to analyze tumor H&E sections. We show our model is generalizable to unseen H&E stained slides from data from TCGA as well as data processed at Weill Cornell Medicine. Funding Starr Cancer Consortium Grant (SCC I15-0027) to Iman Hajirasouliha.
Collapse
|
180
|
When artificial intelligence meets PD-1/PD-L1 inhibitors: Population screening, response prediction and efficacy evaluation. Comput Biol Med 2022; 145:105499. [DOI: 10.1016/j.compbiomed.2022.105499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/26/2022] [Accepted: 04/03/2022] [Indexed: 02/07/2023]
|
181
|
Alam MR, Abdul-Ghafar J, Yim K, Thakur N, Lee SH, Jang HJ, Jung CK, Chong Y. Recent Applications of Artificial Intelligence from Histopathologic Image-Based Prediction of Microsatellite Instability in Solid Cancers: A Systematic Review. Cancers (Basel) 2022; 14:2590. [PMID: 35681570 PMCID: PMC9179592 DOI: 10.3390/cancers14112590] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/07/2022] [Accepted: 05/22/2022] [Indexed: 12/11/2022] Open
Abstract
Cancers with high microsatellite instability (MSI-H) have a better prognosis and respond well to immunotherapy. However, MSI is not tested in all cancers because of the additional costs and time of diagnosis. Therefore, artificial intelligence (AI)-based models have been recently developed to evaluate MSI from whole slide images (WSIs). Here, we aimed to assess the current state of AI application to predict MSI based on WSIs analysis in MSI-related cancers and suggest a better study design for future studies. Studies were searched in online databases and screened by reference type, and only the full texts of eligible studies were reviewed. The included 14 studies were published between 2018 and 2021, and most of the publications were from developed countries. The commonly used dataset is The Cancer Genome Atlas dataset. Colorectal cancer (CRC) was the most common type of cancer studied, followed by endometrial, gastric, and ovarian cancers. The AI models have shown the potential to predict MSI with the highest AUC of 0.93 in the case of CRC. The relatively limited scale of datasets and lack of external validation were the limitations of most studies. Future studies with larger datasets are required to implicate AI models in routine diagnostic practice for MSI prediction.
Collapse
Affiliation(s)
- Mohammad Rizwan Alam
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Jamshid Abdul-Ghafar
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Kwangil Yim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Nishant Thakur
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Hyun-Jong Jang
- Catholic Big Data Integration Center, Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Yosep Chong
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| |
Collapse
|
182
|
Weitz P, Wang Y, Kartasalo K, Egevad L, Lindberg J, Grönberg H, Eklund M, Rantalainen M. Transcriptome-wide prediction of prostate cancer gene expression from histopathology images using co-expression based convolutional neural networks. Bioinformatics 2022; 38:3462-3469. [PMID: 35595235 PMCID: PMC9237721 DOI: 10.1093/bioinformatics/btac343] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 03/18/2022] [Accepted: 05/16/2022] [Indexed: 12/15/2022] Open
Abstract
Motivation Molecular phenotyping by gene expression profiling is central in contemporary cancer research and in molecular diagnostics but remains resource intense to implement. Changes in gene expression occurring in tumours cause morphological changes in tissue, which can be observed on the microscopic level. The relationship between morphological patterns and some of the molecular phenotypes can be exploited to predict molecular phenotypes from routine haematoxylin and eosin-stained whole slide images (WSIs) using convolutional neural networks (CNNs). In this study, we propose a new, computationally efficient approach to model relationships between morphology and gene expression. Results We conducted the first transcriptome-wide analysis in prostate cancer, using CNNs to predict bulk RNA-sequencing estimates from WSIs for 370 patients from the TCGA PRAD study. Out of 15 586 protein coding transcripts, 6618 had predicted expression significantly associated with RNA-seq estimates (FDR-adjusted P-value <1×10−4) in a cross-validation and 5419 (81.9%) of these associations were subsequently validated in a held-out test set. We furthermore predicted the prognostic cell-cycle progression score directly from WSIs. These findings suggest that contemporary computer vision models offer an inexpensive and scalable solution for prediction of gene expression phenotypes directly from WSIs, providing opportunity for cost-effective large-scale research studies and molecular diagnostics. Availability and implementation A self-contained example is available from http://github.com/phiwei/prostate_coexpression. Model predictions and metrics are available from doi.org/10.5281/zenodo.4739097. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Philippe Weitz
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Yinxi Wang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Kimmo Kartasalo
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden.,Faculty of Medicine and Health Technology, Tampere University, Tampere, 33100, Finland
| | - Lars Egevad
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden.,Science for Life Laboratory, Stockholm, 17177, Sweden
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Martin Eklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Mattias Rantalainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| |
Collapse
|
183
|
Bankhead P. Developing image analysis methods for digital pathology. J Pathol 2022; 257:391-402. [PMID: 35481680 PMCID: PMC9324951 DOI: 10.1002/path.5921] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/04/2022]
Abstract
The potential to use quantitative image analysis and artificial intelligence is one of the driving forces behind digital pathology. However, despite novel image analysis methods for pathology being described across many publications, few become widely adopted and many are not applied in more than a single study. The explanation is often straightforward: software implementing the method is simply not available, or is too complex, incomplete, or dataset‐dependent for others to use. The result is a disconnect between what seems already possible in digital pathology based upon the literature, and what actually is possible for anyone wishing to apply it using currently available software. This review begins by introducing the main approaches and techniques involved in analysing pathology images. I then examine the practical challenges inherent in taking algorithms beyond proof‐of‐concept, from both a user and developer perspective. I describe the need for a collaborative and multidisciplinary approach to developing and validating meaningful new algorithms, and argue that openness, implementation, and usability deserve more attention among digital pathology researchers. The review ends with a discussion about how digital pathology could benefit from interacting with and learning from the wider bioimage analysis community, particularly with regard to sharing data, software, and ideas. © 2022 The Author. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Peter Bankhead
- Edinburgh Pathology, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.,Centre for Genomic & Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.,Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
184
|
Swarm learning for decentralized artificial intelligence in cancer histopathology. Nat Med 2022; 28:1232-1239. [PMID: 35469069 PMCID: PMC9205774 DOI: 10.1038/s41591-022-01768-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/02/2022] [Indexed: 02/07/2023]
Abstract
Artificial intelligence (AI) can predict the presence of molecular alterations directly from routine histopathology slides. However, training robust AI systems requires large datasets for which data collection faces practical, ethical and legal obstacles. These obstacles could be overcome with swarm learning (SL), in which partners jointly train AI models while avoiding data transfer and monopolistic data governance. Here, we demonstrate the successful use of SL in large, multicentric datasets of gigapixel histopathology images from over 5,000 patients. We show that AI models trained using SL can predict BRAF mutational status and microsatellite instability directly from hematoxylin and eosin (H&E)-stained pathology slides of colorectal cancer. We trained AI models on three patient cohorts from Northern Ireland, Germany and the United States, and validated the prediction performance in two independent datasets from the United Kingdom. Our data show that SL-trained AI models outperform most locally trained models, and perform on par with models that are trained on the merged datasets. In addition, we show that SL-based AI models are data efficient. In the future, SL can be used to train distributed AI models for any histopathology image analysis task, eliminating the need for data transfer. A decentralized, privacy-preserving machine learning framework used to train a clinically relevant AI system identifies actionable molecular alterations in patients with colorectal cancer by use of routine histopathology slides collected in real-world settings.
Collapse
|
185
|
Communicator-Driven Data Preprocessing Improves Deep Transfer Learning of Histopathological Prediction of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14081964. [PMID: 35454869 PMCID: PMC9031738 DOI: 10.3390/cancers14081964] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Pancreatic cancer has a dismal prognosis and its diagnosis can be challenging. Histopathological slides can be digitalized and their analysis can then be supported by computer algorithms. For this purpose, computer algorithms (neural networks) need to be trained to detect the desired tissue type (e.g., pancreatic cancer). However, raw training data often contain many different tissue types. Here we show a preprocessing step using two communicators that sort unfitting tissue tiles into a new dataset class. Using the improved dataset neural networks distinguished pancreatic cancer from other tissue types on digitalized histopathological slides including lymph node metastases. Abstract Pancreatic cancer is a fatal malignancy with poor prognosis and limited treatment options. Early detection in primary and secondary locations is critical, but fraught with challenges. While digital pathology can assist with the classification of histopathological images, the training of such networks always relies on a ground truth, which is frequently compromised as tissue sections contain several types of tissue entities. Here we show that pancreatic cancer can be detected on hematoxylin and eosin (H&E) sections by convolutional neural networks using deep transfer learning. To improve the ground truth, we describe a preprocessing data clean-up process using two communicators that were generated through existing and new datasets. Specifically, the communicators moved image tiles containing adipose tissue and background to a new data class. Hence, the original dataset exhibited improved labeling and, consequently, a higher ground truth accuracy. Deep transfer learning of a ResNet18 network resulted in a five-class accuracy of about 94% on test data images. The network was validated with independent tissue sections composed of healthy pancreatic tissue, pancreatic ductal adenocarcinoma, and pancreatic cancer lymph node metastases. The screening of different models and hyperparameter fine tuning were performed to optimize the performance with the independent tissue sections. Taken together, we introduce a step of data preprocessing via communicators as a means of improving the ground truth during deep transfer learning and hyperparameter tuning to identify pancreatic ductal adenocarcinoma primary tumors and metastases in histological tissue sections.
Collapse
|
186
|
Carrillo-Perez F, Morales JC, Castillo-Secilla D, Gevaert O, Rojas I, Herrera LJ. Machine-Learning-Based Late Fusion on Multi-Omics and Multi-Scale Data for Non-Small-Cell Lung Cancer Diagnosis. J Pers Med 2022; 12:601. [PMID: 35455716 PMCID: PMC9025878 DOI: 10.3390/jpm12040601] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 03/29/2022] [Accepted: 04/06/2022] [Indexed: 01/27/2023] Open
Abstract
Differentiation between the various non-small-cell lung cancer subtypes is crucial for providing an effective treatment to the patient. For this purpose, machine learning techniques have been used in recent years over the available biological data from patients. However, in most cases this problem has been treated using a single-modality approach, not exploring the potential of the multi-scale and multi-omic nature of cancer data for the classification. In this work, we study the fusion of five multi-scale and multi-omic modalities (RNA-Seq, miRNA-Seq, whole-slide imaging, copy number variation, and DNA methylation) by using a late fusion strategy and machine learning techniques. We train an independent machine learning model for each modality and we explore the interactions and gains that can be obtained by fusing their outputs in an increasing manner, by using a novel optimization approach to compute the parameters of the late fusion. The final classification model, using all modalities, obtains an F1 score of 96.81±1.07, an AUC of 0.993±0.004, and an AUPRC of 0.980±0.016, improving those results that each independent model obtains and those presented in the literature for this problem. These obtained results show that leveraging the multi-scale and multi-omic nature of cancer data can enhance the performance of single-modality clinical decision support systems in personalized medicine, consequently improving the diagnosis of the patient.
Collapse
Affiliation(s)
- Francisco Carrillo-Perez
- Department of Computer Architecture and Technology, University of Granada, C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18170 Granada, Spain; (J.C.M.); (I.R.); (L.J.H.)
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, 1265 Welch Rd, Stanford, CA 94305, USA;
| | - Juan Carlos Morales
- Department of Computer Architecture and Technology, University of Granada, C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18170 Granada, Spain; (J.C.M.); (I.R.); (L.J.H.)
| | - Daniel Castillo-Secilla
- Fujitsu Technology Solutions S.A, CoE Data Intelligence, Camino del Cerro de los Gamos, 1, Pozuelo de Alarcón, 28224 Madrid, Spain;
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, 1265 Welch Rd, Stanford, CA 94305, USA;
| | - Ignacio Rojas
- Department of Computer Architecture and Technology, University of Granada, C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18170 Granada, Spain; (J.C.M.); (I.R.); (L.J.H.)
| | - Luis Javier Herrera
- Department of Computer Architecture and Technology, University of Granada, C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18170 Granada, Spain; (J.C.M.); (I.R.); (L.J.H.)
| |
Collapse
|
187
|
Area under the curve may hide poor generalisation to external datasets. ESMO Open 2022; 7:100429. [PMID: 35397433 PMCID: PMC9006654 DOI: 10.1016/j.esmoop.2022.100429] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 11/23/2022] Open
|
188
|
Echle A, Ghaffari Laleh N, Quirke P, Grabsch HI, Muti HS, Saldanha OL, Brockmoeller SF, van den Brandt PA, Hutchins GGA, Richman SD, Horisberger K, Galata C, Ebert MP, Eckardt M, Boutros M, Horst D, Reissfelder C, Alwers E, Brinker TJ, Langer R, Jenniskens JCA, Offermans K, Mueller W, Gray R, Gruber SB, Greenson JK, Rennert G, Bonner JD, Schmolze D, Chang-Claude J, Brenner H, Trautwein C, Boor P, Jaeger D, Gaisa NT, Hoffmeister M, West NP, Kather JN. Artificial intelligence for detection of microsatellite instability in colorectal cancer-a multicentric analysis of a pre-screening tool for clinical application. ESMO Open 2022; 7:100400. [PMID: 35247870 PMCID: PMC9058894 DOI: 10.1016/j.esmoop.2022.100400] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Microsatellite instability (MSI)/mismatch repair deficiency (dMMR) is a key genetic feature which should be tested in every patient with colorectal cancer (CRC) according to medical guidelines. Artificial intelligence (AI) methods can detect MSI/dMMR directly in routine pathology slides, but the test performance has not been systematically investigated with predefined test thresholds. METHOD We trained and validated AI-based MSI/dMMR detectors and evaluated predefined performance metrics using nine patient cohorts of 8343 patients across different countries and ethnicities. RESULTS Classifiers achieved clinical-grade performance, yielding an area under the receiver operating curve (AUROC) of up to 0.96 without using any manual annotations. Subsequently, we show that the AI system can be applied as a rule-out test: by using cohort-specific thresholds, on average 52.73% of tumors in each surgical cohort [total number of MSI/dMMR = 1020, microsatellite stable (MSS)/ proficient mismatch repair (pMMR) = 7323 patients] could be identified as MSS/pMMR with a fixed sensitivity at 95%. In an additional cohort of N = 1530 (MSI/dMMR = 211, MSS/pMMR = 1319) endoscopy biopsy samples, the system achieved an AUROC of 0.89, and the cohort-specific threshold ruled out 44.12% of tumors with a fixed sensitivity at 95%. As a more robust alternative to cohort-specific thresholds, we showed that with a fixed threshold of 0.25 for all the cohorts, we can rule-out 25.51% in surgical specimens and 6.10% in biopsies. INTERPRETATION When applied in a clinical setting, this means that the AI system can rule out MSI/dMMR in a quarter (with global thresholds) or half of all CRC patients (with local fine-tuning), thereby reducing cost and turnaround time for molecular profiling.
Collapse
Affiliation(s)
- A Echle
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - N Ghaffari Laleh
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - P Quirke
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - H I Grabsch
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK; Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - H S Muti
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - O L Saldanha
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - S F Brockmoeller
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - P A van den Brandt
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - G G A Hutchins
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - S D Richman
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - K Horisberger
- Department of Abdominal and Transplantation Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - C Galata
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Division of Thoracic Surgery, Academic Thoracic Center Mainz, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - M P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Institute for Innate Immunoscience (MI3) and Clinical Cooperation Unit Healthy Metabolism, Center of Preventive Medicine and Digital Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - M Eckardt
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - M Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - D Horst
- Institut für Pathologie Charité, Berlin, Germany
| | - C Reissfelder
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - E Alwers
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - T J Brinker
- Digital Biomarkers for Oncology Group, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - R Langer
- Institute of Pathology, Inselspital, University of Bern, Bern, Switzerland
| | - J C A Jenniskens
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - K Offermans
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - W Mueller
- Gemeinschaftspraxis Pathologie, Starnberg, Germany
| | - R Gray
- Clinical Trial Service Unit, University of Oxford, Oxford, UK
| | - S B Gruber
- Center for Precision Medicine and Department of Medical Oncology, City of Hope National Medical Center, Duarte, USA
| | - J K Greenson
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, USA
| | - G Rennert
- Department of Community Medicine & Epidemiology, Lady Davis Carmel Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Steve and Cindy Rasmussen Institute for Genomic Medicine, Lady Davis Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - J D Bonner
- Center for Precision Medicine and Department of Medical Oncology, City of Hope National Medical Center, Duarte, USA
| | - D Schmolze
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, USA
| | - J Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Cancer Epidemiology Group, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - H Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - P Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany; Department of Nephrology and Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - D Jaeger
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - N T Gaisa
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - M Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - N P West
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - J N Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany; Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
189
|
Cifci D, Foersch S, Kather JN. Artificial intelligence to identify genetic alterations in conventional histopathology. J Pathol 2022; 257:430-444. [PMID: 35342954 DOI: 10.1002/path.5898] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 11/10/2022]
Abstract
Precision oncology relies on the identification of targetable molecular alterations in tumor tissues. In many tumor types, a limited set of molecular tests is currently part of standard diagnostic workflows. However, universal testing for all targetable alterations, especially rare ones, is limited by the cost and availability of molecular assays. From 2017 to 2021, multiple studies have shown that artificial intelligence (AI) methods can predict the probability of specific genetic alterations directly from conventional hematoxylin and eosin (H&E) tissue slides. Although these methods are currently less accurate than gold-standard testing (e.g. immunohistochemistry, polymerase chain reaction or next-generation sequencing), they could be used as pre-screening tools to reduce the workload of genetic analyses. In this systematic literature review, we summarize the state of the art in predicting molecular alterations from H&E using AI. We found that AI methods perform reasonably well across multiple tumor types, although few algorithms have been broadly validated. In addition, we found that genetic alterations in FGFR, IDH, PIK3CA, BRAF, TP53 and DNA repair pathways are predictable from H&E in multiple tumor types, while many other genetic alterations have rarely been investigated or were only poorly predictable. Finally, we discuss the next steps for the implementation of AI-based surrogate tests in diagnostic workflows. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Didem Cifci
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.,Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.,Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
190
|
Zeng Z, Li Y, Li Y, Luo Y. Statistical and machine learning methods for spatially resolved transcriptomics data analysis. Genome Biol 2022; 23:83. [PMID: 35337374 PMCID: PMC8951701 DOI: 10.1186/s13059-022-02653-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 03/15/2022] [Indexed: 01/28/2023] Open
Abstract
The recent advancement in spatial transcriptomics technology has enabled multiplexed profiling of cellular transcriptomes and spatial locations. As the capacity and efficiency of the experimental technologies continue to improve, there is an emerging need for the development of analytical approaches. Furthermore, with the continuous evolution of sequencing protocols, the underlying assumptions of current analytical methods need to be re-evaluated and adjusted to harness the increasing data complexity. To motivate and aid future model development, we herein review the recent development of statistical and machine learning methods in spatial transcriptomics, summarize useful resources, and highlight the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Zexian Zeng
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100084, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100084, China
- Department of Data Sciences, Dana Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Yawei Li
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yiming Li
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yuan Luo
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Northwestern University Clinical and Translational Sciences Institute, Chicago, IL, 60611, USA.
- Institute for Augmented Intelligence in Medicine, Northwestern University, Chicago, IL, 60611, USA.
- Center for Health Information Partnerships, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
191
|
Efficient prediction of a spatial transcriptomics profile better characterizes breast cancer tissue sections without costly experimentation. Sci Rep 2022; 12:4133. [PMID: 35260632 PMCID: PMC8904587 DOI: 10.1038/s41598-022-07685-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/21/2022] [Indexed: 02/07/2023] Open
Abstract
Spatial transcriptomics is an emerging technology requiring costly reagents and considerable skills, limiting the identification of transcriptional markers related to histology. Here, we show that predicted spatial gene-expression in unmeasured regions and tissues can enhance biologists’ histological interpretations. We developed the Deep learning model for Spatial gene Clusters and Expression, DeepSpaCE, and confirmed its performance using the spatial-transcriptome profiles and immunohistochemistry images of consecutive human breast cancer tissue sections. For example, the predicted expression patterns of SPARC, an invasion marker, highlighted a small tumor-invasion region difficult to identify using raw spatial transcriptome data alone because of a lack of measurements. We further developed semi-supervised DeepSpaCE using unlabeled histology images and increased the imputation accuracy of consecutive sections, enhancing applicability for a small sample size. Our method enables users to derive hidden histological characters via spatial transcriptome and gene annotations, leading to accelerated biological discoveries without additional experiments.
Collapse
|
192
|
Lipkova J, Chen TY, Lu MY, Chen RJ, Shady M, Williams M, Wang J, Noor Z, Mitchell RN, Turan M, Coskun G, Yilmaz F, Demir D, Nart D, Basak K, Turhan N, Ozkara S, Banz Y, Odening KE, Mahmood F. Deep learning-enabled assessment of cardiac allograft rejection from endomyocardial biopsies. Nat Med 2022; 28:575-582. [PMID: 35314822 PMCID: PMC9353336 DOI: 10.1038/s41591-022-01709-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 01/19/2022] [Indexed: 02/07/2023]
Abstract
Endomyocardial biopsy (EMB) screening represents the standard of care for detecting allograft rejections after heart transplant. Manual interpretation of EMBs is affected by substantial interobserver and intraobserver variability, which often leads to inappropriate treatment with immunosuppressive drugs, unnecessary follow-up biopsies and poor transplant outcomes. Here we present a deep learning-based artificial intelligence (AI) system for automated assessment of gigapixel whole-slide images obtained from EMBs, which simultaneously addresses detection, subtyping and grading of allograft rejection. To assess model performance, we curated a large dataset from the United States, as well as independent test cohorts from Turkey and Switzerland, which includes large-scale variability across populations, sample preparations and slide scanning instrumentation. The model detects allograft rejection with an area under the receiver operating characteristic curve (AUC) of 0.962; assesses the cellular and antibody-mediated rejection type with AUCs of 0.958 and 0.874, respectively; detects Quilty B lesions, benign mimics of rejection, with an AUC of 0.939; and differentiates between low-grade and high-grade rejections with an AUC of 0.833. In a human reader study, the AI system showed non-inferior performance to conventional assessment and reduced interobserver variability and assessment time. This robust evaluation of cardiac allograft rejection paves the way for clinical trials to establish the efficacy of AI-assisted EMB assessment and its potential for improving heart transplant outcomes.
Collapse
Affiliation(s)
- Jana Lipkova
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tiffany Y Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ming Y Lu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Richard J Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Maha Shady
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Mane Williams
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Jingwen Wang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Computer Science, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Zahra Noor
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard N Mitchell
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Health Sciences and Technology (HST), Cambridge, MA, USA
| | - Mehmet Turan
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| | - Gulfize Coskun
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| | - Funda Yilmaz
- Faculty of Medicine, Department of Pathology, Ege University, Izmir, Turkey
| | - Derya Demir
- Faculty of Medicine, Department of Pathology, Ege University, Izmir, Turkey
| | - Deniz Nart
- Faculty of Medicine, Department of Pathology, Ege University, Izmir, Turkey
| | - Kayhan Basak
- Department of Pathology, University of Health Sciences, Ankara, Turkey
| | - Nesrin Turhan
- Department of Pathology, University of Health Sciences, Ankara, Turkey
| | - Selvinaz Ozkara
- Department of Pathology, University of Health Sciences, Ankara, Turkey
| | - Yara Banz
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Katja E Odening
- Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland
- Institute of Physiology, University of Bern, Bern, Switzerland
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA.
- Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
193
|
Wang X, Wang H, Liu D, Wang N, He D, Wu Z, Zhu X, Wen X, Li X, Li J, Wang Z. Deep learning using bulk RNA-seq data expands cell landscape identification in tumor microenvironment. Oncoimmunology 2022; 11:2043662. [PMID: 35251771 PMCID: PMC8890395 DOI: 10.1080/2162402x.2022.2043662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Xin Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin,China
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongjiu Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin,China
| | - Dan Liu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Na Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin,China
| | - Danni He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin,China
| | - Zheyu Wu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin,China
| | - Xu Zhu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Xiaoling Wen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Xuhua Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Jin Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Zhenzhen Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin,China
| |
Collapse
|
194
|
Park JH, Kim EY, Luchini C, Eccher A, Tizaoui K, Shin JI, Lim BJ. Artificial Intelligence for Predicting Microsatellite Instability Based on Tumor Histomorphology: A Systematic Review. Int J Mol Sci 2022; 23:2462. [PMID: 35269607 PMCID: PMC8910565 DOI: 10.3390/ijms23052462] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 02/21/2022] [Indexed: 02/04/2023] Open
Abstract
Microsatellite instability (MSI)/defective DNA mismatch repair (dMMR) is receiving more attention as a biomarker for eligibility for immune checkpoint inhibitors in advanced diseases. However, due to high costs and resource limitations, MSI/dMMR testing is not widely performed. Some attempts are in progress to predict MSI/dMMR status through histomorphological features on H&E slides using artificial intelligence (AI) technology. In this study, the potential predictive role of this new methodology was reviewed through a systematic review. Studies up to September 2021 were searched through PubMed and Embase database searches. The design and results of each study were summarized, and the risk of bias for each study was evaluated. For colorectal cancer, AI-based systems showed excellent performance with the highest standard of 0.972; for gastric and endometrial cancers they showed a relatively low but satisfactory performance, with the highest standard of 0.81 and 0.82, respectively. However, analyzing the risk of bias, most studies were evaluated at high-risk. AI-based systems showed a high potential in predicting the MSI/dMMR status of different cancer types, and particularly of colorectal cancers. Therefore, a confirmation test should be required only for the results that are positive in the AI test.
Collapse
Affiliation(s)
- Ji Hyun Park
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Eun Young Kim
- Evidence-Based and Clinical Research Laboratory, Department of Health, Social and Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea;
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy;
- ARC-Net Research Center, University and Hospital Trust of Verona, 37134 Verona, Italy
| | - Albino Eccher
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Kalthoum Tizaoui
- Laboratory of Microorganisms and Active Biomolecules, Sciences Faculty of Tunis, Tunis El Manar University, Tunis 2092, Tunisia;
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Beom Jin Lim
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| |
Collapse
|
195
|
Huang W, Randhawa R, Jain P, Hubbard S, Eickhoff J, Kummar S, Wilding G, Basu H, Roy R. A Novel Artificial Intelligence-Powered Method for Prediction of Early Recurrence of Prostate Cancer After Prostatectomy and Cancer Drivers. JCO Clin Cancer Inform 2022; 6:e2100131. [PMID: 35192404 DOI: 10.1200/cci.21.00131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To develop a novel artificial intelligence (AI)-powered method for the prediction of prostate cancer (PCa) early recurrence and identification of driver regions in PCa of all Gleason Grade Group (GGG). MATERIALS AND METHODS Deep convolutional neural networks were used to develop the AI model. The AI model was trained on The Cancer Genome Atlas Prostatic Adenocarcinoma (TCGA-PRAD) whole slide images (WSI) and data set (n = 243) to predict 3-year biochemical recurrence after radical prostatectomy (RP) and was subsequently validated on WSI from patients with PCa (n = 173) from the University of Wisconsin-Madison. RESULTS Our AI-powered platform can extract visual and subvisual morphologic features from WSI to identify driver regions predictive of early recurrence of PCa (regions of interest [ROIs]) after RP. The ROIs were ranked with AI-morphometric scores, which were prognostic for 3-year biochemical recurrence (area under the curve [AUC], 0.78), which is significantly better than the GGG overall (AUC, 0.62). The AI-morphometric scores also showed high accuracy in the prediction of recurrence for low- or intermediate-risk PCa-AUC, 0.76, 0.84, and 0.81 for GGG1, GGG2, and GGG3, respectively. These patients could benefit the most from timely adjuvant therapy after RP. The predictive value of the high-scored ROIs was validated by known PCa biomarkers studied. With this focused biomarker analysis, a potentially new STING pathway-related PCa biomarker-TMEM173-was identified. CONCLUSION Our study introduces a novel approach for identifying patients with PCa at risk for early recurrence regardless of their GGG status and for identifying cancer drivers for focused evolution-aware novel biomarker discovery.
Collapse
Affiliation(s)
- Wei Huang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI.,PathomIQ, Inc, Cupertino, CA
| | - Ramandeep Randhawa
- PathomIQ, Inc, Cupertino, CA.,University of Southern California Marshall School of Business, Los Angeles, CA
| | | | - Samuel Hubbard
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Jens Eickhoff
- Department of Biostatistics and Informatics, University of Wisconsin-Madison, Madison, WI
| | - Shivaani Kummar
- PathomIQ, Inc, Cupertino, CA.,Division of Hematology & Medical Oncology, Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | | | - Hirak Basu
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
196
|
Loeffler CML, Gaisa NT, Muti HS, van Treeck M, Echle A, Ghaffari Laleh N, Trautwein C, Heij LR, Grabsch HI, Ortiz Bruechle N, Kather JN. Predicting Mutational Status of Driver and Suppressor Genes Directly from Histopathology With Deep Learning: A Systematic Study Across 23 Solid Tumor Types. Front Genet 2022; 12:806386. [PMID: 35251119 PMCID: PMC8889144 DOI: 10.3389/fgene.2021.806386] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
In the last four years, advances in Deep Learning technology have enabled the inference of selected mutational alterations directly from routine histopathology slides. In particular, recent studies have shown that genetic changes in clinically relevant driver genes are reflected in the histological phenotype of solid tumors and can be inferred by analysing routine Haematoxylin and Eosin (H&E) stained tissue sections with Deep Learning. However, these studies mostly focused on selected individual genes in selected tumor types. In addition, genetic changes in solid tumors primarily act by changing signaling pathways that regulate cell behaviour. In this study, we hypothesized that Deep Learning networks can be trained to directly predict alterations of genes and pathways across a spectrum of solid tumors. We manually outlined tumor tissue in H&E-stained tissue sections from 7,829 patients with 23 different tumor types from The Cancer Genome Atlas. We then trained convolutional neural networks in an end-to-end way to detect alterations in the most clinically relevant pathways or genes, directly from histology images. Using this automatic approach, we found that alterations in 12 out of 14 clinically relevant pathways and numerous single gene alterations appear to be detectable in tissue sections, many of which have not been reported before. Interestingly, we show that the prediction performance for single gene alterations is better than that for pathway alterations. Collectively, these data demonstrate the predictability of genetic alterations directly from routine cancer histology images and show that individual genes leave a stronger morphological signature than genetic pathways.
Collapse
Affiliation(s)
- Chiara Maria Lavinia Loeffler
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
- *Correspondence: Chiara Maria Lavinia Loeffler,
| | - Nadine T. Gaisa
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Hannah Sophie Muti
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Marko van Treeck
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Amelie Echle
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Narmin Ghaffari Laleh
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Lara R. Heij
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Heike I. Grabsch
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, Netherlands
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, United Kingdom
| | - Nadina Ortiz Bruechle
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, United Kingdom
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
197
|
TAWSEEM: A Deep-Learning-Based Tool for Estimating the Number of Unknown Contributors in DNA Profiling. ELECTRONICS 2022. [DOI: 10.3390/electronics11040548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
DNA profiling involves the analysis of sequences of an individual or mixed DNA profiles to identify the persons that these profiles belong to. A critically important application of DNA profiling is in forensic science to identify criminals by finding a match between their blood samples and the DNA profile found on the crime scene. Other applications include paternity tests, disaster victim identification, missing person investigations, and mapping genetic diseases. A crucial task in DNA profiling is the determination of the number of contributors in a DNA mixture profile, which is challenging due to issues that include allele dropout, stutter, blobs, and noise in DNA profiles; these issues negatively affect the estimation accuracy and the computational complexity. Machine-learning-based methods have been applied for estimating the number of unknowns; however, there is limited work in this area and many more efforts are required to develop robust models and their training on large and diverse datasets. In this paper, we propose and develop a software tool called TAWSEEM that employs a multilayer perceptron (MLP) neural network deep learning model for estimating the number of unknown contributors in DNA mixture profiles using PROVEDIt, the largest publicly available dataset. We investigate the performance of our developed deep learning model using four performance metrics, namely accuracy, F1-score, recall, and precision. The novelty of our tool is evident in the fact that it provides the highest accuracy (97%) compared to any existing work on the most diverse dataset (in terms of the profiles, loci, multiplexes, etc.). We also provide a detailed background on the DNA profiling and literature review, and a detailed account of the deep learning tool development and the performance investigation of the deep learning method.
Collapse
|
198
|
Saad M, He S, Thorstad W, Gay H, Barnett D, Zhao Y, Ruan S, Wang X, Li H. Learning-based Cancer Treatment Outcome Prognosis using Multimodal Biomarkers. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2022; 6:231-244. [PMID: 35520102 PMCID: PMC9066560 DOI: 10.1109/trpms.2021.3104297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Predicting early in treatment whether a tumor is likely to be responsive is a difficult yet important task to support clinical decision-making. Studies have shown that multimodal biomarkers could provide complementary information and lead to more accurate treatment outcome prognosis than unimodal biomarkers. However, the prognosis accuracy could be affected by multimodal data heterogeneity and incompleteness. The small-sized and imbalance datasets also bring additional challenges for training a designed prognosis model. In this study, a modular framework employing multimodal biomarkers for cancer treatment outcome prediction was proposed. It includes four modules of synthetic data generation, deep feature extraction, multimodal feature fusion, and classification to address the challenges described above. The feasibility and advantages of the designed framework were demonstrated through an example study, in which the goal was to stratify oropharyngeal squamous cell carcinoma (OPSCC) patients with low- and high-risks of treatment failures by use of positron emission tomography (PET) image data and microRNA (miRNA) biomarkers. The superior prognosis performance and the comparison with other methods demonstrated the efficiency of the proposed framework and its ability of enabling seamless integration, validation and comparison of various algorithms in each module of the framework. The limitation and future work was discussed as well.
Collapse
Affiliation(s)
- Maliazurina Saad
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA. She is now with the MD Anderson Cancer Center, Houston, TX, USA
| | - Shenghua He
- Department of Computer Science and Engineering, Washington University, Saint louis, MO, USA
| | - Wade Thorstad
- Department of Radiation Oncology, Washington University School of Medicine, Saint louis, MO, USA
| | - Hiram Gay
- Department of Radiation Oncology, Washington University School of Medicine, Saint louis, MO, USA
| | - Daniel Barnett
- Carle Cancer Center, Carle Foundation Hospital, Urbana, IL, USA
| | - Yujie Zhao
- Mao Clinic at Florida, Jacksonville, FL, USA
| | - Su Ruan
- Laboratoire LITIS (EA 4108), Equipe Quantif, University of Rouen, France
| | - Xiaowei Wang
- Department of Pharmacology and Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Hua Li
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Cancer Center at Illinois, and Carle Foundation Hospital, Urbana, IL, USA
| |
Collapse
|
199
|
Alrefaei AF, Hawsawi YM, Almaleki D, Alafif T, Alzahrani FA, Bakhrebah MA. Genetic data sharing and artificial intelligence in the era of personalized medicine based on a cross-sectional analysis of the Saudi human genome program. Sci Rep 2022; 12:1405. [PMID: 35082362 PMCID: PMC8791994 DOI: 10.1038/s41598-022-05296-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/07/2022] [Indexed: 12/21/2022] Open
Abstract
The success of the Saudi Human Genome Program (SHGP), one of the top ten genomic programs worldwide, is highly dependent on the Saudi population embracing the concept of participating in genetic testing. However, genetic data sharing and artificial intelligence (AI) in genomics are critical public issues in medical care and scientific research. The present study was aimed to examine the awareness, knowledge, and attitude of the Saudi society towards the SHGP, the sharing and privacy of genetic data resulting from the SHGP, and the role of AI in genetic data analysis and regulations. Results of a questionnaire survey with 804 respondents revealed moderate awareness and attitude towards the SHGP and minimal knowledge regarding its benefits and applications. Respondents demonstrated a low level of knowledge regarding the privacy of genetic data. A generally positive attitude was found towards the outcomes of the SHGP and genetic data sharing for medical and scientific research. The highest level of knowledge was detected regarding AI use in genetic data analysis and privacy regulation. We recommend that the SHGP’s regulators launch awareness campaigns and educational programs to increase and improve public awareness and knowledge regarding the SHGP’s benefits and applications. Furthermore, we propose a strategy for genetic data sharing which will facilitate genetic data sharing between institutions and advance Personalized Medicine in genetic diseases’ diagnosis and treatment.
Collapse
Affiliation(s)
- Abdulmajeed F Alrefaei
- Department of Biology, Genetic and Molecular Biology Central Lab, Jamoum University College, Umm Al-Qura University, Makkah, 21955, Saudi Arabia.
| | - Yousef M Hawsawi
- Research Centre, King Faisal Specialist Hospital and Research Centre, P.O. Box 40047, Jeddah, 21499, Saudi Arabia.,MBC: J04/ College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Deyab Almaleki
- Department of Evaluation, Measurement, and Research, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Tarik Alafif
- Computer Science Department, Jamoum University College, Umm Al-Qura University, Jamoum, 25375, Saudi Arabia
| | - Faisal A Alzahrani
- Department of Biochemistry, Faculty of Science, Embryonic Stem Cells Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Muhammed A Bakhrebah
- King Abdulaziz City for Science and Technology (KACST), Life Science and Environment Research Institute, P.O. Box 6086, Riyadh, 11442, Saudi Arabia
| |
Collapse
|
200
|
Kang M, Ko E, Mersha TB. A roadmap for multi-omics data integration using deep learning. Brief Bioinform 2022; 23:bbab454. [PMID: 34791014 PMCID: PMC8769688 DOI: 10.1093/bib/bbab454] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 12/18/2022] Open
Abstract
High-throughput next-generation sequencing now makes it possible to generate a vast amount of multi-omics data for various applications. These data have revolutionized biomedical research by providing a more comprehensive understanding of the biological systems and molecular mechanisms of disease development. Recently, deep learning (DL) algorithms have become one of the most promising methods in multi-omics data analysis, due to their predictive performance and capability of capturing nonlinear and hierarchical features. While integrating and translating multi-omics data into useful functional insights remain the biggest bottleneck, there is a clear trend towards incorporating multi-omics analysis in biomedical research to help explain the complex relationships between molecular layers. Multi-omics data have a role to improve prevention, early detection and prediction; monitor progression; interpret patterns and endotyping; and design personalized treatments. In this review, we outline a roadmap of multi-omics integration using DL and offer a practical perspective into the advantages, challenges and barriers to the implementation of DL in multi-omics data.
Collapse
Affiliation(s)
- Mingon Kang
- Department of Computer Science at the University of Nevada, Las Vegas, NV, USA
| | - Euiseong Ko
- Department of Computer Science at the University of Nevada, Las Vegas, NV, USA
| | - Tesfaye B Mersha
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|