1
|
Karz A, Coudray N, Bayraktar E, Galbraith K, Jour G, Shadaloey AAS, Eskow N, Rubanov A, Navarro M, Moubarak R, Baptiste G, Levinson G, Mezzano V, Alu M, Loomis C, Lima D, Rubens A, Jilaveanu L, Tsirigos A, Hernando E. MetFinder: A Tool for Automated Quantitation of Metastatic Burden in Histological Sections From Preclinical Models. Pigment Cell Melanoma Res 2024. [PMID: 39254030 DOI: 10.1111/pcmr.13195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024]
Abstract
As efforts to study the mechanisms of melanoma metastasis and novel therapeutic approaches multiply, researchers need accurate, high-throughput methods to evaluate the effects on tumor burden resulting from specific interventions. We show that automated quantification of tumor content from whole slide images is a compelling solution to assess in vivo experiments. In order to increase the outflow of data collection from preclinical studies, we assembled a large dataset with annotations and trained a deep neural network for the quantitative analysis of melanoma tumor content on histopathological sections of murine models. After assessing its performance in segmenting these images, the tool obtained consistent results with an orthogonal method (bioluminescence) of measuring metastasis in an experimental setting. This AI-based algorithm, made freely available to academic laboratories through a web-interface called MetFinder, promises to become an asset for melanoma researchers and pathologists interested in accurate, quantitative assessment of metastasis burden.
Collapse
Affiliation(s)
- Alcida Karz
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Nicolas Coudray
- Applied Bioinformatics Laboratories, NYU Langone Health, New York, New York, USA
- Department of Cell Biology, NYU School of Medicine, New York, New York, USA
| | - Erol Bayraktar
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Kristyn Galbraith
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
| | - George Jour
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
| | - Arman Alberto Sorin Shadaloey
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Nicole Eskow
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Andrey Rubanov
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Maya Navarro
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Rana Moubarak
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Gillian Baptiste
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Grace Levinson
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Valeria Mezzano
- Experimental Pathology Research Laboratory, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, New York, USA
| | - Mark Alu
- Experimental Pathology Research Laboratory, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, New York, USA
| | - Cynthia Loomis
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Experimental Pathology Research Laboratory, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, New York, USA
| | - Daniel Lima
- Research Software Engineering Core, Medical Center Information Technology Department, NYU Langone Health, New York, New York, USA
| | - Adam Rubens
- Research Software Engineering Core, Medical Center Information Technology Department, NYU Langone Health, New York, New York, USA
| | - Lucia Jilaveanu
- Department of Medicine, Yale University, New Haven, Connecticut, USA
| | - Aristotelis Tsirigos
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Applied Bioinformatics Laboratories, NYU Langone Health, New York, New York, USA
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| |
Collapse
|
2
|
Godson L, Alemi N, Nsengimana J, Cook GP, Clarke EL, Treanor D, Bishop DT, Newton-Bishop J, Gooya A, Magee D. Immune subtyping of melanoma whole slide images using multiple instance learning. Med Image Anal 2024; 93:103097. [PMID: 38325154 DOI: 10.1016/j.media.2024.103097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 02/09/2024]
Abstract
Determining early-stage prognostic markers and stratifying patients for effective treatment are two key challenges for improving outcomes for melanoma patients. Previous studies have used tumour transcriptome data to stratify patients into immune subgroups, which were associated with differential melanoma specific survival and potential predictive biomarkers. However, acquiring transcriptome data is a time-consuming and costly process. Moreover, it is not routinely used in the current clinical workflow. Here, we attempt to overcome this by developing deep learning models to classify gigapixel haematoxylin and eosin (H&E) stained pathology slides, which are well established in clinical workflows, into these immune subgroups. We systematically assess six different multiple instance learning (MIL) frameworks, using five different image resolutions and three different feature extraction methods. We show that pathology-specific self-supervised models using 10x resolution patches generate superior representations for the classification of immune subtypes. In addition, in a primary melanoma dataset, we achieve a mean area under the receiver operating characteristic curve (AUC) of 0.80 for classifying histopathology images into 'high' or 'low immune' subgroups and a mean AUC of 0.82 in an independent TCGA melanoma dataset. Furthermore, we show that these models are able to stratify patients into 'high' and 'low immune' subgroups with significantly different melanoma specific survival outcomes (log rank test, P< 0.005). We anticipate that MIL methods will allow us to find new biomarkers of high importance, act as a tool for clinicians to infer the immune landscape of tumours and stratify patients, without needing to carry out additional expensive genetic tests.
Collapse
Affiliation(s)
- Lucy Godson
- School of Computing, University of Leeds, Woodhouse, Leeds, LS2 9JT, United Kingdom.
| | - Navid Alemi
- School of Computing, University of Leeds, Woodhouse, Leeds, LS2 9JT, United Kingdom
| | - Jérémie Nsengimana
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE1 7RU, United Kingdom
| | - Graham P Cook
- Leeds Institute of Medical Research, University of Leeds School of Medicine, St. James's University Hospital, Leeds, United Kingdom
| | - Emily L Clarke
- Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom; Division of Pathology and Data Analytics, Leeds Institute of Cancer and Pathology, University of Leeds, Beckett Street, Leeds, LS9 7TF, United Kingdom
| | - Darren Treanor
- Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom; Division of Pathology and Data Analytics, Leeds Institute of Cancer and Pathology, University of Leeds, Beckett Street, Leeds, LS9 7TF, United Kingdom; Department of Clinical Pathology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - D Timothy Bishop
- Leeds Institute of Medical Research, University of Leeds School of Medicine, St. James's University Hospital, Leeds, United Kingdom
| | - Julia Newton-Bishop
- Division of Pathology and Data Analytics, Leeds Institute of Cancer and Pathology, University of Leeds, Beckett Street, Leeds, LS9 7TF, United Kingdom
| | - Ali Gooya
- School of Computing, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Derek Magee
- School of Computing, University of Leeds, Woodhouse, Leeds, LS2 9JT, United Kingdom
| |
Collapse
|
3
|
Yee J, Rosendahl C, Aoude LG. The role of artificial intelligence and convolutional neural networks in the management of melanoma: a clinical, pathological, and radiological perspective. Melanoma Res 2024; 34:96-104. [PMID: 38141179 PMCID: PMC10906187 DOI: 10.1097/cmr.0000000000000951] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/29/2023] [Indexed: 12/25/2023]
Abstract
Clinical dermatoscopy and pathological slide assessment are essential in the diagnosis and management of patients with cutaneous melanoma. For those presenting with stage IIC disease and beyond, radiological investigations are often considered. The dermatoscopic, whole slide and radiological images used during clinical care are often stored digitally, enabling artificial intelligence (AI) and convolutional neural networks (CNN) to learn, analyse and contribute to the clinical decision-making. A keyword search of the Medline database was performed to assess the progression, capabilities and limitations of AI and CNN and its use in diagnosis and management of cutaneous melanoma. Full-text articles were reviewed if they related to dermatoscopy, pathological slide assessment or radiology. Through analysis of 95 studies, we demonstrate that diagnostic accuracy of AI/CNN can be superior (or at least equal) to clinicians. However, variability in image acquisition, pre-processing, segmentation, and feature extraction remains challenging. With current technological abilities, AI/CNN and clinicians synergistically working together are better than one another in all subspecialty domains relating to cutaneous melanoma. AI has the potential to enhance the diagnostic capabilities of junior dermatology trainees, primary care skin cancer clinicians and general practitioners. For experienced clinicians, AI provides a cost-efficient second opinion. From a pathological and radiological perspective, CNN has the potential to improve workflow efficiency, allowing clinicians to achieve more in a finite amount of time. Until the challenges of AI/CNN are reliably met, however, they can only remain an adjunct to clinical decision-making.
Collapse
Affiliation(s)
- Joshua Yee
- Faculty of Medicine, University of Queensland, St Lucia
| | - Cliff Rosendahl
- Primary Care Clinical Unit, Medical School, The University of Queensland, Herston
| | - Lauren G. Aoude
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
| |
Collapse
|
4
|
Kehl A, Aupperle-Lellbach H, de Brot S, van der Weyden L. Review of Molecular Technologies for Investigating Canine Cancer. Animals (Basel) 2024; 14:769. [PMID: 38473154 PMCID: PMC10930838 DOI: 10.3390/ani14050769] [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: 12/21/2023] [Revised: 02/09/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Genetic molecular testing is starting to gain traction as part of standard clinical practice for dogs with cancer due to its multi-faceted benefits, such as potentially being able to provide diagnostic, prognostic and/or therapeutic information. However, the benefits and ultimate success of genomic analysis in the clinical setting are reliant on the robustness of the tools used to generate the results, which continually expand as new technologies are developed. To this end, we review the different materials from which tumour cells, DNA, RNA and the relevant proteins can be isolated and what methods are available for interrogating their molecular profile, including analysis of the genetic alterations (both somatic and germline), transcriptional changes and epigenetic modifications (including DNA methylation/acetylation and microRNAs). We also look to the future and the tools that are currently being developed, such as using artificial intelligence (AI) to identify genetic mutations from histomorphological criteria. In summary, we find that the molecular genetic characterisation of canine neoplasms has made a promising start. As we understand more of the genetics underlying these tumours and more targeted therapies become available, it will no doubt become a mainstay in the delivery of precision veterinary care to dogs with cancer.
Collapse
Affiliation(s)
- Alexandra Kehl
- Laboklin GmbH & Co. KG, Steubenstr. 4, 97688 Bad Kissingen, Germany; (A.K.); (H.A.-L.)
- School of Medicine, Institute of Pathology, Technical University of Munich, Trogerstr. 18, 81675 München, Germany
| | - Heike Aupperle-Lellbach
- Laboklin GmbH & Co. KG, Steubenstr. 4, 97688 Bad Kissingen, Germany; (A.K.); (H.A.-L.)
- School of Medicine, Institute of Pathology, Technical University of Munich, Trogerstr. 18, 81675 München, Germany
| | - Simone de Brot
- Institute of Animal Pathology, COMPATH, University of Bern, 3012 Bern, Switzerland;
| | | |
Collapse
|
5
|
Coudray N, Juarez MC, Criscito MC, Quiros AC, Wilken R, Cullison SRJ, Stevenson ML, Doudican NA, Yuan K, Aquino JD, Klufas DM, North JP, Yu SS, Murad F, Ruiz E, Schmults CD, Tsirigos A, Carucci JA. Self-supervised artificial intelligence predicts recurrence, metastasis and disease specific death from primary cutaneous squamous cell carcinoma at diagnosis. RESEARCH SQUARE 2023:rs.3.rs-3607399. [PMID: 38168253 PMCID: PMC10760225 DOI: 10.21203/rs.3.rs-3607399/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Primary cutaneous squamous cell carcinoma (cSCC) is responsible for ~10,000 deaths annually in the United States. Stratification of risk of poor outcome (PO) including recurrence, metastasis and disease specific death (DSD) at initial biopsy would significantly impact clinical decision-making during the initial post operative period where intervention has been shown to be most effective. In this multi-institutional study, we developed a state-of-the-art self-supervised deep-learning approach with interpretability power and demonstrated its ability to predict poor outcomes of cSCCs at the time of initial biopsy. By highlighting histomorphological phenotypes, our approach demonstrates that poor differentiation and deep invasion correlate with poor prognosis. Our approach is particularly efficient at defining poor outcome risk in Brigham and Women's Hospital (BWH) T2a and American Joint Committee on Cancer (AJCC) T2 cSCCs. This bridges a significant gap in our ability to assess risk among T2a/T2 cSCCs and may be useful in defining patients at highest risk of poor outcome at the time of diagnosis. Early identification of highest-risk patients could signal implementation of more stringent surveillance, rigorous diagnostic work up and identify patients who might best respond to early postoperative adjunctive treatment.
Collapse
Affiliation(s)
- Nicolas Coudray
- Applied Bioinformatics Laboratories, New York University School of Medicine, New York, NY, USA
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Michelle C. Juarez
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, USA
| | - Maressa C. Criscito
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, USA
| | | | - Reason Wilken
- Department of Dermatology, Northwell Health, New York, NY, USA
| | | | - Mary L. Stevenson
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, USA
| | - Nicole A. Doudican
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ke Yuan
- School of Computing Science, University of Glasgow, Glasgow, Scotland, UK (Ke Yuan)
- School of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK (Ke Yuan)
- Cancer Research UK Beatson Institute, Glasgow, Scotland, UK (Ke Yuan)
| | - Jamie D. Aquino
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel M. Klufas
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey P. North
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Siegrid S. Yu
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Fadi Murad
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily Ruiz
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Chrysalyne D. Schmults
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, New York University School of Medicine, New York, NY, USA
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - John A. Carucci
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
6
|
Adeuyan O, Gordon ER, Kenchappa D, Bracero Y, Singh A, Espinoza G, Geskin LJ, Saenger YM. An update on methods for detection of prognostic and predictive biomarkers in melanoma. Front Cell Dev Biol 2023; 11:1290696. [PMID: 37900283 PMCID: PMC10611507 DOI: 10.3389/fcell.2023.1290696] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/04/2023] [Indexed: 10/31/2023] Open
Abstract
The approval of immunotherapy for stage II-IV melanoma has underscored the need for improved immune-based predictive and prognostic biomarkers. For resectable stage II-III patients, adjuvant immunotherapy has proven clinical benefit, yet many patients experience significant adverse events and may not require therapy. In the metastatic setting, single agent immunotherapy cures many patients but, in some cases, more intensive combination therapies against specific molecular targets are required. Therefore, the establishment of additional biomarkers to determine a patient's disease outcome (i.e., prognostic) or response to treatment (i.e., predictive) is of utmost importance. Multiple methods ranging from gene expression profiling of bulk tissue, to spatial transcriptomics of single cells and artificial intelligence-based image analysis have been utilized to better characterize the immune microenvironment in melanoma to provide novel predictive and prognostic biomarkers. In this review, we will highlight the different techniques currently under investigation for the detection of prognostic and predictive immune biomarkers in melanoma.
Collapse
Affiliation(s)
- Oluwaseyi Adeuyan
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Emily R. Gordon
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Divya Kenchappa
- Albert Einstein College of Medicine, Bronx, NY, United States
| | - Yadriel Bracero
- Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ajay Singh
- Albert Einstein College of Medicine, Bronx, NY, United States
| | | | - Larisa J. Geskin
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, United States
| | | |
Collapse
|
7
|
Sauter D, Lodde G, Nensa F, Schadendorf D, Livingstone E, Kukuk M. Deep learning in computational dermatopathology of melanoma: A technical systematic literature review. Comput Biol Med 2023; 163:107083. [PMID: 37315382 DOI: 10.1016/j.compbiomed.2023.107083] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/10/2023] [Accepted: 05/27/2023] [Indexed: 06/16/2023]
Abstract
Deep learning (DL) has become one of the major approaches in computational dermatopathology, evidenced by a significant increase in this topic in the current literature. We aim to provide a structured and comprehensive overview of peer-reviewed publications on DL applied to dermatopathology focused on melanoma. In comparison to well-published DL methods on non-medical images (e.g., classification on ImageNet), this field of application comprises a specific set of challenges, such as staining artifacts, large gigapixel images, and various magnification levels. Thus, we are particularly interested in the pathology-specific technical state-of-the-art. We also aim to summarize the best performances achieved thus far with respect to accuracy, along with an overview of self-reported limitations. Accordingly, we conducted a systematic literature review of peer-reviewed journal and conference articles published between 2012 and 2022 in the databases ACM Digital Library, Embase, IEEE Xplore, PubMed, and Scopus, expanded by forward and backward searches to identify 495 potentially eligible studies. After screening for relevance and quality, a total of 54 studies were included. We qualitatively summarized and analyzed these studies from technical, problem-oriented, and task-oriented perspectives. Our findings suggest that the technical aspects of DL for histopathology in melanoma can be further improved. The DL methodology was adopted later in this field, and still lacks the wider adoption of DL methods already shown to be effective for other applications. We also discuss upcoming trends toward ImageNet-based feature extraction and larger models. While DL has achieved human-competitive accuracy in routine pathological tasks, its performance on advanced tasks is still inferior to wet-lab testing (for example). Finally, we discuss the challenges impeding the translation of DL methods to clinical practice and provide insight into future research directions.
Collapse
Affiliation(s)
- Daniel Sauter
- Department of Computer Science, Fachhochschule Dortmund, 44227 Dortmund, Germany.
| | - Georg Lodde
- Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | - Felix Nensa
- Institute for AI in Medicine (IKIM), University Hospital Essen, 45131 Essen, Germany; Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, 45147 Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | | | - Markus Kukuk
- Department of Computer Science, Fachhochschule Dortmund, 44227 Dortmund, Germany
| |
Collapse
|
8
|
Küchler L, Posthaus C, Jäger K, Guscetti F, van der Weyden L, von Bomhard W, Schmidt JM, Farra D, Aupperle-Lellbach H, Kehl A, Rottenberg S, de Brot S. Artificial Intelligence to Predict the BRAF V595E Mutation in Canine Urinary Bladder Urothelial Carcinomas. Animals (Basel) 2023; 13:2404. [PMID: 37570213 PMCID: PMC10416820 DOI: 10.3390/ani13152404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/10/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
In dogs, the BRAF mutation (V595E) is common in bladder and prostate cancer and represents a specific diagnostic marker. Recent advantages in artificial intelligence (AI) offer new opportunities in the field of tumour marker detection. While AI histology studies have been conducted in humans to detect BRAF mutation in cancer, comparable studies in animals are lacking. In this study, we used commercially available AI histology software to predict BRAF mutation in whole slide images (WSI) of bladder urothelial carcinomas (UC) stained with haematoxylin and eosin (HE), based on a training (n = 81) and a validation set (n = 96). Among 96 WSI, 57 showed identical PCR and AI-based BRAF predictions, resulting in a sensitivity of 58% and a specificity of 63%. The sensitivity increased substantially to 89% when excluding small or poor-quality tissue sections. Test reliability depended on tumour differentiation (p < 0.01), presence of inflammation (p < 0.01), slide quality (p < 0.02) and sample size (p < 0.02). Based on a small subset of cases with available adjacent non-neoplastic urothelium, AI was able to distinguish malignant from benign epithelium. This is the first study to demonstrate the use of AI histology to predict BRAF mutation status in canine UC. Despite certain limitations, the results highlight the potential of AI in predicting molecular alterations in routine tissue sections.
Collapse
Affiliation(s)
- Leonore Küchler
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (C.P.); (S.R.)
| | - Caroline Posthaus
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (C.P.); (S.R.)
| | - Kathrin Jäger
- Laboklin GmbH & Co. KG, 97688 Bad Kissingen, Germany; (K.J.); (H.A.-L.); (A.K.)
- Institute of Pathology, Department of Comparative Experimental Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Franco Guscetti
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland;
| | | | | | | | - Dima Farra
- Veterinary Public Health Institute, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland;
| | - Heike Aupperle-Lellbach
- Laboklin GmbH & Co. KG, 97688 Bad Kissingen, Germany; (K.J.); (H.A.-L.); (A.K.)
- Institute of Pathology, Department of Comparative Experimental Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Alexandra Kehl
- Laboklin GmbH & Co. KG, 97688 Bad Kissingen, Germany; (K.J.); (H.A.-L.); (A.K.)
- Institute of Pathology, Department of Comparative Experimental Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (C.P.); (S.R.)
- COMPATH, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, 3008 Bern, Switzerland
| | - Simone de Brot
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (C.P.); (S.R.)
- COMPATH, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
9
|
Doeleman T, Hondelink LM, Vermeer MH, van Dijk MR, Schrader AMR. Artificial intelligence in digital pathology of cutaneous lymphomas: a review of the current state and future perspectives. Semin Cancer Biol 2023:S1044-579X(23)00095-0. [PMID: 37331571 DOI: 10.1016/j.semcancer.2023.06.004] [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: 12/09/2022] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/20/2023]
Abstract
Primary cutaneous lymphomas (CLs) represent a heterogeneous group of T-cell lymphomas and B-cell lymphomas that present in the skin without evidence of extracutaneous involvement at time of diagnosis. CLs are largely distinct from their systemic counterparts in clinical presentation, histopathology, and biological behavior and, therefore, require different therapeutic management. Additional diagnostic burden is added by the fact that several benign inflammatory dermatoses mimic CL subtypes, requiring clinicopathological correlation for definitive diagnosis. Due to the heterogeneity and rarity of CL, adjunct diagnostic tools are welcomed, especially by pathologists without expertise in this field or with limited access to a centralized specialist panel. The transition into digital pathology workflows enables artificial intelligence (AI)-based analysis of patients' whole-slide pathology images (WSIs). AI can be used to automate manual processes in histopathology but, more importantly, can be applied to complex diagnostic tasks, especially suitable for rare disease like CL. To date, AI-based applications for CL have been minimally explored in literature. However, in other skin cancers and systemic lymphomas, disciplines that are recognized here as the building blocks for CLs, several studies demonstrated promising results using AI for disease diagnosis and subclassification, cancer detection, specimen triaging, and outcome prediction. Additionally, AI allows discovery of novel biomarkers or may help to quantify established biomarkers. This review summarizes and blends applications of AI in pathology of skin cancer and lymphoma and proposes how these findings can be applied to diagnostics of CL.
Collapse
Affiliation(s)
- Thom Doeleman
- Department of Pathology, Leiden University Medical Centre, Leiden, the Netherlands; Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Liesbeth M Hondelink
- Department of Pathology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Maarten H Vermeer
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marijke R van Dijk
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Anne M R Schrader
- Department of Pathology, Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
10
|
Grossarth S, Mosley D, Madden C, Ike J, Smith I, Huo Y, Wheless L. Recent Advances in Melanoma Diagnosis and Prognosis Using Machine Learning Methods. Curr Oncol Rep 2023; 25:635-645. [PMID: 37000340 PMCID: PMC10339689 DOI: 10.1007/s11912-023-01407-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 04/01/2023]
Abstract
PURPOSE OF REVIEW The purpose was to summarize the current role and state of artificial intelligence and machine learning in the diagnosis and management of melanoma. RECENT FINDINGS Deep learning algorithms can identify melanoma from clinical, dermoscopic, and whole slide pathology images with increasing accuracy. Efforts to provide more granular annotation to datasets and to identify new predictors are ongoing. There have been many incremental advances in both melanoma diagnostics and prognostic tools using artificial intelligence and machine learning. Higher quality input data will further improve these models' capabilities.
Collapse
Affiliation(s)
- Sarah Grossarth
- Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | | | - Christopher Madden
- Department of Dermatology, Vanderbilt University Medicine Center, Nashville, TN, USA
- State University of New York Downstate College of Medicine, Brooklyn, NY, USA
| | - Jacqueline Ike
- Department of Dermatology, Vanderbilt University Medicine Center, Nashville, TN, USA
- Meharry Medical College, Nashville, TN, USA
| | - Isabelle Smith
- Department of Dermatology, Vanderbilt University Medicine Center, Nashville, TN, USA
- Vanderbilt University, Nashville, TN, USA
| | - Yuankai Huo
- Department of Computer Science and Electrical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Lee Wheless
- Department of Dermatology, Vanderbilt University Medicine Center, Nashville, TN, USA.
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Tennessee Valley Healthcare System VA Medical Center, Nashville, TN, USA.
| |
Collapse
|
11
|
Schneider L, Wies C, Krieghoff-Henning EI, Bucher TC, Utikal JS, Schadendorf D, Brinker TJ. Multimodal integration of image, epigenetic and clinical data to predict BRAF mutation status in melanoma. Eur J Cancer 2023; 183:131-138. [PMID: 36854237 DOI: 10.1016/j.ejca.2023.01.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND In machine learning, multimodal classifiers can provide more generalised performance than unimodal classifiers. In clinical practice, physicians usually also rely on a range of information from different examinations for diagnosis. In this study, we used BRAF mutation status prediction in melanoma as a model system to analyse the contribution of different data types in a combined classifier because BRAF status can be determined accurately by sequencing as the current gold standard, thus nearly eliminating label noise. METHODS We trained a deep learning-based classifier by combining individually trained random forests of image, clinical and methylation data to predict BRAF-V600 mutation status in primary and metastatic melanomas of The Cancer Genome Atlas cohort. RESULTS With our multimodal approach, we achieved an area under the receiver operating characteristic curve of 0.80, whereas the individual classifiers yielded areas under the receiver operating characteristic curve of 0.63 (histopathologic image data), 0.66 (clinical data) and 0.66 (methylation data) on an independent data set. CONCLUSIONS Our combined approach can predict BRAF status to some extent by identifying BRAF-V600 specific patterns at the histologic, clinical and epigenetic levels. The multimodal classifiers have improved generalisability in predicting BRAF mutation status.
Collapse
Affiliation(s)
- Lucas Schneider
- Digital Biomarkers for Oncology Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Christoph Wies
- Digital Biomarkers for Oncology Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Eva I Krieghoff-Henning
- Digital Biomarkers for Oncology Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Tabea-Clara Bucher
- Digital Biomarkers for Oncology Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Jochen S Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| |
Collapse
|
12
|
Ghaffari Laleh N, Ligero M, Perez-Lopez R, Kather JN. Facts and Hopes on the Use of Artificial Intelligence for Predictive Immunotherapy Biomarkers in Cancer. Clin Cancer Res 2023; 29:316-323. [PMID: 36083132 DOI: 10.1158/1078-0432.ccr-22-0390] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/26/2022] [Accepted: 08/29/2022] [Indexed: 01/19/2023]
Abstract
Immunotherapy by immune checkpoint inhibitors has become a standard treatment strategy for many types of solid tumors. However, the majority of patients with cancer will not respond, and predicting response to this therapy is still a challenge. Artificial intelligence (AI) methods can extract meaningful information from complex data, such as image data. In clinical routine, radiology or histopathology images are ubiquitously available. AI has been used to predict the response to immunotherapy from radiology or histopathology images, either directly or indirectly via surrogate markers. While none of these methods are currently used in clinical routine, academic and commercial developments are pointing toward potential clinical adoption in the near future. Here, we summarize the state of the art in AI-based image biomarkers for immunotherapy response based on radiology and histopathology images. We point out limitations, caveats, and pitfalls, including biases, generalizability, and explainability, which are relevant for researchers and health care providers alike, and outline key clinical use cases of this new class of predictive biomarkers.
Collapse
Affiliation(s)
| | - Marta Ligero
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Department of Radiology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Jakob Nikolas 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, United Kingdom.,Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.,Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| |
Collapse
|
13
|
Deep Learning for Skin Melanocytic Tumors in Whole-Slide Images: A Systematic Review. Cancers (Basel) 2022; 15:cancers15010042. [PMID: 36612037 PMCID: PMC9817526 DOI: 10.3390/cancers15010042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/05/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The rise of Artificial Intelligence (AI) has shown promising performance as a support tool in clinical pathology workflows. In addition to the well-known interobserver variability between dermatopathologists, melanomas present a significant challenge in their histological interpretation. This study aims to analyze all previously published studies on whole-slide images of melanocytic tumors that rely on deep learning techniques for automatic image analysis. Embase, Pubmed, Web of Science, and Virtual Health Library were used to search for relevant studies for the systematic review, in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist. Articles from 2015 to July 2022 were included, with an emphasis placed on the used artificial intelligence methods. Twenty-eight studies that fulfilled the inclusion criteria were grouped into four groups based on their clinical objectives, including pathologists versus deep learning models (n = 10), diagnostic prediction (n = 7); prognosis (n = 5), and histological features (n = 6). These were then analyzed to draw conclusions on the general parameters and conditions of AI in pathology, as well as the necessary factors for better performance in real scenarios.
Collapse
|
14
|
Diagnostic and Prognostic Deep Learning Applications for Histological Assessment of Cutaneous Melanoma. Cancers (Basel) 2022; 14:cancers14246231. [PMID: 36551716 PMCID: PMC9776963 DOI: 10.3390/cancers14246231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Melanoma is among the most devastating human malignancies. Accurate diagnosis and prognosis are essential to offer optimal treatment. Histopathology is the gold standard for establishing melanoma diagnosis and prognostic features. However, discrepancies often exist between pathologists, and analysis is costly and time-consuming. Deep-learning algorithms are deployed to improve melanoma diagnosis and prognostication from histological images of melanoma. In recent years, the development of these machine-learning tools has accelerated, and machine learning is poised to become a clinical tool to aid melanoma histology. Nevertheless, a review of the advances in machine learning in melanoma histology was lacking. We performed a comprehensive literature search to provide a complete overview of the recent advances in machine learning in the assessment of melanoma based on hematoxylin eosin digital pathology images. In our work, we review 37 recent publications, compare the methods and performance of the reviewed studies, and highlight the variety of promising machine-learning applications in melanoma histology.
Collapse
|
15
|
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
|
16
|
Hong R, Fenyö D. When blockchain meets artificial intelligence: An application to cancer histopathology. Cell Rep Med 2022; 3:100666. [PMID: 35732149 PMCID: PMC9245033 DOI: 10.1016/j.xcrm.2022.100666] [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] [Indexed: 11/05/2022]
Abstract
A recent study by Saldanha et al. demonstrates that blockchain-based models outcompeted local models and performed similarly with merged models to predict molecular features from cancer histopathology images. The results reveal the capability of decentralized models in molecular diagnosis of cancer.
Collapse
Affiliation(s)
- Runyu Hong
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
17
|
Giannikopoulos P, Parham DM. Pediatric Sarcomas: The Next Generation of Molecular Studies. Cancers (Basel) 2022; 14:2515. [PMID: 35626119 PMCID: PMC9139929 DOI: 10.3390/cancers14102515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Pediatric sarcomas constitute one of the largest groups of childhood cancers, following hematopoietic, neural, and renal lesions. Partly because of their diversity, they continue to offer challenges in diagnosis and treatment. In spite of the diagnostic, nosologic, and therapeutic gains made with genetic technology, newer means for investigation are needed. This article reviews emerging technology being used to study human neoplasia and how these methods might be applicable to pediatric sarcomas. Methods reviewed include single cell RNA sequencing (scRNAseq), spatial multi-omics, high-throughput functional genomics, and clustered regularly interspersed short palindromic sequence-Cas9 (CRISPR-Cas9) technology. In spite of these advances, the field continues to be challenged by a dearth of properly annotated materials, particularly from recurrences and metastases and pre- and post-treatment samples.
Collapse
Affiliation(s)
| | - David M. Parham
- Department of Anatomic Pathology, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Pathology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| |
Collapse
|