51
|
Sun P, Fan S, Li S, Zhao Y, Lu C, Wong KC, Li X. Automated exploitation of deep learning for cancer patient stratification across multiple types. Bioinformatics 2023; 39:btad654. [PMID: 37934154 PMCID: PMC10636288 DOI: 10.1093/bioinformatics/btad654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 10/17/2023] [Indexed: 11/08/2023] Open
Abstract
MOTIVATION Recent frameworks based on deep learning have been developed to identify cancer subtypes from high-throughput gene expression profiles. Unfortunately, the performance of deep learning is highly dependent on its neural network architectures which are often hand-crafted with expertise in deep neural networks, meanwhile, the optimization and adjustment of the network are usually costly and time consuming. RESULTS To address such limitations, we proposed a fully automated deep neural architecture search model for diagnosing consensus molecular subtypes from gene expression data (DNAS). The proposed model uses ant colony algorithm, one of the heuristic swarm intelligence algorithms, to search and optimize neural network architecture, and it can automatically find the optimal deep learning model architecture for cancer diagnosis in its search space. We validated DNAS on eight colorectal cancer datasets, achieving the average accuracy of 95.48%, the average specificity of 98.07%, and the average sensitivity of 96.24%, respectively. Without the loss of generality, we investigated the general applicability of DNAS further on other cancer types from different platforms including lung cancer and breast cancer, and DNAS achieved an area under the curve of 95% and 96%, respectively. In addition, we conducted gene ontology enrichment and pathological analysis to reveal interesting insights into cancer subtype identification and characterization across multiple cancer types. AVAILABILITY AND IMPLEMENTATION The source code and data can be downloaded from https://github.com/userd113/DNAS-main. And the web server of DNAS is publicly accessible at 119.45.145.120:5001.
Collapse
Affiliation(s)
- Pingping Sun
- School of Information Science and Technology, Northeast Normal University, Jilin, China
| | - Shijie Fan
- School of Information Science and Technology, Northeast Normal University, Jilin, China
| | - Shaochuan Li
- School of Information Science and Technology, Northeast Normal University, Jilin, China
- School of Artificial Intelligence, Jilin University, Jilin, China
| | - Yingwei Zhao
- School of Information Science and Technology, Northeast Normal University, Jilin, China
| | - Chang Lu
- School of Information Science and Technology, Northeast Normal University, Jilin, China
- School of Psychology, Northeast Normal University, Jilin, China
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Hong Kong China
| | - Xiangtao Li
- School of Artificial Intelligence, Jilin University, Jilin, China
| |
Collapse
|
52
|
Jafri HSMO, Mushtaq S, Baig S, Bhatty A, Siraj S. Comparison of KRAS gene in circulating tumor DNA levels vs histological grading of colorectal cancer patients through liquid biopsy. Saudi J Gastroenterol 2023; 29:371-375. [PMID: 37602638 PMCID: PMC10754382 DOI: 10.4103/sjg.sjg_85_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/15/2023] [Accepted: 07/04/2023] [Indexed: 08/22/2023] Open
Abstract
Background To determine KRAS gene in circulating tumor DNA in comparison with histological grading through liquid biopsy in colorectal cancer patients. Methods This dual-centered cross-sectional study included 73 diagnosed patients of colorectal cancer at different grading levels [Grade I, well differentiated (n = 7, 9.5%); Grade II, moderately differentiated (n = 14,18.9%); and Grade III, poorly differentiated (n = 52, 70%)]. Blood was collected, and plasma was separated. ctDNA was extracted, using magnetic bead-based technique (MagMAX Cell-Free DNA kit). KRAS gene was quantified through qPCR. STRING database was used to find KRAS interactomes. Results Mean threshold cycle (CT value) of KRAS gene in Grade III samples showed significantly higher (P = 0.001) levels of ctDNA (2.7 ± 1.14) compared with Grade II and Grade I (3.1 ± 0.68, 2.3 ± 0.60), respectively. Grading characterization showed that rectal cancer (n = 22, 42.3%) with Grade III (68.8%) was more prevalent than colon and sigmoid cancer (n = 19, 36.5%, n = 11, 21%, respectively). STRING database showed 10 functional genes interacting with KRAS expressed as gene/proteins. Conclusion Liquid biopsy can be used to detect ctDNA in plasma of CRC patients and enabled to detect the KRAS gene by qPCR. The technique being less invasive and cost-effective is convenient for multiple biopsies in different cancers.
Collapse
Affiliation(s)
| | - Shamim Mushtaq
- Department of Biochemistry, Basic Health Sciences, Ziauddin University, Karachi, Pakistan
| | - Saeeda Baig
- Department of Biochemistry, Basic Health Sciences, Ziauddin University, Karachi, Pakistan
| | - Afreen Bhatty
- Department of Biochemistry, Basic Health Sciences, Ziauddin University, Karachi, Pakistan
| | - Sabra Siraj
- Department of Pharmacology, Basic Health Sciences, Ziauddin University, Karachi, Pakistan
| |
Collapse
|
53
|
Shi T, Li M, Yu Y. Machine learning-enhanced insights into sphingolipid-based prognostication: revealing the immunological landscape and predictive proficiency for immunomotherapy and chemotherapy responses in pancreatic carcinoma. Front Mol Biosci 2023; 10:1284623. [PMID: 38028544 PMCID: PMC10643633 DOI: 10.3389/fmolb.2023.1284623] [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: 08/28/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Background: With a poor prognosis for affected individuals, pancreatic adenocarcinoma (PAAD) is known as a complicated and diverse illness. Immunocytes have become essential elements in the development of PAAD. Notably, sphingolipid metabolism has a dual function in the development of tumors and the invasion of the immune system. Despite these implications, research on the predictive ability of sphingolipid variables for PAAD prognosis is strikingly lacking, and it is yet unclear how they can affect PAAD immunotherapy and targeted pharmacotherapy. Methods: The investigation process included SPG detection while also being pertinent to the prognosis for PAAD. Both the analytical capability of CIBERSORT and the prognostic capability of the pRRophetic R package were used to evaluate the immunological environments of the various HCC subtypes. In addition, CCK-8 experiments on PAAD cell lines were carried out to confirm the accuracy of drug sensitivity estimates. The results of these trials, which also evaluated cell survival and migratory patterns, confirmed the usefulness of sphingolipid-associated genes (SPGs). Results: As a result of this thorough investigation, 32 SPGs were identified, each of which had a measurable influence on the dynamics of overall survival. This collection of genes served as the conceptual framework for the development of a prognostic model, which was carefully assembled from 10 chosen genes. It should be noted that this grouping of patients into cohorts with high and low risk was a sign of different immune profiles and therapy responses. The increased abundance of SPGs was identified as a possible sign of inadequate responses to immune-based treatment approaches. The careful CCK-8 testing carried out on PAAD cell lines was of the highest importance for providing clear confirmation of drug sensitivity estimates. Conclusion: The significance of Sphingolipid metabolism in the complex web of PAAD development is brought home by this study. The novel risk model, built on the complexity of sphingolipid-associated genes, advances our understanding of PAAD and offers doctors a powerful tool for developing personalised treatment plans that are specifically suited to the unique characteristics of each patient.
Collapse
Affiliation(s)
| | | | - Yabin Yu
- Department of Hepatobiliary Surgery, The Affiliated Huaian No 1 People’s Hospital of Nanjing Medical University, Huaian, China
| |
Collapse
|
54
|
Sehring J, Dohmen H, Selignow C, Schmid K, Grau S, Stein M, Uhl E, Mukhopadhyay A, Németh A, Amsel D, Acker T. Leveraging Attention-Based Convolutional Neural Networks for Meningioma Classification in Computational Histopathology. Cancers (Basel) 2023; 15:5190. [PMID: 37958364 PMCID: PMC10647687 DOI: 10.3390/cancers15215190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Convolutional neural networks (CNNs) are becoming increasingly valuable tools for advanced computational histopathology, promoting precision medicine through exceptional visual decoding abilities. Meningiomas, the most prevalent primary intracranial tumors, necessitate accurate grading and classification for informed clinical decision-making. Recently, DNA methylation-based molecular classification of meningiomas has proven to be more effective in predicting tumor recurrence than traditional histopathological methods. However, DNA methylation profiling is expensive, labor-intensive, and not widely accessible. Consequently, a digital histology-based prediction of DNA methylation classes would be advantageous, complementing molecular classification. In this study, we developed and rigorously assessed an attention-based multiple-instance deep neural network for predicting meningioma methylation classes using tumor methylome data from 142 (+51) patients and corresponding hematoxylin-eosin-stained histological sections. Pairwise analysis of sample cohorts from three meningioma methylation classes demonstrated high accuracy in two combinations. The performance of our approach was validated using an independent set of 51 meningioma patient samples. Importantly, attention map visualization revealed that the algorithm primarily focuses on tumor regions deemed significant by neuropathologists, offering insights into the decision-making process of the CNN. Our findings highlight the capacity of CNNs to effectively harness phenotypic information from histological sections through computerized images for precision medicine. Notably, this study is the first demonstration of predicting clinically relevant DNA methylome information using computer vision applied to standard histopathology. The introduced AI framework holds great potential in supporting, augmenting, and expediting meningioma classification in the future.
Collapse
Affiliation(s)
- Jannik Sehring
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Hildegard Dohmen
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Carmen Selignow
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Kai Schmid
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Stefan Grau
- Department of Neurosurgery, Hospital Fulda, Pacelliallee 4, D-36043 Fulda, Germany
| | - Marco Stein
- Department of Neurosurgery, University Hospital Gießen, Klinikstr. 33, D-35392 Giessen, Germany
| | - Eberhard Uhl
- Department of Neurosurgery, University Hospital Gießen, Klinikstr. 33, D-35392 Giessen, Germany
| | - Anirban Mukhopadhyay
- Department of Computer Science, Technical University of Darmstadt, Fraunhoferstraße 5, D-64283 Darmstadt, Germany
| | - Attila Németh
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Daniel Amsel
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Till Acker
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| |
Collapse
|
55
|
Qi L, Liang JY, Li ZW, Xi SY, Lai YN, Gao F, Zhang XR, Wang DS, Hu MT, Cao Y, Xu LJ, Chan RC, Xing BC, Wang X, Li YH. Deep learning-derived spatial organization features on histology images predicts prognosis in colorectal liver metastasis patients after hepatectomy. iScience 2023; 26:107702. [PMID: 37701575 PMCID: PMC10494211 DOI: 10.1016/j.isci.2023.107702] [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: 03/07/2023] [Revised: 07/10/2023] [Accepted: 08/21/2023] [Indexed: 09/14/2023] Open
Abstract
Histopathological images of colorectal liver metastases (CRLM) contain rich morphometric information that may predict patients' outcomes. However, to our knowledge, no study has reported any practical deep learning framework based on the histology images of CRLM, and their direct association with prognosis remains largely unknown. In this study, we developed a deep learning-based framework for fully automated tissue classification and quantification of clinically relevant spatial organization features (SOFs) in H&E-stained images of CRLM. The SOFs based risk-scoring system demonstrated a strong and robust prognostic value that is independent of the current clinical risk score (CRS) system in independent clinical cohorts. Our framework enables fully automated tissue classification of H&E images of CRLM, which could significantly reduce assessment subjectivity and the workload of pathologists. The risk-scoring system provides a time- and cost-efficient tool to assist clinical decision-making for patients with CRLM, which could potentially be implemented in clinical practice.
Collapse
Affiliation(s)
- Lin Qi
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - Jie-ying Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhong-wu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shao-yan Xi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yu-ni Lai
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Feng Gao
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xian-rui Zhang
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - De-shen Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ming-tao Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yi Cao
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - Li-jian Xu
- Centre for Perceptual and Interactive Intelligence, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ronald C.K. Chan
- Department of Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bao-cai Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xin Wang
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - Yu-hong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
56
|
Eminaga O, Leyh-Bannurah SR, Shariat SF, Krabbe LM, Lau H, Xing L, Abbas M. Artificial Intelligence Reveals Distinct Prognostic Subgroups of Muscle-Invasive Bladder Cancer on Histology Images. Cancers (Basel) 2023; 15:4998. [PMID: 37894365 PMCID: PMC10605516 DOI: 10.3390/cancers15204998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/12/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Muscle-invasive bladder cancer (MIBC) is a highly heterogeneous and costly disease with significant morbidity and mortality. Understanding tumor histopathology leads to tailored therapies and improved outcomes. In this study, we employed a weakly supervised learning and neural architecture search to develop a data-driven scoring system. This system aimed to capture prognostic histopathological patterns observed in H&E-stained whole-slide images. We constructed and externally validated our scoring system using multi-institutional datasets with 653 whole-slide images. Additionally, we explored the association between our scoring system, seven histopathological features, and 126 molecular signatures. Through our analysis, we identified two distinct risk groups with varying prognoses, reflecting inherent differences in histopathological and molecular subtypes. The adjusted hazard ratio for overall mortality was 1.46 (95% CI 1.05-2.02; z: 2.23; p = 0.03), thus identifying two prognostic subgroups in high-grade MIBC. Furthermore, we observed an association between our novel digital biomarker and the squamous phenotype, subtypes of miRNA, mRNA, long non-coding RNA, DNA hypomethylation, and several gene mutations, including FGFR3 in MIBC. Our findings underscore the risk of confounding bias when reducing the complex biological and clinical behavior of tumors to a single mutation. Histopathological changes can only be fully captured through comprehensive multi-omics profiles. The introduction of our scoring system has the potential to enhance daily clinical decision making for MIBC. It facilitates shared decision making by offering comprehensive and precise risk stratification, treatment planning, and cost-effective preselection for expensive molecular characterization.
Collapse
Affiliation(s)
| | - Sami-Ramzi Leyh-Bannurah
- Department of Urology, Pediatric Urology and Uro-Oncology, Prostate Center Northwest, St. Antonius-Hospital, 33705 Gronau, Germany
| | - Shahrokh F. Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria;
| | - Laura-Maria Krabbe
- Department of Urology, University Hospital of Muenster, 48419 Muenster, Germany
| | - Hubert Lau
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA;
- Department of Pathology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Lei Xing
- Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mahmoud Abbas
- Department of Pathology, University Hospital of Muenster, 48419 Muenster, Germany
| |
Collapse
|
57
|
Farin HF, Mosa MH, Ndreshkjana B, Grebbin BM, Ritter B, Menche C, Kennel KB, Ziegler PK, Szabó L, Bollrath J, Rieder D, Michels BE, Kress A, Bozlar M, Darvishi T, Stier S, Kur IM, Bankov K, Kesselring R, Fichtner-Feigl S, Brüne B, Goetze TO, Al-Batran SE, Brandts CH, Bechstein WO, Wild PJ, Weigert A, Müller S, Knapp S, Trajanoski Z, Greten FR. Colorectal Cancer Organoid-Stroma Biobank Allows Subtype-Specific Assessment of Individualized Therapy Responses. Cancer Discov 2023; 13:2192-2211. [PMID: 37489084 PMCID: PMC10551667 DOI: 10.1158/2159-8290.cd-23-0050] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/05/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
In colorectal cancers, the tumor microenvironment plays a key role in prognosis and therapy efficacy. Patient-derived tumor organoids (PDTO) show enormous potential for preclinical testing; however, cultured tumor cells lose important characteristics, including the consensus molecular subtypes (CMS). To better reflect the cellular heterogeneity, we established the colorectal cancer organoid-stroma biobank of matched PDTOs and cancer-associated fibroblasts (CAF) from 30 patients. Context-specific phenotyping showed that xenotransplantation or coculture with CAFs improves the transcriptomic fidelity and instructs subtype-specific stromal gene expression. Furthermore, functional profiling in coculture exposed CMS4-specific therapeutic resistance to gefitinib and SN-38 and prognostic expression signatures. Chemogenomic library screening identified patient- and therapy-dependent mechanisms of stromal resistance including MET as a common target. Our results demonstrate that colorectal cancer phenotypes are encrypted in the cancer epithelium in a plastic fashion that strongly depends on the context. Consequently, CAFs are essential for a faithful representation of molecular subtypes and therapy responses ex vivo. SIGNIFICANCE Systematic characterization of the organoid-stroma biobank provides a resource for context dependency in colorectal cancer. We demonstrate a colorectal cancer subtype memory of PDTOs that is independent of specific driver mutations. Our data underscore the importance of functional profiling in cocultures for improved preclinical testing and identification of stromal resistance mechanisms. This article is featured in Selected Articles from This Issue, p. 2109.
Collapse
Affiliation(s)
- Henner F. Farin
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mohammed H. Mosa
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Benardina Ndreshkjana
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Britta M. Grebbin
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Birgit Ritter
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Constantin Menche
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Kilian B. Kennel
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Paul K. Ziegler
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Lili Szabó
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Julia Bollrath
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Dietmar Rieder
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Birgitta E. Michels
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Alena Kress
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Müge Bozlar
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Tahmineh Darvishi
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Sara Stier
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Ivan-Maximilano Kur
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- Institute of Biochemistry I, Goethe University, Frankfurt am Main, Germany
| | - Katrin Bankov
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Rebecca Kesselring
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of General and Visceral Surgery, University of Freiburg, Freiburg, Germany
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, University of Freiburg, Freiburg, Germany
| | - Bernhard Brüne
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Biochemistry I, Goethe University, Frankfurt am Main, Germany
| | | | | | - Christian H. Brandts
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Wolf O. Bechstein
- Department of General and Visceral Surgery, Goethe University, Frankfurt am Main, Germany
| | - Peter J. Wild
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main, Germany
| | - Andreas Weigert
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Biochemistry I, Goethe University, Frankfurt am Main, Germany
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany
- Structural Genomics Consortium, Goethe University, Frankfurt am Main, Germany
| | - Stefan Knapp
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany
- Structural Genomics Consortium, Goethe University, Frankfurt am Main, Germany
| | - Zlatko Trajanoski
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian R. Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
58
|
Prezja F, Äyrämö S, Pölönen I, Ojala T, Lahtinen S, Ruusuvuori P, Kuopio T. Improved accuracy in colorectal cancer tissue decomposition through refinement of established deep learning solutions. Sci Rep 2023; 13:15879. [PMID: 37741820 PMCID: PMC10517936 DOI: 10.1038/s41598-023-42357-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023] Open
Abstract
Hematoxylin and eosin-stained biopsy slides are regularly available for colorectal cancer patients. These slides are often not used to define objective biomarkers for patient stratification and treatment selection. Standard biomarkers often pertain to costly and slow genetic tests. However, recent work has shown that relevant biomarkers can be extracted from these images using convolutional neural networks (CNNs). The CNN-based biomarkers predicted colorectal cancer patient outcomes comparably to gold standards. Extracting CNN-biomarkers is fast, automatic, and of minimal cost. CNN-based biomarkers rely on the ability of CNNs to recognize distinct tissue types from microscope whole slide images. The quality of these biomarkers (coined 'Deep Stroma') depends on the accuracy of CNNs in decomposing all relevant tissue classes. Improving tissue decomposition accuracy is essential for improving the prognostic potential of CNN-biomarkers. In this study, we implemented a novel training strategy to refine an established CNN model, which then surpassed all previous solutions . We obtained a 95.6% average accuracy in the external test set and 99.5% in the internal test set. Our approach reduced errors in biomarker-relevant classes, such as Lymphocytes, and was the first to include interpretability methods. These methods were used to better apprehend our model's limitations and capabilities.
Collapse
Affiliation(s)
- Fabi Prezja
- Faculty of Information Technology, University of Jyväskylä, Jyväskylä, 40014, Finland.
- Digital Health Intelligence Laboratory, University of Jyväskylä, Jyväskylä, 40014, Finland.
| | - Sami Äyrämö
- Faculty of Information Technology, University of Jyväskylä, Jyväskylä, 40014, Finland
- Digital Health Intelligence Laboratory, University of Jyväskylä, Jyväskylä, 40014, Finland
| | - Ilkka Pölönen
- Faculty of Information Technology, University of Jyväskylä, Jyväskylä, 40014, Finland
- Spectral Imaging Laboratory, University of Jyväskylä, Jyväskylä, 40014, Finland
| | - Timo Ojala
- Faculty of Information Technology, University of Jyväskylä, Jyväskylä, 40014, Finland
- Digital Health Intelligence Laboratory, University of Jyväskylä, Jyväskylä, 40014, Finland
| | - Suvi Lahtinen
- Faculty of Information Technology, University of Jyväskylä, Jyväskylä, 40014, Finland
- Department of Biological and Environmental Science, Faculty of Mathematics and Science, University of Jyväskylä, Jyväskylä, 40014, Finland
| | - Pekka Ruusuvuori
- Institute of Biomedicine, Cancer Research Unit, University of Turku, Turku, 20014, Finland
- FICAN West Cancer Centre, Turku University Hospital, Turku, 20521, Finland
| | - Teijo Kuopio
- Department of Education and Research, Hospital Nova of Central Finland, Jyväskylä, 40620, Finland
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, 40014, Finland
- Department of Pathology, Hospital Nova of Central Finland, Jyväskylä, 40620, Finland
| |
Collapse
|
59
|
Zhao Q, Li H, Li W, Guo Z, Jia W, Xu S, Chen S, Shen X, Wang C. Identification and verification of a prognostic signature based on a miRNA-mRNA interaction pattern in colon adenocarcinoma. Front Cell Dev Biol 2023; 11:1161667. [PMID: 37745305 PMCID: PMC10511881 DOI: 10.3389/fcell.2023.1161667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 08/04/2023] [Indexed: 09/26/2023] Open
Abstract
The expression characteristics of non-coding RNA (ncRNA) in colon adenocarcinoma (COAD) are involved in regulating various biological processes. To achieve these functions, ncRNA and a member of the Argonaute protein family form an RNA-induced silencing complex (RISC). The RISC is directed by ncRNA, especially microRNA (miRNA), to bind the target complementary mRNAs and regulate their expression by interfering with mRNA cleavage, degradation, or translation. However, how to identify potential miRNA biomarkers and therapeutic targets remains unclear. Here, we performed differential gene screening based on The Cancer Genome Atlas dataset and annotated meaningful differential genes to enrich related biological processes and regulatory cancer pathways. According to the overlap between the screened differential mRNAs and differential miRNAs, a prognosis model based on a least absolute shrinkage and selection operator-based Cox proportional hazards regression analysis can be established to obtain better prognosis characteristics. To further explore the therapeutic potential of miRNA as a target of mRNA intervention, we conducted an immunohistochemical analysis and evaluated the expression level in the tissue microarray of 100 colorectal cancer patients. The results demonstrated that the expression level of POU4F1, DNASE1L2, and WDR72 in the signature was significantly upregulated in COAD and correlated with poor prognosis. Establishing a prognostic signature based on miRNA target genes will help elucidate the molecular pathogenesis of COAD and provide novel potential targets for RNA therapy.
Collapse
Affiliation(s)
- Qiwu Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haosheng Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenchang Li
- Department of Interventional Radiography, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zichao Guo
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqing Jia
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuiyu Xu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sixia Chen
- Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Xiaonan Shen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changgang Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
60
|
Halawani R, Buchert M, Chen YPP. Deep learning exploration of single-cell and spatially resolved cancer transcriptomics to unravel tumour heterogeneity. Comput Biol Med 2023; 164:107274. [PMID: 37506451 DOI: 10.1016/j.compbiomed.2023.107274] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 07/03/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023]
Abstract
Tumour heterogeneity is one of the critical confounding aspects in decoding tumour growth. Malignant cells display variations in their gene transcription profiles and mutation spectra even when originating from a single progenitor cell. Single-cell and spatial transcriptomics sequencing have recently emerged as key technologies for unravelling tumour heterogeneity. Single-cell sequencing promotes individual cell-type identification through transcriptome-wide gene expression measurements of each cell. Spatial transcriptomics facilitates identification of cell-cell interactions and the structural organization of heterogeneous cells within a tumour tissue through associating spatial RNA abundance of cells at distinct spots in the tissue section. However, extracting features and analyzing single-cell and spatial transcriptomics data poses challenges. Single-cell transcriptome data is extremely noisy and its sparse nature and dropouts can lead to misinterpretation of gene expression and the misclassification of cell types. Deep learning predictive power can overcome data challenges, provide high-resolution analysis and enhance precision oncology applications that involve early cancer prognosis, diagnosis, patient survival estimation and anti-cancer therapy planning. In this paper, we provide a background to and review of the recent progress of deep learning frameworks to investigate tumour heterogeneity using both single-cell and spatial transcriptomics data types.
Collapse
Affiliation(s)
- Raid Halawani
- Department of Computer Science and Information Technology, La Trobe University, Melbourne, Australia
| | - Michael Buchert
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Yi-Ping Phoebe Chen
- Department of Computer Science and Information Technology, La Trobe University, Melbourne, Australia.
| |
Collapse
|
61
|
Cooper M, Ji Z, Krishnan RG. Machine learning in computational histopathology: Challenges and opportunities. Genes Chromosomes Cancer 2023; 62:540-556. [PMID: 37314068 DOI: 10.1002/gcc.23177] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/15/2023] Open
Abstract
Digital histopathological images, high-resolution images of stained tissue samples, are a vital tool for clinicians to diagnose and stage cancers. The visual analysis of patient state based on these images are an important part of oncology workflow. Although pathology workflows have historically been conducted in laboratories under a microscope, the increasing digitization of histopathological images has led to their analysis on computers in the clinic. The last decade has seen the emergence of machine learning, and deep learning in particular, a powerful set of tools for the analysis of histopathological images. Machine learning models trained on large datasets of digitized histopathology slides have resulted in automated models for prediction and stratification of patient risk. In this review, we provide context for the rise of such models in computational histopathology, highlight the clinical tasks they have found success in automating, discuss the various machine learning techniques that have been applied to this domain, and underscore open problems and opportunities.
Collapse
Affiliation(s)
- Michael Cooper
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- University Health Network, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Zongliang Ji
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Rahul G Krishnan
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
62
|
Fu X, Sahai E, Wilkins A. Application of digital pathology-based advanced analytics of tumour microenvironment organisation to predict prognosis and therapeutic response. J Pathol 2023; 260:578-591. [PMID: 37551703 PMCID: PMC10952145 DOI: 10.1002/path.6153] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 08/09/2023]
Abstract
In recent years, the application of advanced analytics, especially artificial intelligence (AI), to digital H&E images, and other histological image types, has begun to radically change how histological images are used in the clinic. Alongside the recognition that the tumour microenvironment (TME) has a profound impact on tumour phenotype, the technical development of highly multiplexed immunofluorescence platforms has enhanced the biological complexity that can be captured in the TME with high precision. AI has an increasingly powerful role in the recognition and quantitation of image features and the association of such features with clinically important outcomes, as occurs in distinct stages in conventional machine learning. Deep-learning algorithms are able to elucidate TME patterns inherent in the input data with minimum levels of human intelligence and, hence, have the potential to achieve clinically relevant predictions and discovery of important TME features. Furthermore, the diverse repertoire of deep-learning algorithms able to interrogate TME patterns extends beyond convolutional neural networks to include attention-based models, graph neural networks, and multimodal models. To date, AI models have largely been evaluated retrospectively, outside the well-established rigour of prospective clinical trials, in part because traditional clinical trial methodology may not always be suitable for the assessment of AI technology. However, to enable digital pathology-based advanced analytics to meaningfully impact clinical care, specific measures of 'added benefit' to the current standard of care and validation in a prospective setting are important. This will need to be accompanied by adequate measures of explainability and interpretability. Despite such challenges, the combination of expanding datasets, increased computational power, and the possibility of integration of pre-clinical experimental insights into model development means there is exciting potential for the future progress of these AI applications. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Xiao Fu
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
- Biomolecular Modelling LaboratoryThe Francis Crick InstituteLondonUK
| | - Erik Sahai
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
| | - Anna Wilkins
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
- Division of Radiotherapy and ImagingInstitute of Cancer ResearchLondonUK
- Royal Marsden Hospitals NHS TrustLondonUK
| |
Collapse
|
63
|
Reis-Filho JS, Kather JN. Overcoming the challenges to implementation of artificial intelligence in pathology. J Natl Cancer Inst 2023; 115:608-612. [PMID: 36929936 PMCID: PMC10248832 DOI: 10.1093/jnci/djad048] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/02/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
Pathologists worldwide are facing remarkable challenges with increasing workloads and lack of time to provide consistently high-quality patient care. The application of artificial intelligence (AI) to digital whole-slide images has the potential of democratizing the access to expert pathology and affordable biomarkers by supporting pathologists in the provision of timely and accurate diagnosis as well as supporting oncologists by directly extracting prognostic and predictive biomarkers from tissue slides. The long-awaited adoption of AI in pathology, however, has not materialized, and the transformation of pathology is happening at a much slower pace than that observed in other fields (eg, radiology). Here, we provide a critical summary of the developments in digital and computational pathology in the last 10 years, outline key hurdles and ways to overcome them, and provide a perspective for AI-supported precision oncology in the future.
Collapse
Affiliation(s)
- Jorge S Reis-Filho
- Experimental Pathology, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jakob Nikolas Kather
- Department of Medicine I, University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, UK
| |
Collapse
|
64
|
Najdawi F, Sucipto K, Mistry P, Hennek S, Jayson CKB, Lin M, Fahy D, Kinsey S, Wapinski I, Beck AH, Resnick MB, Khosla A, Drage MG. Artificial Intelligence Enables Quantitative Assessment of Ulcerative Colitis Histology. Mod Pathol 2023; 36:100124. [PMID: 36841434 DOI: 10.1016/j.modpat.2023.100124] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/23/2022] [Accepted: 01/28/2023] [Indexed: 02/17/2023]
Abstract
Ulcerative colitis is a chronic inflammatory bowel disease that is characterized by a relapsing and remitting course. Assessment of disease activity critically informs treatment decisions. In addition to endoscopic remission, histologic remission is emerging as a treatment target and a key factor in the evaluation of disease activity and therapeutic efficacy. However, manual pathologist evaluation is semiquantitative and limited in granularity. Machine learning approaches are increasingly being developed to aid pathologists in accurate and reproducible scoring of histology, enabling precise quantitation of clinically relevant features. Here, we report the development and validation of convolutional neural network models that quantify histologic features pertinent to ulcerative colitis disease activity, directly from hematoxylin and eosin-stained whole slide images. Tissue and cell model predictions were used to generate quantitative human-interpretable features to fully characterize the histology samples. Tissue and cell predictions showed comparable agreement to pathologist annotations, and the extracted slide-level human-interpretable features demonstrated strong correlations with disease severity and pathologist-assigned Nancy histological index scores. Moreover, using a random forest classifier based on 13 human-interpretable features derived from the tissue and cell models, we were able to accurately predict Nancy histological index scores, with a weighted kappa (κ = 0.91) and Spearman correlation (⍴ = 0.89, P < .001) when compared with pathologist consensus Nancy histological index scores. We were also able to predict histologic remission, based on the absence of neutrophil extravasation, with a high accuracy of 0.97. This work demonstrates the potential of computer vision to enable a standardized and robust assessment of ulcerative colitis histopathology for translational research and improved evaluation of disease activity and prognosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Mary Lin
- PathAI, Inc, Boston, Massachusetts
| | | | | | | | | | | | | | | |
Collapse
|
65
|
Alam MR, Seo KJ, Abdul-Ghafar J, Yim K, Lee SH, Jang HJ, Jung CK, Chong Y. Recent application of artificial intelligence on histopathologic image-based prediction of gene mutation in solid cancers. Brief Bioinform 2023; 24:bbad151. [PMID: 37114657 DOI: 10.1093/bib/bbad151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
PURPOSE Evaluation of genetic mutations in cancers is important because distinct mutational profiles help determine individualized drug therapy. However, molecular analyses are not routinely performed in all cancers because they are expensive, time-consuming and not universally available. Artificial intelligence (AI) has shown the potential to determine a wide range of genetic mutations on histologic image analysis. Here, we assessed the status of mutation prediction AI models on histologic images by a systematic review. METHODS A literature search using the MEDLINE, Embase and Cochrane databases was conducted in August 2021. The articles were shortlisted by titles and abstracts. After a full-text review, publication trends, study characteristic analysis and comparison of performance metrics were performed. RESULTS Twenty-four studies were found mostly from developed countries, and their number is increasing. The major targets were gastrointestinal, genitourinary, gynecological, lung and head and neck cancers. Most studies used the Cancer Genome Atlas, with a few using an in-house dataset. The area under the curve of some of the cancer driver gene mutations in particular organs was satisfactory, such as 0.92 of BRAF in thyroid cancers and 0.79 of EGFR in lung cancers, whereas the average of all gene mutations was 0.64, which is still suboptimal. CONCLUSION AI has the potential to predict gene mutations on histologic images with appropriate caution. Further validation with larger datasets is still required before AI models can be used in clinical practice to predict gene mutations.
Collapse
Affiliation(s)
- Mohammad Rizwan Alam
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Kyung Jin Seo
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jamshid Abdul-Ghafar
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Kwangil Yim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hyun-Jong Jang
- Catholic Big Data Integration Center, Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Yosep Chong
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
66
|
Stahler A, Hoppe B, Na IK, Keilholz L, Müller L, Karthaus M, Fruehauf S, Graeven U, Fischer von Weikersthal L, Goekkurt E, Kasper S, Kind AJ, Kurreck A, Alig AHS, Held S, Reinacher-Schick A, Heinemann V, Horst D, Jarosch A, Stintzing S, Trarbach T, Modest DP. Consensus Molecular Subtypes as Biomarkers of Fluorouracil and Folinic Acid Maintenance Therapy With or Without Panitumumab in RAS Wild-Type Metastatic Colorectal Cancer (PanaMa, AIO KRK 0212). J Clin Oncol 2023; 41:2975-2987. [PMID: 37018649 DOI: 10.1200/jco.22.02582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
Abstract
PURPOSE Consensus molecular subtypes (CMSs) were evaluated as prognostic and predictive biomarkers of patients with RAS wild-type metastatic colorectal cancer (mCRC) receiving fluorouracil and folinic acid (FU/FA) with or without panitumumab (Pmab) after Pmab + mFOLFOX6 induction within the randomized phase II PanaMa trial. METHODS CMSs were determined in the safety set (ie, patients that received induction) and full analysis set (FAS; ie, randomly assigned patients who received maintenance) and correlated with median progression-free survival (PFS) and overall survival (OS) since the start of induction or maintenance treatment and objective response rates (ORRs). Hazard ratios (HRs) and 95% CI were calculated by univariate/multivariate Cox regression analyses. RESULTS Of 377 patients of the safety set, 296 (78.5%) had available CMS data: CMS1/2/3/4: 29 (9.8%)/122 (41.2%)/33 (11.2%)/112 (37.8%) and unclassifiable: 17 (5.7%). The CMSs were prognostic biomarkers in terms of PFS (P < .0001), OS (P < .0001), and ORR (P = .02) since the start of induction treatment. In FAS patients (n = 196), with CMS2/4 tumors, the addition of Pmab to FU/FA maintenance therapy was associated with longer PFS (CMS2: HR, 0.58 [95% CI, 0.36 to 0.95], P = .03; CMS4: HR, 0.63 [95% CI, 0.38 to 1.03], P = .07) and OS (CMS2: HR, 0.88 [95% CI, 0.52 to 1.52], P = .66; CMS4: HR, 0.54 [95% CI, 0.30 to 0.96], P = .04). The CMS interacted significantly with treatment in terms of PFS (CMS2 v CMS1/3: P = .02; CMS4 v CMS1/3: P = .03) and OS (CMS2 v CMS1/3: P = .03; CMS4 v CMS1/3: P < .001). CONCLUSION The CMS had a prognostic impact on PFS, OS, and ORR in RAS wild-type mCRC. In PanaMa, Pmab + FU/FA maintenance was associated with beneficial outcomes in CMS2/4, whereas no benefit was observed in CMS1/3 tumors.
Collapse
Affiliation(s)
- Arndt Stahler
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Beeke Hoppe
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Il-Kang Na
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DKTK, German Cancer Consortium, German Cancer Research Centre (DKFZ), Heidelberg, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, ECRC Experimental and Clinical Research Center, Berlin, Germany
| | - Luisa Keilholz
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Meinolf Karthaus
- Department of Hematology and Oncology, Munich Hospital Neuperlach, Munich, Germany
| | | | | | | | - Eray Goekkurt
- Practice of Hematology and Oncology (HOPE), Hamburg, Germany
- University Cancer Center Hamburg (UCCH), Hamburg, Germany
| | - Stefan Kasper
- DKTK, German Cancer Consortium, German Cancer Research Centre (DKFZ), Heidelberg, Germany
- Department of Medical Oncology, West German Cancer Center, Westdeutsches Tumorzentrum, University Hospital of Essen, Essen, Germany
| | - Andreas Jay Kind
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Annika Kurreck
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Annabel Helga Sophie Alig
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Anke Reinacher-Schick
- Department of Hematology, Oncology and Palliative Care, St Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Volker Heinemann
- DKTK, German Cancer Consortium, German Cancer Research Centre (DKFZ), Heidelberg, Germany
- Department of Medicine III and Comprehensive Cancer Center, University Hospital (LMU), Munich, Germany
| | - David Horst
- DKTK, German Cancer Consortium, German Cancer Research Centre (DKFZ), Heidelberg, Germany
- Department of Pathology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Armin Jarosch
- Department of Pathology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Stintzing
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DKTK, German Cancer Consortium, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Tanja Trarbach
- Department of Medical Oncology, West German Cancer Center, Westdeutsches Tumorzentrum, University Hospital of Essen, Essen, Germany
- Reha-Zentrum am Meer, Bad Zwischenahn, Niedersachsen, Germany
| | - Dominik Paul Modest
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DKTK, German Cancer Consortium, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| |
Collapse
|
67
|
Zhang X, Zhuge J, Liu J, Xia Z, Wang H, Gao Q, Jiang H, Qu Y, Fan L, Ma J, Tan C, Luo W, Luo Y. Prognostic signatures of sphingolipids: Understanding the immune landscape and predictive role in immunotherapy response and outcomes of hepatocellular carcinoma. Front Immunol 2023; 14:1153423. [PMID: 37006285 PMCID: PMC10063861 DOI: 10.3389/fimmu.2023.1153423] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a complex disease with a poor outlook for patients in advanced stages. Immune cells play an important role in the progression of HCC. The metabolism of sphingolipids functions in both tumor growth and immune infiltration. However, little research has focused on using sphingolipid factors to predict HCC prognosis. This study aimed to identify the key sphingolipids genes (SPGs) in HCC and develop a reliable prognostic model based on these genes. Methods The TCGA, GEO, and ICGC datasets were grouped using SPGs obtained from the InnateDB portal. A prognostic gene signature was created by applying LASSO-Cox analysis and evaluating it with Cox regression. The validity of the signature was verified using ICGC and GEO datasets. The tumor microenvironment (TME) was examined using ESTIMATE and CIBERSORT, and potential therapeutic targets were identified through machine learning. Single-cell sequencing was used to examine the distribution of signature genes in cells within the TME. Cell viability and migration were tested to confirm the role of the key SPGs. Results We identified 28 SPGs that have an impact on survival. Using clinicopathological features and 6 genes, we developed a nomogram for HCC. The high- and low-risk groups were found to have distinct immune characteristics and response to drugs. Unlike CD8 T cells, M0 and M2 macrophages were found to be highly infiltrated in the TME of the high-risk subgroup. High levels of SPGs were found to be a good indicator of response to immunotherapy. In cell function experiments, SMPD2 and CSTA were found to enhance survival and migration of Huh7 cells, while silencing these genes increased the sensitivity of Huh7 cells to lapatinib. Conclusion The study presents a six-gene signature and a nomogram that can aid clinicians in choosing personalized treatments for HCC patients. Furthermore, it uncovers the connection between sphingolipid-related genes and the immune microenvironment, offering a novel approach for immunotherapy. By focusing on crucial sphingolipid genes like SMPD2 and CSTA, the efficacy of anti-tumor therapy can be increased in HCC cells.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pathology, the Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Jinke Zhuge
- Department of Respiratory Medicine, Hainan Cancer Hospital, Hainan, China
| | - Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Huixiong Wang
- Department of Hepatobiliary Surgery, Hospital of Inner Mongolia Baotou Steel, Baotou, Inner Mongolia, China
| | - Qiang Gao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hao Jiang
- Department of Pathology, the Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Yanyu Qu
- Department of Pathology, the Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Linlin Fan
- Department of Pathology, the Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Jiali Ma
- Department of Pathology, the Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Chunhua Tan
- Department of Pathology, the Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Wei Luo
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yong Luo
- Department of Urology, The Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| |
Collapse
|
68
|
Wen Z, Wang S, Yang DM, Xie Y, Chen M, Bishop J, Xiao G. Deep learning in digital pathology for personalized treatment plans of cancer patients. Semin Diagn Pathol 2023; 40:109-119. [PMID: 36890029 DOI: 10.1053/j.semdp.2023.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Over the past decade, many new cancer treatments have been developed and made available to patients. However, in most cases, these treatments only benefit a specific subgroup of patients, making the selection of treatment for a specific patient an essential but challenging task for oncologists. Although some biomarkers were found to associate with treatment response, manual assessment is time-consuming and subjective. With the rapid developments and expanded implementation of artificial intelligence (AI) in digital pathology, many biomarkers can be quantified automatically from histopathology images. This approach allows for a more efficient and objective assessment of biomarkers, aiding oncologists in formulating personalized treatment plans for cancer patients. This review presents an overview and summary of the recent studies on biomarker quantification and treatment response prediction using hematoxylin-eosin (H&E) stained pathology images. These studies have shown that an AI-based digital pathology approach can be practical and will become increasingly important in improving the selection of cancer treatments for patients.
Collapse
Affiliation(s)
- Zhuoyu Wen
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shidan Wang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Donghan M Yang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yang Xie
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mingyi Chen
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Justin Bishop
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
69
|
Predicting gene mutation status via artificial intelligence technologies based on multimodal integration (MMI) to advance precision oncology. Semin Cancer Biol 2023; 91:1-15. [PMID: 36801447 DOI: 10.1016/j.semcancer.2023.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/30/2023] [Accepted: 02/15/2023] [Indexed: 02/21/2023]
Abstract
Personalized treatment strategies for cancer frequently rely on the detection of genetic alterations which are determined by molecular biology assays. Historically, these processes typically required single-gene sequencing, next-generation sequencing, or visual inspection of histopathology slides by experienced pathologists in a clinical context. In the past decade, advances in artificial intelligence (AI) technologies have demonstrated remarkable potential in assisting physicians with accurate diagnosis of oncology image-recognition tasks. Meanwhile, AI techniques make it possible to integrate multimodal data such as radiology, histology, and genomics, providing critical guidance for the stratification of patients in the context of precision therapy. Given that the mutation detection is unaffordable and time-consuming for a considerable number of patients, predicting gene mutations based on routine clinical radiological scans or whole-slide images of tissue with AI-based methods has become a hot issue in actual clinical practice. In this review, we synthesized the general framework of multimodal integration (MMI) for molecular intelligent diagnostics beyond standard techniques. Then we summarized the emerging applications of AI in the prediction of mutational and molecular profiles of common cancers (lung, brain, breast, and other tumor types) pertaining to radiology and histology imaging. Furthermore, we concluded that there truly exist multiple challenges of AI techniques in the way of its real-world application in the medical field, including data curation, feature fusion, model interpretability, and practice regulations. Despite these challenges, we still prospect the clinical implementation of AI as a highly potential decision-support tool to aid oncologists in future cancer treatment management.
Collapse
|
70
|
Zhang S, Mu W, Dong D, Wei J, Fang M, Shao L, Zhou Y, He B, Zhang S, Liu Z, Liu J, Tian J. The Applications of Artificial Intelligence in Digestive System Neoplasms: A Review. HEALTH DATA SCIENCE 2023; 3:0005. [PMID: 38487199 PMCID: PMC10877701 DOI: 10.34133/hds.0005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/05/2022] [Indexed: 03/17/2024]
Abstract
Importance Digestive system neoplasms (DSNs) are the leading cause of cancer-related mortality with a 5-year survival rate of less than 20%. Subjective evaluation of medical images including endoscopic images, whole slide images, computed tomography images, and magnetic resonance images plays a vital role in the clinical practice of DSNs, but with limited performance and increased workload of radiologists or pathologists. The application of artificial intelligence (AI) in medical image analysis holds promise to augment the visual interpretation of medical images, which could not only automate the complicated evaluation process but also convert medical images into quantitative imaging features that associated with tumor heterogeneity. Highlights We briefly introduce the methodology of AI for medical image analysis and then review its clinical applications including clinical auxiliary diagnosis, assessment of treatment response, and prognosis prediction on 4 typical DSNs including esophageal cancer, gastric cancer, colorectal cancer, and hepatocellular carcinoma. Conclusion AI technology has great potential in supporting the clinical diagnosis and treatment decision-making of DSNs. Several technical issues should be overcome before its application into clinical practice of DSNs.
Collapse
Affiliation(s)
- Shuaitong Zhang
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Wei Mu
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jingwei Wei
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Mengjie Fang
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Lizhi Shao
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Yu Zhou
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Bingxi He
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Song Zhang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jianhua Liu
- Department of Oncology, Guangdong Provincial People's Hospital/Second Clinical Medical College of Southern Medical University/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jie Tian
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
71
|
Fremond S, Andani S, Barkey Wolf J, Dijkstra J, Melsbach S, Jobsen JJ, Brinkhuis M, Roothaan S, Jurgenliemk-Schulz I, Lutgens LCHW, Nout RA, van der Steen-Banasik EM, de Boer SM, Powell ME, Singh N, Mileshkin LR, Mackay HJ, Leary A, Nijman HW, Smit VTHBM, Creutzberg CL, Horeweg N, Koelzer VH, Bosse T. Interpretable deep learning model to predict the molecular classification of endometrial cancer from haematoxylin and eosin-stained whole-slide images: a combined analysis of the PORTEC randomised trials and clinical cohorts. Lancet Digit Health 2023; 5:e71-e82. [PMID: 36496303 DOI: 10.1016/s2589-7500(22)00210-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Endometrial cancer can be molecularly classified into POLEmut, mismatch repair deficient (MMRd), p53 abnormal (p53abn), and no specific molecular profile (NSMP) subgroups. We aimed to develop an interpretable deep learning pipeline for whole-slide-image-based prediction of the four molecular classes in endometrial cancer (im4MEC), to identify morpho-molecular correlates, and to refine prognostication. METHODS This combined analysis included diagnostic haematoxylin and eosin-stained slides and molecular and clinicopathological data from 2028 patients with intermediate-to-high-risk endometrial cancer from the PORTEC-1 (n=466), PORTEC-2 (n=375), and PORTEC-3 (n=393) randomised trials and the TransPORTEC pilot study (n=110), the Medisch Spectrum Twente cohort (n=242), a case series of patients with POLEmut endometrial cancer in the Leiden Endometrial Cancer Repository (n=47), and The Cancer Genome Atlas-Uterine Corpus Endometrial Carcinoma cohort (n=395). PORTEC-3 was held out as an independent test set and a four-fold cross validation was performed. Performance was measured with the macro and class-wise area under the receiver operating characteristic curve (AUROC). Whole-slide images were segmented into tiles of 360 μm resized to 224 × 224 pixels. im4MEC was trained to learn tile-level morphological features with self-supervised learning and to molecularly classify whole-slide images with an attention mechanism. The top 20 tiles with the highest attention scores were reviewed to identify morpho-molecular correlates. Predictions of a nuclear classification deep learning model serve to derive interpretable morphological features. We analysed 5-year recurrence-free survival and explored prognostic refinement by molecular class using the Kaplan-Meier method. FINDINGS im4MEC attained macro-average AUROCs of 0·874 (95% CI 0·856-0·893) on four-fold cross-validation and 0·876 on the independent test set. The class-wise AUROCs were 0·849 for POLEmut (n=51), 0·844 for MMRd (n=134), 0·883 for NSMP (n=120), and 0·928 for p53abn (n=88). POLEmut and MMRd tiles had a high density of lymphocytes, p53abn tiles had strong nuclear atypia, and the morphology of POLEmut and MMRd endometrial cancer overlapped. im4MEC highlighted a low tumour-to-stroma ratio as a potentially novel characteristic feature of the NSMP class. 5-year recurrence-free survival was significantly different between im4MEC predicted molecular classes in PORTEC-3 (log-rank p<0·0001). The ten patients with aggressive p53abn endometrial cancer that was predicted as MMRd showed inflammatory morphology and appeared to have a better prognosis than patients with correctly predicted p53abn endometrial cancer (p=0·30). The four patients with NSMP endometrial cancer that was predicted as p53abn showed higher nuclear atypia and appeared to have a worse prognosis than patients with correctly predicted NSMP (p=0·13). Patients with MMRd endometrial cancer predicted as POLEmut had an excellent prognosis, as do those with true POLEmut endometrial cancer. INTERPRETATION We present the first interpretable deep learning model, im4MEC, for haematoxylin and eosin-based prediction of molecular endometrial cancer classification. im4MEC robustly identified morpho-molecular correlates and could enable further prognostic refinement of patients with endometrial cancer. FUNDING The Hanarth Foundation, the Promedica Foundation, and the Swiss Federal Institutes of Technology.
Collapse
Affiliation(s)
- Sarah Fremond
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Sonali Andani
- Department of Computer Science, ETH Zurich, Zurich, Switzerland; Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Jouke Dijkstra
- Department of Vascular and Molecular Imaging, Leiden University Medical Center, Leiden, Netherlands
| | - Sinéad Melsbach
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Jan J Jobsen
- Department of Radiation Oncology, Medisch Spectrum Twente, Enschede, Netherlands
| | | | | | - Ina Jurgenliemk-Schulz
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ludy C H W Lutgens
- Department of Radiation Oncology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Remi A Nout
- Department of Radiation Oncology, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Stephanie M de Boer
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Melanie E Powell
- Department of Clinical Oncology, Barts Health NHS Trust, London, UK
| | - Naveena Singh
- Department of Pathology, Barts Health NHS Trust, London, UK
| | - Linda R Mileshkin
- Department of Medical Oncology, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | - Helen J Mackay
- Department of Medical Oncology and Hematology, Odette Cancer Center Sunnybrook Health Sciences Center, Toronto, ON, Canada
| | - Alexandra Leary
- Medical Oncology Department, Gustave Roussy Institute, Villejuif, France
| | - Hans W Nijman
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
| | | | - Carien L Creutzberg
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Nanda Horeweg
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
72
|
Wu P, Wu K, Li Z, Liu H, Yang K, Zhou R, Zhou Z, Xing N, Wu S. Multimodal investigation of bladder cancer data based on computed tomography, whole slide imaging, and transcriptomics. Quant Imaging Med Surg 2023; 13:1023-1035. [PMID: 36819263 PMCID: PMC9929396 DOI: 10.21037/qims-22-679] [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: 06/27/2022] [Accepted: 12/08/2022] [Indexed: 01/11/2023]
Abstract
Background Multimodal analysis has shown great potential in the diagnosis and management of cancer. This study aimed to determine the multimodal data associations between radiological, pathologic, and molecular characteristics in bladder cancer. Methods A retrospective study of computed tomography (CT), pathologic slice, and RNA sequencing data from 127 consecutive adult patients in China who underwent bladder surgery and were pathologically diagnosed with bladder cancer was conducted. A total of 200 radiological and 1,029 pathologic features were extracted by radiomics and pathomics. Multimodal associations analysis and structural equation modeling were used to measure the cross-modal associations and structural relationships between CT and pathologic slice. A convolutional neural network was constructed for molecular subtyping based on multimodal imaging features. Class activation maps were used to examine the feature contribution in model decision-making. Cox regression and Kaplan-Meier survival analysis were used to explore the relevance of multimodal features to the prognosis of patients with bladder cancer. Results A total of 77 densely associated blocks of feature pairs were identified between CT and whole slide images. The largest cross-modal associated block reflected the tumor-grade properties. A significant relation was found between pathological features and molecular subtypes (β=0.396; P<0.001). High-grade bladder cancer showed heterogeneity of significance across different scales and higher disorders at the microscopic level. The fused radiological and pathologic features achieved higher accuracy (area under the curve: 0.89; 95% CI: 0.75-1.0) than the unimodal method. Thirteen prognosis-related features from CT and whole slide images were identified. Conclusions Our work demonstrated the associations between CT, pathologic slices, and molecular signatures, and the potential to use multimodal data analysis in related clinical applications. Multimodal data analysis showed the potential of cross-inference of modal data and had higher diagnostic accuracy than the unimodal method.
Collapse
Affiliation(s)
- Peng Wu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China;,Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
| | - Kai Wu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China;,Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
| | - Zhe Li
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China;,Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
| | - Hanlin Liu
- Department of Radiology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
| | - Kai Yang
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Rong Zhou
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China;,Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
| | - Ziyu Zhou
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Nianzeng Xing
- State Key Laboratory of Molecular Oncology and Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Song Wu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China;,Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
73
|
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: 21] [Impact Index Per Article: 10.5] [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
|
74
|
Zhang H, Xi Q, Zhang F, Li Q, Jiao Z, Ni X. Application of Deep Learning in Cancer Prognosis Prediction Model. Technol Cancer Res Treat 2023; 22:15330338231199287. [PMID: 37709267 PMCID: PMC10503281 DOI: 10.1177/15330338231199287] [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] [Indexed: 09/16/2023] Open
Abstract
As an important branch of artificial intelligence and machine learning, deep learning (DL) has been widely used in various aspects of cancer auxiliary diagnosis, among which cancer prognosis is the most important part. High-accuracy cancer prognosis is beneficial to the clinical management of patients with cancer. Compared with other methods, DL models can significantly improve the accuracy of prediction. Therefore, this article is a systematic review of the latest research on DL in cancer prognosis prediction. First, the data type, construction process, and performance evaluation index of the DL model are introduced in detail. Then, the current mainstream baseline DL cancer prognosis prediction models, namely, deep neural networks, convolutional neural networks, deep belief networks, deep residual networks, and vision transformers, including network architectures, the latest application in cancer prognosis, and their respective characteristics, are discussed. Next, some key factors that affect the predictive performance of the model and common performance enhancement techniques are listed. Finally, the limitations of the DL cancer prognosis prediction model in clinical practice are summarized, and the future research direction is prospected. This article could provide relevant researchers with a comprehensive understanding of DL cancer prognostic models and is expected to promote the research progress of cancer prognosis prediction.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Radiotherapy Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, China
| | - Qianyi Xi
- Department of Radiotherapy Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, China
- School of Microelectronics and Control Engineering, Changzhou University, Changzhou, China
| | - Fan Zhang
- Department of Radiotherapy Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, China
- School of Microelectronics and Control Engineering, Changzhou University, Changzhou, China
| | - Qixuan Li
- Department of Radiotherapy Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, China
- School of Microelectronics and Control Engineering, Changzhou University, Changzhou, China
| | - Zhuqing Jiao
- School of Microelectronics and Control Engineering, Changzhou University, Changzhou, China
| | - Xinye Ni
- Department of Radiotherapy Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, China
| |
Collapse
|
75
|
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
|
76
|
Pai RK, Banerjee I, Shivji S, Jain S, Hartman D, Buchanan DD, Jenkins MA, Schaeffer DF, Rosty C, Como J, Phipps AI, Newcomb PA, Burnett-Hartman AN, Marchand LL, Samadder NJ, Patel B, Swallow C, Lindor NM, Gallinger SJ, Grant RC, Westerling-Bui T, Conner J, Cyr DP, Kirsch R, Pai RK. Quantitative Pathologic Analysis of Digitized Images of Colorectal Carcinoma Improves Prediction of Recurrence-Free Survival. Gastroenterology 2022; 163:1531-1546.e8. [PMID: 35985511 PMCID: PMC9716432 DOI: 10.1053/j.gastro.2022.08.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/02/2022] [Accepted: 08/09/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS To examine whether quantitative pathologic analysis of digitized hematoxylin and eosin slides of colorectal carcinoma (CRC) correlates with clinicopathologic features, molecular alterations, and prognosis. METHODS A quantitative segmentation algorithm (QuantCRC) was applied to 6468 digitized hematoxylin and eosin slides of CRCs. Fifteen parameters were recorded from each image and tested for associations with clinicopathologic features and molecular alterations. A prognostic model was developed to predict recurrence-free survival using data from the internal cohort (n = 1928) and validated on an internal test (n = 483) and external cohort (n = 938). RESULTS There were significant differences in QuantCRC according to stage, histologic subtype, grade, venous/lymphatic/perineural invasion, tumor budding, CD8 immunohistochemistry, mismatch repair status, KRAS mutation, BRAF mutation, and CpG methylation. A prognostic model incorporating stage, mismatch repair, and QuantCRC resulted in a Harrell's concordance (c)-index of 0.714 (95% confidence interval [CI], 0.702-0.724) in the internal test and 0.744 (95% CI, 0.741-0.754) in the external cohort. Removing QuantCRC from the model reduced the c-index to 0.679 (95% CI, 0.673-0.694) in the external cohort. Prognostic risk groups were identified, which provided a hazard ratio of 2.24 (95% CI, 1.33-3.87, P = .004) for low vs high-risk stage III CRCs and 2.36 (95% CI, 1.07-5.20, P = .03) for low vs high-risk stage II CRCs, in the external cohort after adjusting for established risk factors. The predicted median 36-month recurrence rate for high-risk stage III CRCs was 32.7% vs 13.4% for low-risk stage III and 15.8% for high-risk stage II vs 5.4% for low-risk stage II CRCs. CONCLUSIONS QuantCRC provides a powerful adjunct to routine pathologic reporting of CRC. A prognostic model using QuantCRC improves prediction of recurrence-free survival.
Collapse
Affiliation(s)
- Reetesh K. Pai
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Imon Banerjee
- Department of Radiology and Machine Intelligence in Medicine and Imaging Center (MI-2), Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Sameer Shivji
- Department of Pathology, Mount Sinai Hospital, Toronto, ON, Canada
| | - Suchit Jain
- Department of Radiology and Machine Intelligence in Medicine and Imaging Center (MI-2), Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Douglas Hartman
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Daniel D. Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, Australia
- Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Mark A. Jenkins
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| | - David F. Schaeffer
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC, Canada
| | - Christophe Rosty
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
- Envoi Specialist Pathologists, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Julia Como
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, Australia
| | - Amanda I. Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Polly A. Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Andrea N. Burnett-Hartman
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Institute for Health Research, Kaiser Permanente Colorado, Denver, Colorado, USA
| | - Loic Le Marchand
- Department of Epidemiology, University of Hawaii, Seattle, Washington, USA
| | - Niloy J. Samadder
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Bhavik Patel
- Department of Radiology and Machine Intelligence in Medicine and Imaging Center (MI-2), Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Carol Swallow
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Surgical Oncology, Princess Margaret Cancer Centre and Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of General Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Noralane M. Lindor
- Department of Health Sciences Research Mayo Clinic, Scottsdale, Arizona, USA
| | - Steven J. Gallinger
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Hepatobiliary/Pancreatic Surgical Oncology Program, University Health Network, Toronto, Ontario, Canada
| | - Robert C. Grant
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - James Conner
- Department of Pathology, Mount Sinai Hospital, Toronto, ON, Canada
| | - David P. Cyr
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Surgical Oncology, Princess Margaret Cancer Centre and Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of General Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Richard Kirsch
- Department of Pathology, Mount Sinai Hospital, Toronto, ON, Canada
| | - Rish K. Pai
- Department of Pathology and Laboratory Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| |
Collapse
|
77
|
Chen Z, Li X, Yang M, Zhang H, Xu XS. Optimization of deep learning models for the prediction of gene mutations using unsupervised clustering. J Pathol Clin Res 2022; 9:3-17. [PMID: 36376239 PMCID: PMC9732687 DOI: 10.1002/cjp2.302] [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: 06/09/2022] [Revised: 10/03/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022]
Abstract
Deep learning models are increasingly being used to interpret whole-slide images (WSIs) in digital pathology and to predict genetic mutations. Currently, it is commonly assumed that tumor regions have most of the predictive power. However, it is reasonable to assume that other tissues from the tumor microenvironment may also provide important predictive information. In this paper, we propose an unsupervised clustering-based multiple-instance deep learning model for the prediction of genetic mutations using WSIs of three cancer types obtained from The Cancer Genome Atlas. Our proposed model facilitates the identification of spatial regions related to specific gene mutations and exclusion of patches that lack predictive information through the use of unsupervised clustering. This results in a more accurate prediction of gene mutations when compared with models using all image patches on WSIs and two recently published algorithms for all three different cancer types evaluated in this study. In addition, our study validates the hypothesis that the prediction of gene mutations solely based on tumor regions on WSI slides may not always provide the best performance. Other tissue types in the tumor microenvironment could provide a better prediction ability than tumor tissues alone. These results highlight the heterogeneity in the tumor microenvironment and the importance of identification of predictive image patches in digital pathology prediction tasks.
Collapse
Affiliation(s)
- Zihan Chen
- School of Data ScienceUniversity of Science and Technology of ChinaHefeiPR China
| | - Xingyu Li
- Department of Statistics and Finance, School of ManagementUniversity of Science and Technology of ChinaHefeiPR China
| | - Miaomiao Yang
- Clinical Pathology CenterThe Fourth Affiliated Hospital of Anhui Medical UniversityHefeiPR China
| | - Hong Zhang
- Department of Statistics and Finance, School of ManagementUniversity of Science and Technology of ChinaHefeiPR China
| | - Xu Steven Xu
- Clinical Pharmacology and Quantitative ScienceGenmab Inc.PrincetonNJUSA
| |
Collapse
|
78
|
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
|
79
|
Househam J, Heide T, Cresswell GD, Spiteri I, Kimberley C, Zapata L, Lynn C, James C, Mossner M, Fernandez-Mateos J, Vinceti A, Baker AM, Gabbutt C, Berner A, Schmidt M, Chen B, Lakatos E, Gunasri V, Nichol D, Costa H, Mitchinson M, Ramazzotti D, Werner B, Iorio F, Jansen M, Caravagna G, Barnes CP, Shibata D, Bridgewater J, Rodriguez-Justo M, Magnani L, Sottoriva A, Graham TA. Phenotypic plasticity and genetic control in colorectal cancer evolution. Nature 2022; 611:744-753. [PMID: 36289336 PMCID: PMC9684078 DOI: 10.1038/s41586-022-05311-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/01/2022] [Indexed: 12/12/2022]
Abstract
Genetic and epigenetic variation, together with transcriptional plasticity, contribute to intratumour heterogeneity1. The interplay of these biological processes and their respective contributions to tumour evolution remain unknown. Here we show that intratumour genetic ancestry only infrequently affects gene expression traits and subclonal evolution in colorectal cancer (CRC). Using spatially resolved paired whole-genome and transcriptome sequencing, we find that the majority of intratumour variation in gene expression is not strongly heritable but rather 'plastic'. Somatic expression quantitative trait loci analysis identified a number of putative genetic controls of expression by cis-acting coding and non-coding mutations, the majority of which were clonal within a tumour, alongside frequent structural alterations. Consistently, computational inference on the spatial patterning of tumour phylogenies finds that a considerable proportion of CRCs did not show evidence of subclonal selection, with only a subset of putative genetic drivers associated with subclone expansions. Spatial intermixing of clones is common, with some tumours growing exponentially and others only at the periphery. Together, our data suggest that most genetic intratumour variation in CRC has no major phenotypic consequence and that transcriptional plasticity is, instead, widespread within a tumour.
Collapse
Affiliation(s)
- Jacob Househam
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Timon Heide
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - George D Cresswell
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Inmaculada Spiteri
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Chris Kimberley
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Luis Zapata
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Claire Lynn
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Chela James
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Maximilian Mossner
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | | | - Ann-Marie Baker
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Calum Gabbutt
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Alison Berner
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Melissa Schmidt
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Bingjie Chen
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Eszter Lakatos
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Vinaya Gunasri
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Daniel Nichol
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Helena Costa
- UCL Cancer Institute, University College London, London, UK
| | - Miriam Mitchinson
- Histopathology Department, University College London Hospitals NHS Foundation Trust, London, UK
| | - Daniele Ramazzotti
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Benjamin Werner
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Francesco Iorio
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Marnix Jansen
- UCL Cancer Institute, University College London, London, UK
| | - Giulio Caravagna
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Department of Mathematics and Geosciences, University of Trieste, Trieste, Italy
| | - Chris P Barnes
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Darryl Shibata
- Department of Pathology, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | | | | | - Luca Magnani
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
- Computational Biology Research Centre, Human Technopole, Milan, Italy.
| | - Trevor A Graham
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
80
|
van de Weerd S, Smit MA, Roelands J, Mesker WE, Bedognetti D, Kuppen PJK, Putter H, Tollenaar RAEM, Roodhart JML, Hendrickx W, Medema JP, van Krieken JHJM. Correlation of Immunological and Histopathological Features with Gene Expression-Based Classifiers in Colon Cancer Patients. Int J Mol Sci 2022; 23:ijms232012707. [PMID: 36293565 PMCID: PMC9604175 DOI: 10.3390/ijms232012707] [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: 09/23/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/25/2022] Open
Abstract
The purpose of this study was to evaluate the association between four distinct histopathological features: (1) tumor infiltrating lymphocytes, (2) mucinous differentiation, (3) tumor-stroma ratio, plus (4) tumor budding and two gene expression-based classifiers—(1) consensus molecular subtypes (CMS) plus (2) colorectal cancer intrinsic subtypes (CRIS). All four histopathological features were retrospectively scored on hematoxylin and eosin sections of the most invasive part of the primary tumor in 218 stage II and III colon cancer patients from two independent cohorts (AMC-AJCC-90 and AC-ICAM). RNA-based CMS and CRIS assignments were independently obtained for all patients. Contingency tables were constructed and a χ2 test was used to test for statistical significance. Odds ratios with 95% confidence intervals were calculated. The presence of tumor infiltrating lymphocytes and a mucinous phenotype (>50% mucinous surface area) were strongly correlated with CMS1 (p < 0.001 and p = 0.008) and CRIS-A (p = 0.006 and p < 0.001). The presence of mucus (≥ 10%) was associated with CMS3: mucus was present in 64.1% of all CMS3 tumors (p < 0.001). Although a clear association between tumor-stroma ratio and CMS4 was established in this study (p = 0.006), still 32 out of 61 (52.5%) CMS4 tumors were scored as stroma-low, indicating that CMS4 tumors cannot be identified solely based on stromal content. Higher budding counts were seen in CMS4 and CRIS-B tumors (p = 0.045 and p = 0.046). No other associations of the measured parameters were seen for any of the other CRIS subtypes. Our analysis revealed clear associations between histopathologic features and CMS or CRIS subtypes. However, identification of distinct molecular subtypes solely based on histopathology proved to be infeasible. Combining both molecular and morphologic features could potentially improve patient stratification.
Collapse
Affiliation(s)
- Simone van de Weerd
- Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Pathology, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
- Oncode Institute, Amsterdam UMC, University of Amsterdam, 3521 AL Amsterdam, The Netherlands
| | - Marloes A. Smit
- Department of Surgery, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Jessica Roelands
- Department of Surgery, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
- Translational Medicine Department, Research Branch, Sidra Medicine, Doha 26999, Qatar
| | - Wilma E. Mesker
- Department of Surgery, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Davide Bedognetti
- Translational Medicine Department, Research Branch, Sidra Medicine, Doha 26999, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
| | - Peter J. K. Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Hein Putter
- Department of Medical Statistics, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Rob A. E. M. Tollenaar
- Department of Surgery, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Jeanine M. L. Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Wouter Hendrickx
- Translational Medicine Department, Research Branch, Sidra Medicine, Doha 26999, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
| | - Jan Paul Medema
- Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Oncode Institute, Amsterdam UMC, University of Amsterdam, 3521 AL Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-20-566-2368
| | | |
Collapse
|
81
|
Volovat SR, Augustin I, Zob D, Boboc D, Amurariti F, Volovat C, Stefanescu C, Stolniceanu CR, Ciocoiu M, Dumitras EA, Danciu M, Apostol DGC, Drug V, Shurbaji SA, Coca LG, Leon F, Iftene A, Herghelegiu PC. Use of Personalized Biomarkers in Metastatic Colorectal Cancer and the Impact of AI. Cancers (Basel) 2022; 14:4834. [PMID: 36230757 PMCID: PMC9562853 DOI: 10.3390/cancers14194834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/18/2022] [Accepted: 09/29/2022] [Indexed: 12/09/2022] Open
Abstract
Colorectal cancer is a major cause of cancer-related death worldwide and is correlated with genetic and epigenetic alterations in the colonic epithelium. Genetic changes play a major role in the pathophysiology of colorectal cancer through the development of gene mutations, but recent research has shown an important role for epigenetic alterations. In this review, we try to describe the current knowledge about epigenetic alterations, including DNA methylation and histone modifications, as well as the role of non-coding RNAs as epigenetic regulators and the prognostic and predictive biomarkers in metastatic colorectal disease that can allow increases in the effectiveness of treatments. Additionally, the intestinal microbiota's composition can be an important biomarker for the response to strategies based on the immunotherapy of CRC. The identification of biomarkers in mCRC can be enhanced by developing artificial intelligence programs. We present the actual models that implement AI technology as a bridge connecting ncRNAs with tumors and conducted some experiments to improve the quality of the model used as well as the speed of the model that provides answers to users. In order to carry out this task, we implemented six algorithms: the naive Bayes classifier, the random forest classifier, the decision tree classifier, gradient boosted trees, logistic regression and SVM.
Collapse
Affiliation(s)
- Simona-Ruxandra Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Iolanda Augustin
- Department of Medical Oncology, AI.Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Daniela Zob
- Department of Medical Oncology, AI.Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Diana Boboc
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Florin Amurariti
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Constantin Volovat
- Department of Medical Oncology, “Euroclinic” Center of Oncology, 2 Vasile Conta Str., 700106 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Eduard Alexandru Dumitras
- Department of Pathophysiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Anesthesiology and Intensive Care, Regional Institute of Oncology, 700115 Iasi, Romania
| | - Mihai Danciu
- Pathology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | | | - Vasile Drug
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
- Gastroenterology Clinic, Institute of Gastroenterology and Hepatology, ‘St. Spiridon’ Clinical Hospital, 700115 Iasi, Romania
| | - Sinziana Al Shurbaji
- Gastroenterology Clinic, Institute of Gastroenterology and Hepatology, ‘St. Spiridon’ Clinical Hospital, 700115 Iasi, Romania
| | - Lucia-Georgiana Coca
- Faculty of Computer Science, Alexandru Ioan Cuza University, 700115 Iasi, Romania
| | - Florin Leon
- Faculty of Automatic Control and Computer Engineering, Gheorghe Asachi Technical University, 700115 Iasi, Romania
| | - Adrian Iftene
- Faculty of Computer Science, Alexandru Ioan Cuza University, 700115 Iasi, Romania
| | - Paul-Corneliu Herghelegiu
- Faculty of Automatic Control and Computer Engineering, Gheorghe Asachi Technical University, 700115 Iasi, Romania
| |
Collapse
|
82
|
Jakab A, Patai ÁV, Micsik T. Digital image analysis provides robust tissue microenvironment-based prognosticators in patients with stage I-IV colorectal cancer. Hum Pathol 2022; 128:141-151. [PMID: 35820451 DOI: 10.1016/j.humpath.2022.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/03/2022] [Accepted: 07/02/2022] [Indexed: 11/26/2022]
Abstract
In patients with colorectal cancer (CRC), a promising marker is tumor-stroma ratio (TSR). Quantification issues highlight the importance of precise assessment that might be solved by artificial intelligence-based digital image analysis systems. Some alternatives have been offered so far, although these platforms are either proprietary developments or require additional programming skills. Our aim was to validate a user-friendly, commercially available software running in everyday computational environment to improve TSR assessment and also to compare the prognostic value of assessing TSR in 3 distinct regions of interests, like hotspot, invasive front, and whole tumor. Furthermore, we compared the prognostic power of TSR with the newly suggested carcinoma percentage (CP) and carcinoma-stroma percentage (CSP). Slides of 185 patients with stage I-IV CRC with clinical follow-up data were scanned and evaluated by a senior pathologist. A machine learning-based digital pathology software was trained to recognize tumoral and stromal compartments. The aforementioned parameters were evaluated in the hotspot, invasive front, and whole tumor area, both visually and by machine learning. Patients were classified based on TSR, CP, and CSP values. On multivariate analysis, TSR-hotspot was found to be an independent prognostic factor of overall survival (hazard ratio for TSR-hotspotsoftware: 2.005 [95% confidence interval (CI): 1.146-3.507], P = .011, for TSR-hotspotvisual: 1.781 [CI: 1.060-2.992], P = .029). Also, TSR was an independent predictor for distant metastasis and local relapse in most settings. Generally, software performance was comparable to visual evaluation and delivered reliable prognostication in more settings also with CP and CSP values. This study presents that software-assisted evaluation is a robust prognosticator. Our approach used a less sophisticated and thus easily accessible software without the aid of a convolutional neural network; however, it was still effective enough to deliver reliable prognostic information.
Collapse
Affiliation(s)
- Anna Jakab
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Üllői őt 26, H-1085, Hungary; Interdisciplinary Gastroenterology Working Group, Semmelweis University, Budapest, Üllői út 78, H-1082, Hungary.
| | - Árpád V Patai
- Interdisciplinary Gastroenterology Working Group, Semmelweis University, Budapest, Üllői út 78, H-1082, Hungary; Department of Surgery, Transplantation and Gastroenterology, Semmelweis University, Budapest, Üllői út 78, H-1082, Hungary
| | - Tamás Micsik
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Üllői őt 26, H-1085, Hungary; Interdisciplinary Gastroenterology Working Group, Semmelweis University, Budapest, Üllői út 78, H-1082, Hungary; Saint George Teaching Hospital of Fejér County, Székesfehérvár, Seregélyesi út 3, HU-8000, Hungary
| |
Collapse
|
83
|
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
|
84
|
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
|
85
|
Peters NA, Constantinides A, Ubink I, van Kuik J, Bloemendal HJ, van Dodewaard JM, Brink MA, Schwartz TP, Lolkema MP, Lacle MM, Moons LM, Geesing J, van Grevenstein WM, Roodhart JML, Koopman M, Elias SG, Borel Rinkes IH, Kranenburg O. Consensus molecular subtype 4 (CMS4)-targeted therapy in primary colon cancer: A proof-of-concept study. Front Oncol 2022; 12:969855. [PMID: 36147916 PMCID: PMC9486194 DOI: 10.3389/fonc.2022.969855] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundMesenchymal Consensus Molecular Subtype 4 (CMS4) colon cancer is associated with poor prognosis and therapy resistance. In this proof-of-concept study, we assessed whether a rationally chosen drug could mitigate the distinguishing molecular features of primary CMS4 colon cancer.MethodsIn the ImPACCT trial, informed consent was obtained for molecular subtyping at initial diagnosis of colon cancer using a validated RT-qPCR CMS4-test on three biopsies per tumor (Phase-1, n=69 patients), and for neoadjuvant CMS4-targeting therapy with imatinib (Phase-2, n=5). Pre- and post-treatment tumor biopsies were analyzed by RNA-sequencing and immunohistochemistry. Imatinib-induced gene expression changes were associated with molecular subtypes and survival in an independent cohort of 3232 primary colon cancer.ResultsThe CMS4-test classified 52/172 biopsies as CMS4 (30%). Five patients consented to imatinib treatment prior to surgery, yielding 15 pre- and 15 post-treatment samples for molecular analysis. Imatinib treatment caused significant suppression of mesenchymal genes and upregulation of genes encoding epithelial junctions. The gene expression changes induced by imatinib were associated with improved survival and a shift from CMS4 to CMS2.ConclusionImatinib may have value as a CMS-switching drug in primary colon cancer and induces a gene expression program that is associated with improved survival.
Collapse
Affiliation(s)
- Niek A. Peters
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Alexander Constantinides
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Inge Ubink
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Joyce van Kuik
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Haiko J. Bloemendal
- Department of Internal Medicine, Meander Medical Center, Amersfoort, Netherlands
- Department of Internal Medicine/Oncology, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | | | - Menno A. Brink
- Department of Gastroenterology, Meander Medical Center, Amersfoort, Netherlands
| | - Thijs P. Schwartz
- Department of Gastroenterology, Meander Medical Center, Amersfoort, Netherlands
| | | | - Miangela M. Lacle
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Leon M. Moons
- Department of Gastroenterology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Joost Geesing
- Department of Gastroenterology, Diakonessenhuis, Utrecht, Netherlands
| | - Wilhelmina M.U. van Grevenstein
- Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jeanine M. L. Roodhart
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Sjoerd G. Elias
- Julius Centre for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Inne H.M. Borel Rinkes
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- *Correspondence: Inne H.M. Borel Rinkes, ; Onno Kranenburg,
| | - Onno Kranenburg
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- *Correspondence: Inne H.M. Borel Rinkes, ; Onno Kranenburg,
| |
Collapse
|
86
|
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: 179] [Impact Index Per Article: 59.7] [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
|
87
|
Krieg C, Weber LM, Fosso B, Marzano M, Hardiman G, Olcina MM, Domingo E, El Aidy S, Mallah K, Robinson MD, Guglietta S. Complement downregulation promotes an inflammatory signature that renders colorectal cancer susceptible to immunotherapy. J Immunother Cancer 2022; 10:e004717. [PMID: 36137652 PMCID: PMC9511657 DOI: 10.1136/jitc-2022-004717] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND AND AIMS The role of inflammatory immune responses in colorectal cancer (CRC) development and response to therapy is a matter of intense debate. While inflammation is a known driver of CRC, inflammatory immune infiltrates are a positive prognostic factor in CRC and predispose to response to immune checkpoint blockade (ICB) therapy. Unfortunately, over 85% of CRC cases are primarily unresponsive to ICB due to the absence of an immune infiltrate, and even the cases that show an initial immune infiltration can become refractory to ICB. The identification of therapy supportive immune responses in the field has been partially hindered by the sparsity of suitable mouse models to recapitulate the human disease. In this study, we aimed to understand how the dysregulation of the complement anaphylatoxin C3a receptor (C3aR), observed in subsets of patients with CRC, affects the immune responses, the development of CRC, and response to ICB therapy. METHODS We use a comprehensive approach encompassing analysis of publicly available human CRC datasets, inflammation-driven and newly generated spontaneous mouse models of CRC, and multiplatform high-dimensional analysis of immune responses using microbiota sequencing, RNA sequencing, and mass cytometry. RESULTS We found that patients' regulation of the complement C3aR is associated with epigenetic modifications. Specifically, downregulation of C3ar1 in human CRC promotes a tumor microenvironment characterized by the accumulation of innate and adaptive immune cells that support antitumor immunity. In addition, in vivo studies in our newly generated mouse model revealed that the lack of C3a in the colon activates a microbiota-mediated proinflammatory program which promotes the development of tumors with an immune signature that renders them responsive to the ICB therapy. CONCLUSIONS Our findings reveal that C3aR may act as a previously unrecognized checkpoint to enhance antitumor immunity in CRC. C3aR can thus be exploited to overcome ICB resistance in a larger group of patients with CRC.
Collapse
Affiliation(s)
- Carsten Krieg
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center Charleston, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lukas M Weber
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Bruno Fosso
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy
| | - Marinella Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy
| | - Gary Hardiman
- School of Biological Sciences and Institute for Global Food Security, Queens University of Belfast, Belfast, UK
| | - Monica M Olcina
- Institute of Radiation Oncology, Medical Research Council Oxford Institute for Radiation Oncology, Oxford, UK
| | - Enric Domingo
- Institute of Radiation Oncology, Medical Research Council Oxford Institute for Radiation Oncology, Oxford, UK
| | - Sahar El Aidy
- Host-microbe Metabolic Interactions, Microbiology, University of Groningen, Groningen, The Netherlands
| | - Khalil Mallah
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Mark D Robinson
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Silvia Guglietta
- Hollings Cancer Center Charleston, Medical University of South Carolina, Charleston, South Carolina, USA
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
88
|
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
|
89
|
Wong ANN, He Z, Leung KL, To CCK, Wong CY, Wong SCC, Yoo JS, Chan CKR, Chan AZ, Lacambra MD, Yeung MHY. Current Developments of Artificial Intelligence in Digital Pathology and Its Future Clinical Applications in Gastrointestinal Cancers. Cancers (Basel) 2022; 14:3780. [PMID: 35954443 PMCID: PMC9367360 DOI: 10.3390/cancers14153780] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 02/05/2023] Open
Abstract
The implementation of DP will revolutionize current practice by providing pathologists with additional tools and algorithms to improve workflow. Furthermore, DP will open up opportunities for development of AI-based tools for more precise and reproducible diagnosis through computational pathology. One of the key features of AI is its capability to generate perceptions and recognize patterns beyond the human senses. Thus, the incorporation of AI into DP can reveal additional morphological features and information. At the current rate of AI development and adoption of DP, the interest in computational pathology is expected to rise in tandem. There have already been promising developments related to AI-based solutions in prostate cancer detection; however, in the GI tract, development of more sophisticated algorithms is required to facilitate histological assessment of GI specimens for early and accurate diagnosis. In this review, we aim to provide an overview of the current histological practices in AP laboratories with respect to challenges faced in image preprocessing, present the existing AI-based algorithms, discuss their limitations and present clinical insight with respect to the application of AI in early detection and diagnosis of GI cancer.
Collapse
Affiliation(s)
- Alex Ngai Nick Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (A.N.N.W.); (Z.H.); (K.L.L.); (C.Y.W.); (S.C.C.W.); (J.S.Y.)
| | - Zebang He
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (A.N.N.W.); (Z.H.); (K.L.L.); (C.Y.W.); (S.C.C.W.); (J.S.Y.)
| | - Ka Long Leung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (A.N.N.W.); (Z.H.); (K.L.L.); (C.Y.W.); (S.C.C.W.); (J.S.Y.)
| | - Curtis Chun Kit To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China; (C.C.K.T.); (C.K.R.C.); (M.D.L.)
| | - Chun Yin Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (A.N.N.W.); (Z.H.); (K.L.L.); (C.Y.W.); (S.C.C.W.); (J.S.Y.)
| | - Sze Chuen Cesar Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (A.N.N.W.); (Z.H.); (K.L.L.); (C.Y.W.); (S.C.C.W.); (J.S.Y.)
| | - Jung Sun Yoo
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (A.N.N.W.); (Z.H.); (K.L.L.); (C.Y.W.); (S.C.C.W.); (J.S.Y.)
| | - Cheong Kin Ronald Chan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China; (C.C.K.T.); (C.K.R.C.); (M.D.L.)
| | - Angela Zaneta Chan
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, Shatin, Hong Kong SAR, China;
| | - Maribel D. Lacambra
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China; (C.C.K.T.); (C.K.R.C.); (M.D.L.)
| | - Martin Ho Yin Yeung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (A.N.N.W.); (Z.H.); (K.L.L.); (C.Y.W.); (S.C.C.W.); (J.S.Y.)
| |
Collapse
|
90
|
Ahuja A, Kefalakes H. Clinical Applications of Artificial Intelligence in Gastroenterology: Excitement and Evidence. Gastroenterology 2022; 163:341-344. [PMID: 35489435 DOI: 10.1053/j.gastro.2022.04.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/14/2022] [Accepted: 04/23/2022] [Indexed: 12/04/2022]
Affiliation(s)
- Amisha Ahuja
- Temple University Hospital, Philadelphia, Pennsylvania
| | | |
Collapse
|
91
|
Wu X, Ye Y, Vega KJ, Yao J. Consensus Molecular Subtypes Efficiently Classify Gastric Adenocarcinomas and Predict the Response to Anti-PD-1 Immunotherapy. Cancers (Basel) 2022; 14:3740. [PMID: 35954402 PMCID: PMC9367605 DOI: 10.3390/cancers14153740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/14/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
Background: Gastric adenocarcinoma (GAC) is highly heterogeneous and closely related to colorectal cancer (CRC) both molecularly and functionally. GAC is currently subtyped using a system developed by TCGA. However, with the emergence of immunotherapies, this system has failed to identify suitable treatment candidates. Methods: Consensus molecular subtypes (CMSs) developed for CRC were used for molecular subtyping in GAC based on public expression cohorts, including TCGA, ACRG, and a cohort of GAC patients treated with the programmed cell death 1 (PD-1) inhibitor pembrolizumab. All aspects of each subtype, including clinical outcome, molecular characteristics, oncogenic pathway activity, and the response to immunotherapy, were fully explored. Results: CMS classification was efficiently applied to GAC. CMS4, characterized by EMT activation, stromal invasion, angiogenesis, and the worst clinical outcomes (median OS 24.2 months), was the predominant subtype (38.8%~44.3%) and an independent prognostic indicator that outperformed classical TCGA subtyping. CMS1 (20.9%~21.5%) displayed hypermutation, low SCNV, immune activation, and best clinical outcomes (median OS > 120 months). CMS3 (17.95%~25.7%) was characterized by overactive metabolism, KRAS mutation, and intermediate outcomes (median OS 85.6 months). CMS2 (14.6%~16.3%) was enriched for WNT and MYC activation, differentiated epithelial characteristics, APC mutation, lack of ARID1A, and intermediate outcomes (median OS 48.7 months). Notably, CMS1 was strongly correlated with immunotherapy biomarkers and favorable for the anti-PD-1 drug pembrolizumab, whereas CMS4 was poorly responsive but became more sensitive after EMT-based stratification. Conclusions: Our study reveals the practical utility of CMS classification for GAC to improve clinical outcomes and identify candidates who will respond to immunotherapy.
Collapse
Affiliation(s)
- Xiangyan Wu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350122, China;
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou 350122, China
| | - Yuhan Ye
- Department of Pathology, Zhongshan Hospital, Xiamen University, Xiamen 361004, China;
| | - Kenneth J. Vega
- Department of Gastroenterology and Hepatology, Augusta University, Augusta, GA 30912, USA;
| | - Jiannan Yao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
92
|
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
|
93
|
Kouzu K, Nearchou IP, Kajiwara Y, Tsujimoto H, Lillard K, Kishi Y, Ueno H. Deep-learning-based classification of desmoplastic reaction on H&E predicts poor prognosis in oesophageal squamous cell carcinoma. Histopathology 2022; 81:255-263. [PMID: 35758184 DOI: 10.1111/his.14708] [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: 03/10/2022] [Revised: 05/16/2022] [Accepted: 05/31/2022] [Indexed: 12/24/2022]
Abstract
AIMS Desmoplastic reaction (DR) categorisation has been shown to be a promising prognostic factor in oesophageal squamous cell carcinoma (ESCC). The usual DR evaluation is performed using semiquantitative scores, which can be subjective. This study aimed to investigate whether a deep-learning classifier could be used for DR classification. We further assessed the prognostic significance of the deep-learning classifier and compared it to that of manual DR reporting and other pathological factors currently used in the clinic. METHODS AND RESULTS From 222 surgically resected ESCC cases, 31 randomly selected haematoxylin-eosin-digitised whole slides of patients with immature DR were used to train and develop a deep-learning classifier. The classifier was trained for 89 370 iterations. The accuracy of the deep-learning classifier was assessed to 30 unseen cases, and the results revealed a Dice coefficient score of 0.81. For survival analysis, the classifier was then applied to the entire cohort of patients, which was split into a training (n = 156) and a test (n = 66) cohort. The automated DR classification had a higher prognostic significance for disease-specific survival than the manually classified DR in both the training and test cohorts. In addition, the automated DR classification outperformed the prognostic accuracy of the gold-standard factors of tumour depth and lymph node metastasis. CONCLUSIONS This study demonstrated that DR can be objectively and quantitatively assessed in ESCC using a deep-learning classifier and that automatically classed DR has a higher prognostic significance than manual DR and other features currently used in the clinic.
Collapse
Affiliation(s)
- Keita Kouzu
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Ines P Nearchou
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Yoshiki Kajiwara
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | | | | | - Yoji Kishi
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Saitama, Japan
| |
Collapse
|
94
|
Bell PD, Pai RK. Immune Response in Colorectal Carcinoma: A Review of Its Significance as a Predictive and Prognostic Biomarker. Histopathology 2022; 81:696-714. [PMID: 35758208 DOI: 10.1111/his.14713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022]
Abstract
Colorectal carcinoma is a leading cause of cancer-related death worldwide. There is significant prognostic heterogeneity in stage II and III tumours, necessitating the development of new biomarkers to better identify patients at risk of disease progression. Recently, the tumour immune environment, particularly the type and quantity of T lymphocytes, has been shown to be a useful biomarker in predicting prognosis for patients with colorectal carcinoma. In this review, the significance of the immune response in colorectal carcinoma, including its influence on prognosis and response to therapy, will be detailed.
Collapse
Affiliation(s)
- Phoenix D Bell
- Department of Pathology, University of Pittsburgh Medical Centre, Pittsburgh, PA, 15213, USA
| | - Reetesh K Pai
- Department of Pathology, University of Pittsburgh Medical Centre, Pittsburgh, PA, 15213, USA
| |
Collapse
|
95
|
Towards a national strategy for digital pathology in Switzerland. Virchows Arch 2022; 481:647-652. [PMID: 35622144 PMCID: PMC9534807 DOI: 10.1007/s00428-022-03345-0] [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: 01/20/2022] [Revised: 04/25/2022] [Accepted: 05/18/2022] [Indexed: 11/02/2022]
Abstract
Precision medicine is entering a new era of digital diagnostics; the availability of integrated digital pathology (DP) and structured clinical datasets has the potential to become a key catalyst for biomedical research, education and business development. In Europe, national programs for sharing of this data will be crucial for the development, testing, and validation of machine learning-enabled tools supporting clinical decision-making. Here, the Swiss Digital Pathology Consortium (SDiPath) discusses the creation of a Swiss Digital Pathology Infrastructure (SDPI), which aims to develop a unified national DP network bringing together the Swiss Personalized Health Network (SPHN) with Swiss university hospitals and subsequent inclusion of cantonal and private institutions. This effort builds on existing developments for the national implementation of structured pathology reporting. Opening this national infrastructure and data to international researchers in a sequential rollout phase can enable the large-scale integration of health data and pooling of resources for research purposes and clinical trials. Therefore, the concept of a SDPI directly synergizes with the priorities of the European Commission communication on the digital transformation of healthcare on an international level, and with the aims of the Swiss State Secretariat for Economic Affairs (SECO) for advancing research and innovation in the digitalization domain. SDPI directly addresses the needs of existing national and international research programs in neoplastic and non-neoplastic diseases by providing unprecedented access to well-curated clinicopathological datasets for the development and implementation of novel integrative methods for analysis of clinical outcomes and treatment response. In conclusion, a SDPI would facilitate and strengthen inter-institutional collaboration in technology, clinical development, business and research at a national and international scale, promoting improved patient care via precision medicine.
Collapse
|
96
|
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
|
97
|
Zheng B, Fang L. Spatially resolved transcriptomics provide a new method for cancer research. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:179. [PMID: 35590346 PMCID: PMC9118771 DOI: 10.1186/s13046-022-02385-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/06/2022] [Indexed: 12/22/2022]
Abstract
A major feature of cancer is the heterogeneity, both intratumoral and intertumoral. Traditional single-cell techniques have given us a comprehensive understanding of the biological characteristics of individual tumor cells, but the lack of spatial context of the transcriptome has limited the study of cell-to-cell interaction patterns and hindered further exploration of tumor heterogeneity. In recent years, the advent of spatially resolved transcriptomics (SRT) technology has made possible the multidimensional analysis of the tumor microenvironment in the context of intact tissues. Different SRT methods are applicable to different working ranges due to different working principles. In this paper, we review the advantages and disadvantages of various current SRT methods and the overall idea of applying these techniques to oncology studies, hoping to help researchers find breakthroughs. Finally, we discussed the future direction of SRT technology, and deeper investigation into the complex mechanisms of tumor development from different perspectives through multi-omics fusion, paving the way for precisely targeted tumor therapy.
Collapse
Affiliation(s)
- Bowen Zheng
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, People's Republic of China
| | - Lin Fang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, People's Republic of China.
| |
Collapse
|
98
|
Li CH, Cai D, Zhong ME, Lv MY, Huang ZP, Zhu Q, Hu C, Qi H, Wu X, Gao F. Multi-Size Deep Learning Based Preoperative Computed Tomography Signature for Prognosis Prediction of Colorectal Cancer. Front Genet 2022; 13:880093. [PMID: 35646105 PMCID: PMC9133721 DOI: 10.3389/fgene.2022.880093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Preoperative and postoperative evaluation of colorectal cancer (CRC) patients is crucial for subsequent treatment guidance. Our study aims to provide a timely and rapid assessment of the prognosis of CRC patients with deep learning according to non-invasive preoperative computed tomography (CT) and explore the underlying biological explanations.Methods: A total of 808 CRC patients with preoperative CT (development cohort: n = 426, validation cohort: n = 382) were enrolled in our study. We proposed a novel end-to-end Multi-Size Convolutional Neural Network (MSCNN) to predict the risk of CRC recurrence with CT images (CT signature). The prognostic performance of CT signature was evaluated by Kaplan-Meier curve. An integrated nomogram was constructed to improve the clinical utility of CT signature by combining with other clinicopathologic factors. Further visualization and correlation analysis for CT deep features with paired gene expression profiles were performed to reveal the molecular characteristics of CRC tumors learned by MSCNN in radiographic imaging.Results: The Kaplan-Meier analysis showed that CT signature was a significant prognostic factor for CRC disease-free survival (DFS) prediction [development cohort: hazard ratio (HR): 50.7, 95% CI: 28.4–90.6, p < 0.001; validation cohort: HR: 2.04, 95% CI: 1.44–2.89, p < 0.001]. Multivariable analysis confirmed the independence prognostic value of CT signature (development cohort: HR: 30.7, 95% CI: 19.8–69.3, p < 0.001; validation cohort: HR: 1.83, 95% CI: 1.19–2.83, p = 0.006). Dimension reduction and visualization of CT deep features demonstrated a high correlation with the prognosis of CRC patients. Functional pathway analysis further indicated that CRC patients with high CT signature presented down-regulation of several immunology pathways. Correlation analysis found that CT deep features were mainly associated with activation of metabolic and proliferative pathways.Conclusions: Our deep learning based preoperative CT signature can effectively predict prognosis of CRC patients. Integration analysis of multi-omic data revealed that some molecular characteristics of CRC tumor can be captured by deep learning in CT images.
Collapse
Affiliation(s)
- Cheng-Hang Li
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- School of Computer Science and Engineering, Guangzhou Higher Education Mega Center, Sun Yat-sen University, Guangzhou, China
| | - Du Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min-Er Zhong
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min-Yi Lv
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ze-Ping Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiqi Zhu
- Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Chuling Hu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haoning Qi
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaojian Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xiaojian Wu, ; Feng Gao,
| | - Feng Gao
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xiaojian Wu, ; Feng Gao,
| |
Collapse
|
99
|
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
|
100
|
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: 82] [Impact Index Per Article: 27.3] [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
|