651
|
Rakha EA, Alsaleem M, ElSharawy KA, Toss MS, Raafat S, Mihai R, Minhas FA, Green AR, Rajpoot NM, Dalton LW, Mongan NP. Visual histological assessment of morphological features reflects the underlying molecular profile in invasive breast cancer: a morphomolecular study. Histopathology 2020; 77:631-645. [PMID: 32618014 DOI: 10.1111/his.14199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/22/2020] [Accepted: 06/26/2020] [Indexed: 12/29/2022]
Abstract
AIMS Tumour genotype and phenotype are related and can predict outcome. In this study, we hypothesised that the visual assessment of breast cancer (BC) morphological features can provide valuable insight into underlying molecular profiles. METHODS AND RESULTS The Cancer Genome Atlas (TCGA) BC cohort was used (n = 743) and morphological features, including Nottingham grade and its components and nucleolar prominence, were assessed utilising whole-slide images (WSIs). Two independent scores were assigned, and discordant cases were utilised to represent cases with intermediate morphological features. Differentially expressed genes (DEGs) were identified for each feature, compared among concordant/discordant cases and tested for specific pathways. Concordant grading was observed in 467 of 743 (63%) of cases. Among concordant case groups, eight common DEGs (UGT8, DDC, RGR, RLBP1, SPRR1B, CXorf49B, PSAPL1 and SPRR2G) were associated with overall tumour grade and its components. These genes are related mainly to cellular proliferation, differentiation and metabolism. The number of DEGs in cases with discordant grading was larger than those identified in concordant cases. The largest number of DEGs was observed in discordant grade 1:3 cases (n = 1185). DEGs were identified for each discordant component. Some DEGs were uniquely associated with well-defined specific morphological features, whereas expression/co-expression of other genes was identified across multiple features and underlined intermediate morphological features. CONCLUSION Morphological features are probably related to distinct underlying molecular profiles that drive both morphology and behaviour. This study provides further evidence to support the use of image-based analysis of WSIs, including artificial intelligence algorithms, to predict tumour molecular profiles and outcome.
Collapse
Affiliation(s)
- Emad A Rakha
- School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Mansour Alsaleem
- School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Khloud A ElSharawy
- School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Michael S Toss
- School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Sara Raafat
- School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Raluca Mihai
- School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Fayyaz A Minhas
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Andrew R Green
- School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Nasir M Rajpoot
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Leslie W Dalton
- Department of Histopathology, South Austin Hospital, Austin, TX, USA
| | - Nigel P Mongan
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.,Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, University of Nottingham Biodiscovery Institute, Nottingham, UK
| |
Collapse
|
652
|
Zhang H, Kalirai H, Acha-Sagredo A, Yang X, Zheng Y, Coupland SE. Piloting a Deep Learning Model for Predicting Nuclear BAP1 Immunohistochemical Expression of Uveal Melanoma from Hematoxylin-and-Eosin Sections. Transl Vis Sci Technol 2020; 9:50. [PMID: 32953248 PMCID: PMC7476670 DOI: 10.1167/tvst.9.2.50] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
Background Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. Monosomy 3 and BAP1 mutation are strong prognostic factors predicting metastatic risk in UM. Nuclear BAP1 (nBAP1) expression is a close immunohistochemical surrogate for both genetic alterations. Not all laboratories perform routine BAP1 immunohistochemistry or genetic testing, and rely mainly on clinical information and anatomic/morphologic analyses for UM prognostication. The purpose of our study was to pilot deep learning (DL) techniques to predict nBAP1 expression on whole slide images (WSIs) of hematoxylin and eosin (H&E) stained UM sections. Methods One hundred forty H&E-stained UMs were scanned at 40 × magnification, using commercially available WSI image scanners. The training cohort comprised 66 BAP1+ and 74 BAP1− UM, with known chromosome 3 status and clinical outcomes. Nonoverlapping areas of three different dimensions (512 × 512, 1024 × 1024, and 2048 × 2048 pixels) for comparison were extracted from tumor regions in each WSI, and were resized to 256 × 256 pixels. Deep convolutional neural networks (Resnet18 pre-trained on Imagenet) and auto-encoder-decoders (U-Net) were trained to predict nBAP1 expression of these patches. Trained models were tested on the patches cropped from a test cohort of WSIs of 16 BAP1+ and 28 BAP1− UM cases. Results The trained model with best performance achieved area under the curve values of 0.90 for patches and 0.93 for slides on the test set. Conclusions Our results show the effectiveness of DL for predicting nBAP1 expression in UM on the basis of H&E sections only. Translational Relevance Our pilot demonstrates a high capacity of artificial intelligence-related techniques for automated prediction on the basis of histomorphology, and may be translatable into routine histology laboratories.
Collapse
Affiliation(s)
- Hongrun Zhang
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.,Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Amelia Acha-Sagredo
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Xiaoyun Yang
- Chinese Academy of Sciences (CAS) IntelliCloud Technology Co., Ltd., Shanghai, China
| | - Yalin Zheng
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Sarah E Coupland
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.,Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
653
|
Radakovich N, Cortese M, Nazha A. Acute myeloid leukemia and artificial intelligence, algorithms and new scores. Best Pract Res Clin Haematol 2020; 33:101192. [PMID: 33038981 PMCID: PMC7548395 DOI: 10.1016/j.beha.2020.101192] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/27/2020] [Indexed: 12/23/2022]
Abstract
Artificial intelligence, and more narrowly machine-learning, is beginning to expand humanity's capacity to analyze increasingly large and complex datasets. Advances in computer hardware and software have led to breakthroughs in multiple sectors of our society, including a burgeoning role in medical research and clinical practice. As the volume of medical data grows at an apparently exponential rate, particularly since the human genome project laid the foundation for modern genetic inquiry, informatics tools like machine learning are becoming crucial in analyzing these data to provide meaningful tools for diagnostic, prognostic, and therapeutic purposes. Within medicine, hematologic diseases can be particularly challenging to understand and treat given the increasingly complex and intercalated genetic, epigenetic, immunologic, and regulatory pathways that must be understood to optimize patient outcomes. In acute myeloid leukemia (AML), new developments in machine learning algorithms have enabled a deeper understanding of disease biology and the development of better prognostic and predictive tools. Ongoing work in the field brings these developments incrementally closer to clinical implementation.
Collapse
Affiliation(s)
- Nathan Radakovich
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, United States
| | - Matthew Cortese
- Department of Hematology and Medical Oncology, Cleveland Clinic, United States
| | - Aziz Nazha
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, United States; Department of Hematology and Medical Oncology, Cleveland Clinic, United States; Center for Clinical Artificial Intelligence, Cleveland Clinic, United States.
| |
Collapse
|
654
|
Jin P, Ji X, Kang W, Li Y, Liu H, Ma F, Ma S, Hu H, Li W, Tian Y. Artificial intelligence in gastric cancer: a systematic review. J Cancer Res Clin Oncol 2020; 146:2339-2350. [PMID: 32613386 DOI: 10.1007/s00432-020-03304-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 06/26/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVE This study aims to systematically review the application of artificial intelligence (AI) techniques in gastric cancer and to discuss the potential limitations and future directions of AI in gastric cancer. METHODS A systematic review was performed that follows the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Pubmed, EMBASE, the Web of Science, and the Cochrane Library were used to search for gastric cancer publications with an emphasis on AI that were published up to June 2020. The terms "artificial intelligence" and "gastric cancer" were used to search for the publications. RESULTS A total of 64 articles were included in this review. In gastric cancer, AI is mainly used for molecular bio-information analysis, endoscopic detection for Helicobacter pylori infection, chronic atrophic gastritis, early gastric cancer, invasion depth, and pathology recognition. AI may also be used to establish predictive models for evaluating lymph node metastasis, response to drug treatments, and prognosis. In addition, AI can be used for surgical training, skill assessment, and surgery guidance. CONCLUSIONS In the foreseeable future, AI applications can play an important role in gastric cancer management in the era of precision medicine.
Collapse
Affiliation(s)
- Peng Jin
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Xiaoyan Ji
- Department of Emergency Ward, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Wenzhe Kang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yang Li
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Hao Liu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Fuhai Ma
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Shuai Ma
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Haitao Hu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Weikun Li
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yantao Tian
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
655
|
Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet 2020; 396:635-648. [PMID: 32861308 DOI: 10.1016/s0140-6736(20)31288-5] [Citation(s) in RCA: 2584] [Impact Index Per Article: 516.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
Gastric cancer is the fifth most common cancer and the third most common cause of cancer death globally. Risk factors for the condition include Helicobacter pylori infection, age, high salt intake, and diets low in fruit and vegetables. Gastric cancer is diagnosed histologically after endoscopic biopsy and staged using CT, endoscopic ultrasound, PET, and laparoscopy. It is a molecularly and phenotypically highly heterogeneous disease. The main treatment for early gastric cancer is endoscopic resection. Non-early operable gastric cancer is treated with surgery, which should include D2 lymphadenectomy (including lymph node stations in the perigastric mesentery and along the celiac arterial branches). Perioperative or adjuvant chemotherapy improves survival in patients with stage 1B or higher cancers. Advanced gastric cancer is treated with sequential lines of chemotherapy, starting with a platinum and fluoropyrimidine doublet in the first line; median survival is less than 1 year. Targeted therapies licensed to treat gastric cancer include trastuzumab (HER2-positive patients first line), ramucirumab (anti-angiogenic second line), and nivolumab or pembrolizumab (anti-PD-1 third line).
Collapse
Affiliation(s)
- Elizabeth C Smyth
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Hill's Road, Cambridge, UK.
| | - Magnus Nilsson
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden; Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Heike I Grabsch
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands; Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Nicole Ct van Grieken
- Department of Pathology, Amsterdam University Medical Centre, Cancer Center Amsterdam, VU University, Amsterdam, Netherlands
| | - Florian Lordick
- University Cancer Center Leipzig, Leipzig University Medical Center, Leipzig, Germany
| |
Collapse
|
656
|
Kutzner H, Jutzi TB, Krahl D, Krieghoff-Henning EI, Heppt MV, Hekler A, Schmitt M, Maron RCR, Fröhling S, von Kalle C, Brinker TJ. Overdiagnosis of melanoma - causes, consequences and solutions. J Dtsch Dermatol Ges 2020; 18:1236-1243. [PMID: 32841508 DOI: 10.1111/ddg.14233] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
Malignant melanoma is the skin tumor that causes most deaths in Germany. At an early stage, melanoma is well treatable, so early detection is essential. However, the skin cancer screening program in Germany has been criticized because although melanomas have been diagnosed more frequently since introduction of the program, the mortality from malignant melanoma has not decreased. This indicates that the observed increase in melanoma diagnoses be due to overdiagnosis, i.e. to the detection of lesions that would never have created serious health problems for the patients. One of the reasons is the challenging distinction between some benign and malignant lesions. In addition, there may be lesions that are biologically equivocal, and other lesions that are classified as malignant according to current criteria, but that grow so slowly that they would never have posed a threat to patient's life. So far, these "indolent" melanomas cannot be identified reliably due to a lack of biomarkers. Moreover, the likelihood that an in-situ melanoma will progress to an invasive tumor still cannot be determined with any certainty. When benign lesions are diagnosed as melanoma, the consequences are unnecessary psychological and physical stress for the affected patients and incurred therapy costs. Vice versa, underdiagnoses in the sense of overlooked melanomas can adversely affect patients' prognoses and may necessitate more intense therapies. Novel diagnostic options could reduce the number of over- and underdiagnoses and contribute to more objective diagnoses in borderline cases. One strategy that has yielded promising results in pilot studies is the use of artificial intelligence-based diagnostic tools. However, these applications still await translation into clinical and pathological routine.
Collapse
Affiliation(s)
| | - Tanja B Jutzi
- Digital Biomarkers for Oncology group (DBO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dieter Krahl
- Private Laboratory for Dermatohistopathology, Mönchhofstraße 52, Heidelberg, Germany
| | - Eva I Krieghoff-Henning
- Digital Biomarkers for Oncology group (DBO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Markus V Heppt
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Achim Hekler
- Digital Biomarkers for Oncology group (DBO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Max Schmitt
- Digital Biomarkers for Oncology group (DBO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roman C R Maron
- Digital Biomarkers for Oncology group (DBO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Fröhling
- Digital Biomarkers for Oncology group (DBO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christof von Kalle
- Berlin Institute of Health (BIH) and Charité-University Medical Center Berlin, Berlin, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology group (DBO), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
657
|
Schulz MA, Yeo BTT, Vogelstein JT, Mourao-Miranada J, Kather JN, Kording K, Richards B, Bzdok D. Different scaling of linear models and deep learning in UKBiobank brain images versus machine-learning datasets. Nat Commun 2020; 11:4238. [PMID: 32843633 PMCID: PMC7447816 DOI: 10.1038/s41467-020-18037-z] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
Recently, deep learning has unlocked unprecedented success in various domains, especially using images, text, and speech. However, deep learning is only beneficial if the data have nonlinear relationships and if they are exploitable at available sample sizes. We systematically profiled the performance of deep, kernel, and linear models as a function of sample size on UKBiobank brain images against established machine learning references. On MNIST and Zalando Fashion, prediction accuracy consistently improves when escalating from linear models to shallow-nonlinear models, and further improves with deep-nonlinear models. In contrast, using structural or functional brain scans, simple linear models perform on par with more complex, highly parameterized models in age/sex prediction across increasing sample sizes. In sum, linear models keep improving as the sample size approaches ~10,000 subjects. Yet, nonlinearities for predicting common phenotypes from typical brain scans remain largely inaccessible to the examined kernel and deep learning methods.
Collapse
Affiliation(s)
- Marc-Andre Schulz
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Rheinisch-Westfälische Technische Hochschule (RWTH), Aachen University, Aachen, Germany
| | - B T Thomas Yeo
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore
- Centre for Sleep and Cognition (CSC) and Centre for Translational Magnetic Resonance Research (TMR), National University of Singapore, Singapore, Singapore
- N.1 Institute for Health and Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore
| | - Joshua T Vogelstein
- Department of Biomedical Engineering, Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Janaina Mourao-Miranada
- Max Planck University College London Centre for Computational Psychiatry and Ageing Research, University College London, London, UK
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Jakob N Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Konrad Kording
- Department of Neuroscience and Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Blake Richards
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
- School of Computer Science, McGill University, Montréal, Québec, Canada
- Canadian Institute for Advanced Research, Toronto, Ontario, Canada
- Mila - Quebec Artificial Intelligence Institute, Montréal, Québec, Canada
| | - Danilo Bzdok
- Mila - Quebec Artificial Intelligence Institute, Montréal, Québec, Canada.
- Neurospin, Commissariat à l'Energie Atomique (CEA) Saclay, Gif-sur-Yvette, France.
- Parietal Team, Institut National de Recherche en Informatique et en Automatique (INRIA), Gif-sur-Yvette, France.
- Faculty of Medicine, Department of Biomedical Engineering, McConnell Brain imaging Centre, Montreal Neurological Institute (MNI), McGill University, Montreal, Québec, Canada.
| |
Collapse
|
658
|
Liu Y, Li X, Zheng A, Zhu X, Liu S, Hu M, Luo Q, Liao H, Liu M, He Y, Chen Y. Predict Ki-67 Positive Cells in H&E-Stained Images Using Deep Learning Independently From IHC-Stained Images. Front Mol Biosci 2020; 7:183. [PMID: 32903653 PMCID: PMC7438787 DOI: 10.3389/fmolb.2020.00183] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To obtain molecular information in slides directly from H&E staining slides, which apparently display morphological information, to show that some differences in molecular level have already encoded in morphology. METHODS In this paper, we selected Ki-67-expression as the representative of molecular information. We proposed a method that can predict Ki-67 positive cells directly from H&E stained slides by a deep convolutional network model. To train this model, we constructed a dataset containing Ki-67 negative or positive cell images and background images. These images were all extracted from H&E stained WSIs and the Ki-67 expression was acquired from the corresponding IHC stained WSIs. The trained model was evaluated both on classification performance and the ability to quantify Ki-67 expression in H&E stained images. RESULTS The model achieved an average accuracy of 0.9371 in discrimination of Ki-67 negative cell images, positive cell images and background images. As for evaluation of quantification performance, the correlation coefficient between the quantification results of H&E stained images predicted by our model and that of IHC stained images obtained by color channel filtering is 0.80. CONCLUSION AND SIGNIFICANCE Our study indicates that the deep learning model has a good performance both on prediction of Ki-67 positive cells and quantification of Ki-67 expression in cancer samples stained by H&E. More generally, this study shows that deep learning is a powerful tool in exploring the relationship between morphological information and molecular information. AVAILABILITY AND IMPLEMENTATION The main program is available at https://github.com/liuyiqing2018/predict_Ki-67_from_HE.
Collapse
Affiliation(s)
- Yiqing Liu
- Department of Life and Health, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Xi Li
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Aiping Zheng
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xihan Zhu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Shuting Liu
- Department of Life and Health, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Mengying Hu
- Department of Life and Health, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Qianjiang Luo
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Huina Liao
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Mubiao Liu
- Department of Obstetrics and Gynecology, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Yonghong He
- Department of Life and Health, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | | |
Collapse
|
659
|
Schmauch B, Romagnoni A, Pronier E, Saillard C, Maillé P, Calderaro J, Kamoun A, Sefta M, Toldo S, Zaslavskiy M, Clozel T, Moarii M, Courtiol P, Wainrib G. A deep learning model to predict RNA-Seq expression of tumours from whole slide images. Nat Commun 2020; 11:3877. [PMID: 32747659 PMCID: PMC7400514 DOI: 10.1038/s41467-020-17678-4] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Deep learning methods for digital pathology analysis are an effective way to address multiple clinical questions, from diagnosis to prediction of treatment outcomes. These methods have also been used to predict gene mutations from pathology images, but no comprehensive evaluation of their potential for extracting molecular features from histology slides has yet been performed. We show that HE2RNA, a model based on the integration of multiple data modes, can be trained to systematically predict RNA-Seq profiles from whole-slide images alone, without expert annotation. Through its interpretable design, HE2RNA provides virtual spatialization of gene expression, as validated by CD3- and CD20-staining on an independent dataset. The transcriptomic representation learned by HE2RNA can also be transferred on other datasets, even of small size, to increase prediction performance for specific molecular phenotypes. We illustrate the use of this approach in clinical diagnosis purposes such as the identification of tumors with microsatellite instability. RNA-sequencing of tumour tissue can provide important diagnostic and prognostic information but this is costly and not routinely performed in all clinical settings. Here, the authors show that whole slide histology slides—part of routine care—can be used to predict RNA-sequencing data and thus reduce the need for additional analyses.
Collapse
Affiliation(s)
| | | | | | | | - Pascale Maillé
- INSERM U955, Team "Pathophysiology and Therapy of Chronic Viral Hepatitis and Related Cancers", Créteil, France.,APHP, Department of Pathology, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
| | - Julien Calderaro
- INSERM U955, Team "Pathophysiology and Therapy of Chronic Viral Hepatitis and Related Cancers", Créteil, France.,APHP, Department of Pathology, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
| | | | | | | | | | | | | | | | | |
Collapse
|
660
|
Coudray N, Tsirigos A. Deep learning links histology, molecular signatures and prognosis in cancer. NATURE CANCER 2020; 1:755-757. [PMID: 35122048 PMCID: PMC11330634 DOI: 10.1038/s43018-020-0099-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Deep learning can be used to predict genomic alterations based on morphological features learned from digital histopathology. Two independent pan-cancer studies now show that automated learning from digital pathology slides and genomics can potentially decipher broader classes of molecular signatures and prognostic associations across cancer types.
Collapse
Affiliation(s)
- Nicolas Coudray
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, NY, USA.
- Skirball Institute, NYU Grossman School of Medicine, New York, NY, USA.
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, NY, USA.
- Institute for Computational Medicine, NYU Langone Health, New York, NY, USA.
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
661
|
Choi G, Kim YG, Cho H, Kim N, Lee H, Moon KC, Go H. Automated detection algorithm for C4d immunostaining showed comparable diagnostic performance to pathologists in renal allograft biopsy. Mod Pathol 2020; 33:1626-1634. [PMID: 32218521 DOI: 10.1038/s41379-020-0529-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 11/09/2022]
Abstract
A deep learning-based image analysis could improve diagnostic accuracy and efficiency in pathology work. Recently, we proposed a deep learning-based detection algorithm for C4d immunostaining in renal allografts. The objective of this study is to assess the diagnostic performance of the algorithm by comparing pathologists' diagnoses and analyzing the associations of the algorithm with clinical data. C4d immunostaining slides of renal allografts were obtained from two different institutions (100 slides from the Asan Medical Center and 86 slides from the Seoul National University Hospital) and scanned using two different slide scanners. Three pathologists and the algorithm independently evaluated each slide according to the Banff 2017 criteria. Subsequently, they jointly reviewed the results for consensus scoring. The result of the algorithm was compared with that of each pathologist and the consensus diagnosis. Clinicopathological associations of the results of the algorithm with allograft survival, histologic evidence of microvascular inflammation, and serologic results for donor-specific antibodies were also analyzed. As a result, the reproducibility between the pathologists was fair to moderate (kappa 0.36-0.54), which is comparable to that between the algorithm and each pathologist (kappa 0.34-0.51). The C4d scores predicted by the algorithm achieved substantial concordance with the consensus diagnosis (kappa = 0.61), and they were significantly associated with remarkable microvascular inflammation (P = 0.001), higher detection rate of donor-specific antibody (P = 0.003), and shorter graft survival (P < 0.001). In conclusion, the deep learning-based C4d detection algorithm showed a diagnostic performance similar to that of the pathologists.
Collapse
Affiliation(s)
- Gyuheon Choi
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Young-Gon Kim
- Department of Biomedical Engineering, Asan Institute of Life Science, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Haeyon Cho
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Namkug Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Hyunna Lee
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehang-ro, Jongro-gu, Seoul, 03080, South Korea
| | - Heounjeong Go
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
662
|
Benzekry S. Artificial Intelligence and Mechanistic Modeling for Clinical Decision Making in Oncology. Clin Pharmacol Ther 2020; 108:471-486. [PMID: 32557598 DOI: 10.1002/cpt.1951] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/04/2020] [Indexed: 12/24/2022]
Abstract
The amount of "big" data generated in clinical oncology, whether from molecular, imaging, pharmacological, or biological origin, brings novel challenges. To mine efficiently this source of information, mathematical models able to produce predictive algorithms and simulations are required, with applications for diagnosis, prognosis, drug development, or prediction of the response to therapy. Such mathematical and computational constructs can be subdivided into two broad classes: biologically agnostic, statistical models using artificial intelligence techniques, and physiologically based, mechanistic models. In this review, recent advances in the applications of such methods in clinical oncology are outlined. These include machine learning applied to big data (omics, imaging, or electronic health records), pharmacometrics and quantitative systems pharmacology, as well as tumor kinetics and metastasis modeling. Focus is set on studies with high potential of clinical translation, and particular attention is given to cancer immunotherapy. Perspectives are given in terms of combinations of the two approaches: "mechanistic learning."
Collapse
Affiliation(s)
- Sebastien Benzekry
- MONC Team, Inria Bordeaux Sud-Ouest, Talence, France
- Institut de Mathématiques de Bordeaux, CNRS UMR 5251, Bordeaux University, Talence, France
| |
Collapse
|
663
|
Deng S, Zhang X, Yan W, Chang EIC, Fan Y, Lai M, Xu Y. Deep learning in digital pathology image analysis: a survey. Front Med 2020; 14:470-487. [PMID: 32728875 DOI: 10.1007/s11684-020-0782-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/05/2020] [Indexed: 12/21/2022]
Abstract
Deep learning (DL) has achieved state-of-the-art performance in many digital pathology analysis tasks. Traditional methods usually require hand-crafted domain-specific features, and DL methods can learn representations without manually designed features. In terms of feature extraction, DL approaches are less labor intensive compared with conventional machine learning methods. In this paper, we comprehensively summarize recent DL-based image analysis studies in histopathology, including different tasks (e.g., classification, semantic segmentation, detection, and instance segmentation) and various applications (e.g., stain normalization, cell/gland/region structure analysis). DL methods can provide consistent and accurate outcomes. DL is a promising tool to assist pathologists in clinical diagnosis.
Collapse
Affiliation(s)
- Shujian Deng
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education and State Key Laboratory of Software Development Environment, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| | - Xin Zhang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education and State Key Laboratory of Software Development Environment, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| | - Wen Yan
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education and State Key Laboratory of Software Development Environment, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| | | | - Yubo Fan
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education and State Key Laboratory of Software Development Environment, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| | - Maode Lai
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, 310007, China
| | - Yan Xu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education and State Key Laboratory of Software Development Environment, Beihang University, Beijing, 100191, China.
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China.
- Microsoft Research Asia, Beijing, 100080, China.
| |
Collapse
|
664
|
Shao L, Liu Z, Feng L, Lou X, Li Z, Zhang XY, Wan X, Zhou X, Sun K, Zhang DF, Wu L, Yang G, Sun YS, Xu R, Fan X, Tian J. Multiparametric MRI and Whole Slide Image-Based Pretreatment Prediction of Pathological Response to Neoadjuvant Chemoradiotherapy in Rectal Cancer: A Multicenter Radiopathomic Study. Ann Surg Oncol 2020; 27:4296-4306. [PMID: 32729045 PMCID: PMC7497677 DOI: 10.1245/s10434-020-08659-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Indexed: 01/01/2023]
Abstract
Background The aim of this work is to combine radiological and pathological information of tumor to develop a signature for pretreatment prediction of discrepancies of pathological response at several centers and restage patients with locally advanced rectal cancer (LARC) for individualized treatment planning. Patients and Methods A total of 981 consecutive patients with evaluation of response according to tumor regression grade (TRG) who received nCRT were retrospectively recruited from four hospitals (primary cohort and external validation cohort 1–3); both pretreatment multiparametric MRI (mp-MRI) and whole slide image (WSI) of biopsy specimens were available for each patient. Quantitative image features were extracted from mp-MRI and WSI and used to construct a radiopathomics signature (RPS) powered by an artificial-intelligence model. Models based on mp-MRI or WSI alone were also constructed for comparison. Results The RPS showed overall accuracy of 79.66–87.66% in validation cohorts. The areas under the curve of RPS at specific response grades were 0.98 (TRG0), 0.93 (≤ TRG1), and 0.84 (≤ TRG2). RPS at each grade of pathological response revealed significant improvement compared with both signatures constructed without combining multiscale tumor information (P < 0.01). Moreover, RPS showed relevance to distinct probabilities of overall survival and disease-free survival in patients with LARC who underwent nCRT (P < 0.05). Conclusions The results of this study suggest that radiopathomics, combining both radiological information of the whole tumor and pathological information of local lesions from biopsy, could potentially predict discrepancies of pathological response prior to nCRT for better treatment planning. Electronic supplementary material The online version of this article (10.1245/s10434-020-08659-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lizhi Shao
- School of Computer Science and Engineering, Southeast University, Nanjing, China.,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Lili Feng
- Department of Radiation Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoying Lou
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhenhui Li
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiao-Yan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangbo Wan
- Department of Radiation Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuezhi Zhou
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China.,Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Kai Sun
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China.,Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Da-Fu Zhang
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Wu
- Department of Pathology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Guanyu Yang
- School of Computer Science and Engineering, Southeast University, Nanjing, China.,LIST, Key Laboratory of Computer Network and Information Integration, Southeast University, Ministry of Education, Nanjing, China
| | - Ying-Shi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ruihua Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Xinjuan Fan
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China. .,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China. .,Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China. .,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China.
| |
Collapse
|
665
|
Abstract
Molecular alterations in cancer can cause phenotypic changes in tumor cells and their micro-environment. Routine histopathology tissue slides - which are ubiquitously available - can reflect such morphological changes. Here, we show that deep learning can consistently infer a wide range of genetic mutations, molecular tumor subtypes, gene expression signatures and standard pathology biomarkers directly from routine histology. We developed, optimized, validated and publicly released a one-stop-shop workflow and applied it to tissue slides of more than 5000 patients across multiple solid tumors. Our findings show that a single deep learning algorithm can be trained to predict a wide range of molecular alterations from routine, paraffin-embedded histology slides stained with hematoxylin and eosin. These predictions generalize to other populations and are spatially resolved. Our method can be implemented on mobile hardware, potentially enabling point-of-care diagnostics for personalized cancer treatment. More generally, this approach could elucidate and quantify genotype-phenotype links in cancer.
Collapse
|
666
|
Pan-cancer computational histopathology reveals mutations, tumor composition and prognosis. ACTA ACUST UNITED AC 2020; 1:800-810. [DOI: 10.1038/s43018-020-0085-8] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023]
|
667
|
Zou XL, Ren Y, Feng DY, He XQ, Guo YF, Yang HL, Li X, Fang J, Li Q, Ye JJ, Han LQ, Zhang TT. A promising approach for screening pulmonary hypertension based on frontal chest radiographs using deep learning: A retrospective study. PLoS One 2020; 15:e0236378. [PMID: 32706807 PMCID: PMC7380616 DOI: 10.1371/journal.pone.0236378] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND To date, the missed diagnosis rate of pulmonary hypertension (PH) was high, and there has been limited development of a rapid, simple, and effective way to screen the disease. The purpose of this study is to develop a deep learning approach to achieve rapid detection of possible abnormalities in chest radiographs suggesting PH for screening patients suspected of PH. METHODS We retrospectively collected frontal chest radiographs and the pulmonary artery systolic pressure (PASP) value measured by Doppler transthoracic echocardiography from 762 patients (357 healthy controls and 405 with PH) from three institutes in China from January 2013 to May 2019. The wohle sample comprised 762 images (641 for training, 80 for internal test, and 41 for external test). We firstly performed a 8-fold cross-validation on the 641 images selected for training (561 for pre-training, 80 for validation), then decided to tune learning rate to 0.0008 according to the best score on validation data. Finally, we used all the pre-training and validation data (561+80 = 641) to train our models (Resnet50, Xception, and Inception V3), evaluated them on internal and external test dataset to classify the images as having manifestations of PH or healthy according to the area under the receiver operating characteristic curve (AUC/ROC). After that, the three deep learning models were further used for prediction of PASP using regression algorithm. Moreover, we invited an experienced chest radiologist to classify the images in the test dataset as having PH or not, and compared the prediction accuracy performed by deep learing models with that of manual classification. RESULTS The AUC performed by the best model (Inception V3) achieved 0.970 in the internal test, and slightly declined in the external test (0.967) when using deep learning algorithms to classify PH from normal based on chest X-rays. The mean absolute error (MAE) of the best model for prediction of PASP value was smaller in the internal test (7.45) compared to 9.95 in the external test. Manual classification of PH based on chest X-rays showed much lower AUCs compared to that performed by deep learning models both in the internal and external test. CONCLUSIONS The present study used deep learning algorithms to classify abnormalities suggesting PH in chest radiographs with high accuracy and good generalizability. Once tested prospectively in clinical settings, the technology could provide a non-invasive and easy-to-use method to screen patients suspected of having PH.
Collapse
Affiliation(s)
- Xiao-Ling Zou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, China
| | - Yong Ren
- Center for Artificial Intelligence in Medicine, Research Institute of Tsinghua, Pearl River Delta, Guangzhou, China
| | - Ding-Yun Feng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, China
| | - Xu-Qi He
- Department of Medical Ultrasound, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yue-Fei Guo
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hai-Ling Yang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, China
| | - Xian Li
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Yuedong Hospital, Meizhou, China
| | - Jia Fang
- Department of Pumonary Diseases, Dongguan Tangxia Hospital, Dongguan, China
| | - Quan Li
- Center for Artificial Intelligence in Medicine, Research Institute of Tsinghua, Pearl River Delta, Guangzhou, China
| | - Jun-Jie Ye
- Center for Artificial Intelligence in Medicine, Research Institute of Tsinghua, Pearl River Delta, Guangzhou, China
| | - Lan-Qing Han
- Center for Artificial Intelligence in Medicine, Research Institute of Tsinghua, Pearl River Delta, Guangzhou, China
| | - Tian-Tuo Zhang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
668
|
Automated identification of chest radiographs with referable abnormality with deep learning: need for recalibration. Eur Radiol 2020; 30:6902-6912. [PMID: 32661584 DOI: 10.1007/s00330-020-07062-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/07/2020] [Accepted: 07/01/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVES To evaluate the calibration of a deep learning (DL) model in a diagnostic cohort and to improve model's calibration through recalibration procedures. METHODS Chest radiographs (CRs) from 1135 consecutive patients (M:F = 582:553; mean age, 52.6 years) who visited our emergency department were included. A commercialized DL model was utilized to identify abnormal CRs, with a continuous probability score for each CR. After evaluation of the model calibration, eight different methods were used to recalibrate the original model based on the probability score. The original model outputs were recalibrated using 681 randomly sampled CRs and validated using the remaining 454 CRs. The Brier score for overall performance, average and maximum calibration error, absolute Spiegelhalter's Z for calibration, and area under the receiver operating characteristic curve (AUROC) for discrimination were evaluated in 1000-times repeated, randomly split datasets. RESULTS The original model tended to overestimate the likelihood for the presence of abnormalities, exhibiting average and maximum calibration error of 0.069 and 0.179, respectively; an absolute Spiegelhalter's Z value of 2.349; and an AUROC of 0.949. After recalibration, significant improvements in the average (range, 0.015-0.036) and maximum (range, 0.057-0.172) calibration errors were observed in eight and five methods, respectively. Significant improvement in absolute Spiegelhalter's Z (range, 0.809-4.439) was observed in only one method (the recalibration constant). Discriminations were preserved in six methods (AUROC, 0.909-0.949). CONCLUSION The calibration of DL algorithm can be augmented through simple recalibration procedures. Improved calibration may enhance the interpretability and credibility of the model for users. KEY POINTS • A deep learning model tended to overestimate the likelihood of the presence of abnormalities in chest radiographs. • Simple recalibration of the deep learning model using output scores could improve the calibration of model while maintaining discrimination. • Improved calibration of a deep learning model may enhance the interpretability and the credibility of the model for users.
Collapse
|
669
|
Yu S, Li H, Li X, Fu YV, Liu F. Classification of pathogens by Raman spectroscopy combined with generative adversarial networks. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 726:138477. [PMID: 32315848 DOI: 10.1016/j.scitotenv.2020.138477] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 06/11/2023]
Abstract
Rapid identification of marine pathogens is very important in marine ecology. Artificial intelligence combined with Raman spectroscopy is a promising choice for identifying marine pathogens due to its rapidity and efficiency. However, considering the cost of sample collection and the challenging nature of the experimental environment, only limited spectra are typically available to build a classification model, which hinders qualitative analysis. In this paper, we propose a novel method to classify marine pathogens by means of Raman spectroscopy combined with generative adversarial networks (GANs). Three marine strains, namely, Staphylococcus hominis, Vibrio alginolyticus, and Bacillus licheniformis, were cultured. Using Raman spectroscopy, we acquired 100 spectra of each strain, and we fitted them into GAN models for training. After 30,000 training iterations, the spectra generated by G were similar to the actual spectra, and D was used to test the accuracy of the spectra. Our results demonstrate that our method not only improves the accuracy of machine learning classification but also solves the problem of requiring a large amount of training data. Moreover, we have attempted to find potential identifying regions in the Raman spectra that can be used for reference in subsequent related work in this field. Therefore, this method has tremendous potential to be developed as a tool for pathogen identification.
Collapse
Affiliation(s)
- Shixiang Yu
- Key Laboratory of Coastal Biology and Biological Resources Utilization, CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, P. R. China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Hanfei Li
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xin Li
- Key Laboratory of Coastal Biology and Biological Resources Utilization, CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, P. R. China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yu Vincent Fu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China.
| | - Fanghua Liu
- Key Laboratory of Coastal Biology and Biological Resources Utilization, CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, P. R. China; National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Guangdong Institute of Eco-environmental Science & Technology, Guangdong Academy of Sciences, Guangzhou 510650, PR China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China.
| |
Collapse
|
670
|
Thakur N, Yoon H, Chong Y. Current Trends of Artificial Intelligence for Colorectal Cancer Pathology Image Analysis: A Systematic Review. Cancers (Basel) 2020; 12:E1884. [PMID: 32668721 PMCID: PMC7408874 DOI: 10.3390/cancers12071884] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers requiring early pathologic diagnosis using colonoscopy biopsy samples. Recently, artificial intelligence (AI) has made significant progress and shown promising results in the field of medicine despite several limitations. We performed a systematic review of AI use in CRC pathology image analysis to visualize the state-of-the-art. Studies published between January 2000 and January 2020 were searched in major online databases including MEDLINE (PubMed, Cochrane Library, and EMBASE). Query terms included "colorectal neoplasm," "histology," and "artificial intelligence." Of 9000 identified studies, only 30 studies consisting of 40 models were selected for review. The algorithm features of the models were gland segmentation (n = 25, 62%), tumor classification (n = 8, 20%), tumor microenvironment characterization (n = 4, 10%), and prognosis prediction (n = 3, 8%). Only 20 gland segmentation models met the criteria for quantitative analysis, and the model proposed by Ding et al. (2019) performed the best. Studies with other features were in the elementary stage, although most showed impressive results. Overall, the state-of-the-art is promising for CRC pathological analysis. However, datasets in most studies had relatively limited scale and quality for clinical application of this technique. Future studies with larger datasets and high-quality annotations are required for routine practice-level validation.
Collapse
Affiliation(s)
- Nishant Thakur
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Korea;
| | - Hongjun Yoon
- AI Lab, Deepnoid, #1305 E&C Venture Dream Tower 2, 55, Digital-ro 33-Gil, Guro-gu, Seoul 06216, Korea;
| | - Yosep Chong
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Korea;
| |
Collapse
|
671
|
Tolkach Y, Dohmgörgen T, Toma M, Kristiansen G. High-accuracy prostate cancer pathology using deep learning. NAT MACH INTELL 2020. [DOI: 10.1038/s42256-020-0200-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
672
|
Yamamoto H, Watanabe Y, Maehata T, Imai K, Itoh F. Microsatellite instability in cancer: a novel landscape for diagnostic and therapeutic approach. Arch Toxicol 2020; 94:3349-3357. [DOI: 10.1007/s00204-020-02833-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
|
673
|
Panayides AS, Amini A, Filipovic ND, Sharma A, Tsaftaris SA, Young A, Foran D, Do N, Golemati S, Kurc T, Huang K, Nikita KS, Veasey BP, Zervakis M, Saltz JH, Pattichis CS. AI in Medical Imaging Informatics: Current Challenges and Future Directions. IEEE J Biomed Health Inform 2020; 24:1837-1857. [PMID: 32609615 PMCID: PMC8580417 DOI: 10.1109/jbhi.2020.2991043] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This paper reviews state-of-the-art research solutions across the spectrum of medical imaging informatics, discusses clinical translation, and provides future directions for advancing clinical practice. More specifically, it summarizes advances in medical imaging acquisition technologies for different modalities, highlighting the necessity for efficient medical data management strategies in the context of AI in big healthcare data analytics. It then provides a synopsis of contemporary and emerging algorithmic methods for disease classification and organ/ tissue segmentation, focusing on AI and deep learning architectures that have already become the de facto approach. The clinical benefits of in-silico modelling advances linked with evolving 3D reconstruction and visualization applications are further documented. Concluding, integrative analytics approaches driven by associate research branches highlighted in this study promise to revolutionize imaging informatics as known today across the healthcare continuum for both radiology and digital pathology applications. The latter, is projected to enable informed, more accurate diagnosis, timely prognosis, and effective treatment planning, underpinning precision medicine.
Collapse
|
674
|
Becker JU, Mayerich D, Padmanabhan M, Barratt J, Ernst A, Boor P, Cicalese PA, Mohan C, Nguyen HV, Roysam B. Artificial intelligence and machine learning in nephropathology. Kidney Int 2020; 98:65-75. [PMID: 32475607 PMCID: PMC8906056 DOI: 10.1016/j.kint.2020.02.027] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/03/2020] [Accepted: 02/12/2020] [Indexed: 12/15/2022]
Abstract
Artificial intelligence (AI) for the purpose of this review is an umbrella term for technologies emulating a nephropathologist's ability to extract information on diagnosis, prognosis, and therapy responsiveness from native or transplant kidney biopsies. Although AI can be used to analyze a wide variety of biopsy-related data, this review focuses on whole slide images traditionally used in nephropathology. AI applications in nephropathology have recently become available through several advancing technologies, including (i) widespread introduction of glass slide scanners, (ii) data servers in pathology departments worldwide, and (iii) through greatly improved computer hardware to enable AI training. In this review, we explain how AI can enhance the reproducibility of nephropathology results for certain parameters in the context of precision medicine using advanced architectures, such as convolutional neural networks, that are currently the state of the art in machine learning software for this task. Because AI applications in nephropathology are still in their infancy, we show the power and potential of AI applications mostly in the example of oncopathology. Moreover, we discuss the technological obstacles as well as the current stakeholder and regulatory concerns about developing AI applications in nephropathology from the perspective of nephropathologists and the wider nephrology community. We expect the gradual introduction of these technologies into routine diagnostics and research for selective tasks, suggesting that this technology will enhance the performance of nephropathologists rather than making them redundant.
Collapse
Affiliation(s)
- Jan U Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany.
| | - David Mayerich
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, USA
| | - Meghana Padmanabhan
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, USA
| | - Jonathan Barratt
- The Mayer IgA Nephropathy Laboratories, Department of Cardiovascular, University of Leicester, Leicester, UK
| | - Angela Ernst
- Faculty of Medicine, Institute of Medical Statistics and Computational Biology, University of Cologne, Cologne, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen, Germany; Department of Nephrology, RWTH Aachen, Germany
| | | | - Chandra Mohan
- College of Engineering, University of Houston, Houston, Texas, USA
| | - Hien V Nguyen
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, USA
| | - Badrinath Roysam
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, USA
| |
Collapse
|
675
|
Acs B, Rantalainen M, Hartman J. Artificial intelligence as the next step towards precision pathology. J Intern Med 2020; 288:62-81. [PMID: 32128929 DOI: 10.1111/joim.13030] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/16/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022]
Abstract
Pathology is the cornerstone of cancer care. The need for accuracy in histopathologic diagnosis of cancer is increasing as personalized cancer therapy requires accurate biomarker assessment. The appearance of digital image analysis holds promise to improve both the volume and precision of histomorphological evaluation. Recently, machine learning, and particularly deep learning, has enabled rapid advances in computational pathology. The integration of machine learning into routine care will be a milestone for the healthcare sector in the next decade, and histopathology is right at the centre of this revolution. Examples of potential high-value machine learning applications include both model-based assessment of routine diagnostic features in pathology, and the ability to extract and identify novel features that provide insights into a disease. Recent groundbreaking results have demonstrated that applications of machine learning methods in pathology significantly improves metastases detection in lymph nodes, Ki67 scoring in breast cancer, Gleason grading in prostate cancer and tumour-infiltrating lymphocyte (TIL) scoring in melanoma. Furthermore, deep learning models have also been demonstrated to be able to predict status of some molecular markers in lung, prostate, gastric and colorectal cancer based on standard HE slides. Moreover, prognostic (survival outcomes) deep neural network models based on digitized HE slides have been demonstrated in several diseases, including lung cancer, melanoma and glioma. In this review, we aim to present and summarize the latest developments in digital image analysis and in the application of artificial intelligence in diagnostic pathology.
Collapse
Affiliation(s)
- B Acs
- From the, Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - M Rantalainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - J Hartman
- From the, Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
676
|
Xie R, Li J, Wang J, Dai W, Leier A, Marquez-Lago TT, Akutsu T, Lithgow T, Song J, Zhang Y. DeepVF: a deep learning-based hybrid framework for identifying virulence factors using the stacking strategy. Brief Bioinform 2020; 22:5864586. [PMID: 32599617 DOI: 10.1093/bib/bbaa125] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Virulence factors (VFs) enable pathogens to infect their hosts. A wealth of individual, disease-focused studies has identified a wide variety of VFs, and the growing mass of bacterial genome sequence data provides an opportunity for computational methods aimed at predicting VFs. Despite their attractive advantages and performance improvements, the existing methods have some limitations and drawbacks. Firstly, as the characteristics and mechanisms of VFs are continually evolving with the emergence of antibiotic resistance, it is more and more difficult to identify novel VFs using existing tools that were previously developed based on the outdated data sets; secondly, few systematic feature engineering efforts have been made to examine the utility of different types of features for model performances, as the majority of tools only focused on extracting very few types of features. By addressing the aforementioned issues, the accuracy of VF predictors can likely be significantly improved. This, in turn, would be particularly useful in the context of genome wide predictions of VFs. In this work, we present a deep learning (DL)-based hybrid framework (termed DeepVF) that is utilizing the stacking strategy to achieve more accurate identification of VFs. Using an enlarged, up-to-date dataset, DeepVF comprehensively explores a wide range of heterogeneous features with popular machine learning algorithms. Specifically, four classical algorithms, including random forest, support vector machines, extreme gradient boosting and multilayer perceptron, and three DL algorithms, including convolutional neural networks, long short-term memory networks and deep neural networks are employed to train 62 baseline models using these features. In order to integrate their individual strengths, DeepVF effectively combines these baseline models to construct the final meta model using the stacking strategy. Extensive benchmarking experiments demonstrate the effectiveness of DeepVF: it achieves a more accurate and stable performance compared with baseline models on the benchmark dataset and clearly outperforms state-of-the-art VF predictors on the independent test. Using the proposed hybrid ensemble model, a user-friendly online predictor of DeepVF (http://deepvf.erc.monash.edu/) is implemented. Furthermore, its utility, from the user's viewpoint, is compared with that of existing toolkits. We believe that DeepVF will be exploited as a useful tool for screening and identifying potential VFs from protein-coding gene sequences in bacterial genomes.
Collapse
Affiliation(s)
- Ruopeng Xie
- Bioinformatics Lab at Guilin University of Electronic Technology
| | - Jiahui Li
- Bioinformatics Lab at Guilin University of Electronic Technology
| | - Jiawei Wang
- Biomedicine Discovery Institute and the Department of Microbiology at Monash University, Australia
| | - Wei Dai
- School of Computer Science and Information Security, Guilin University of Electronic Technology, China
| | - André Leier
- Department of Genetics and the Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham (UAB) School of Medicine, USA
| | - Tatiana T Marquez-Lago
- Department of Genetics and the Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham (UAB) School of Medicine, USA
| | | | - Trevor Lithgow
- Biomedicine Discovery Institute and the Director of the Centre to Impact AMR at Monash University, Australia
| | - Jiangning Song
- Group Leader in the Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Yanju Zhang
- Leiden Institute of Advanced Computer Science, Leiden University
| |
Collapse
|
677
|
Zhang F, Yao S, Li Z, Liang C, Zhao K, Huang Y, Gao Y, Qu J, Li Z, Liu Z. Predicting treatment response to neoadjuvant chemoradiotherapy in local advanced rectal cancer by biopsy digital pathology image features. Clin Transl Med 2020; 10:e110. [PMID: 32594660 PMCID: PMC7403709 DOI: 10.1002/ctm2.110] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022] Open
Affiliation(s)
- Fang Zhang
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China.,Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Su Yao
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhi Li
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Ke Zhao
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Gao
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Jinrong Qu
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospita, Yunnan Cancer Center, Kunming, Yunnan, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
678
|
Kallenbach-Thieltges A, Großerueschkamp F, Jütte H, Kuepper C, Reinacher-Schick A, Tannapfel A, Gerwert K. Label-free, automated classification of microsatellite status in colorectal cancer by infrared imaging. Sci Rep 2020; 10:10161. [PMID: 32576892 PMCID: PMC7311536 DOI: 10.1038/s41598-020-67052-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
Challenging histopathological diagnostics in cancer include microsatellite instability-high (MSI-H) colorectal cancer (CRC), which occurs in 15% of early-stage CRC and is caused by a deficiency in the mismatch repair system. The diagnosis of MSI-H cannot be reliably achieved by visual inspection of a hematoxylin and eosin stained thin section alone, but additionally requires subsequent molecular analysis. Time- and sample-intensive immunohistochemistry with subsequent fragment length analysis is used. The aim of the presented feasibility study is to test the ability of quantum cascade laser (QCL)-based infrared (IR) imaging as an alternative diagnostic tool for MSI-H in CRC. We analyzed samples from 100 patients with sporadic CRC UICC stage II and III. Forty samples were used to develop the random forest classifier and 60 samples to verify the results on an independent blinded dataset. Specifically, 100% sensitivity and 93% specificity were achieved based on the independent 30 MSI-H- and 30 microsatellite stable (MSS)-patient validation cohort. This showed that QCL-based IR imaging is able to distinguish between MSI-H and MSS for sporadic CRC - a question that goes beyond morphological features - based on the use of spatially resolved infrared spectra used as biomolecular fingerprints.
Collapse
Affiliation(s)
- Angela Kallenbach-Thieltges
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany.,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany
| | - Frederik Großerueschkamp
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany.,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany
| | - Hendrik Jütte
- Institute of Pathology, Ruhr University Bochum, Bochum, Germany
| | - Claus Kuepper
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany.,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany
| | - Anke Reinacher-Schick
- Department of Hematology, Oncology and Palliative Care, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | | | - Klaus Gerwert
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany. .,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany.
| |
Collapse
|
679
|
Kriegsmann M, Haag C, Weis CA, Steinbuss G, Warth A, Zgorzelski C, Muley T, Winter H, Eichhorn ME, Eichhorn F, Kriegsmann J, Christopolous P, Thomas M, Witzens-Harig M, Sinn P, von Winterfeld M, Heussel CP, Herth FJF, Klauschen F, Stenzinger A, Kriegsmann K. Deep Learning for the Classification of Small-Cell and Non-Small-Cell Lung Cancer. Cancers (Basel) 2020; 12:cancers12061604. [PMID: 32560475 PMCID: PMC7352768 DOI: 10.3390/cancers12061604] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022] Open
Abstract
Reliable entity subtyping is paramount for therapy stratification in lung cancer. Morphological evaluation remains the basis for entity subtyping and directs the application of additional methods such as immunohistochemistry (IHC). The decision of whether to perform IHC for subtyping is subjective, and access to IHC is not available worldwide. Thus, the application of additional methods to support morphological entity subtyping is desirable. Therefore, the ability of convolutional neuronal networks (CNNs) to classify the most common lung cancer subtypes, pulmonary adenocarcinoma (ADC), pulmonary squamous cell carcinoma (SqCC), and small-cell lung cancer (SCLC), was evaluated. A cohort of 80 ADC, 80 SqCC, 80 SCLC, and 30 skeletal muscle specimens was assembled; slides were scanned; tumor areas were annotated; image patches were extracted; and cases were randomly assigned to a training, validation or test set. Multiple CNN architectures (VGG16, InceptionV3, and InceptionResNetV2) were trained and optimized to classify the four entities. A quality control (QC) metric was established. An optimized InceptionV3 CNN architecture yielded the highest classification accuracy and was used for the classification of the test set. Image patch and patient-based CNN classification results were 95% and 100% in the test set after the application of strict QC. Misclassified cases mainly included ADC and SqCC. The QC metric identified cases that needed further IHC for definite entity subtyping. The study highlights the potential and limitations of CNN image classification models for tumor differentiation.
Collapse
Affiliation(s)
- Mark Kriegsmann
- Institute of Pathology, Heidelberg University, 69120 Heidelberg, Germany; (C.H.); (G.S.); (C.Z.); (P.S.); (M.v.W.); (A.S.)
- Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (T.M.); (H.W.); (M.E.E.); (F.E.); (P.C.); (M.T.); (C.P.H.); (F.J.F.H.)
- Correspondence: (M.K.); (K.K.); Tel.: +49-6221-56-36930 (M.K.); +49-6221-56-37238 (K.K.)
| | - Christian Haag
- Institute of Pathology, Heidelberg University, 69120 Heidelberg, Germany; (C.H.); (G.S.); (C.Z.); (P.S.); (M.v.W.); (A.S.)
- Department Hematology, Oncology and Rheumatology, Heidelberg University, 69120 Heidelberg, Germany
| | - Cleo-Aron Weis
- Institute of Pathology, University Medical Centre Mannheim, Heidelberg University, 68782 Mannheim, Germany;
| | - Georg Steinbuss
- Institute of Pathology, Heidelberg University, 69120 Heidelberg, Germany; (C.H.); (G.S.); (C.Z.); (P.S.); (M.v.W.); (A.S.)
- Department Hematology, Oncology and Rheumatology, Heidelberg University, 69120 Heidelberg, Germany
| | - Arne Warth
- Institute of Pathology, Cytopathology, and Molecular Pathology, UEGP MVZ Gießen/Wetzlar/Limburg, 65549 Limburg, Germany;
| | - Christiane Zgorzelski
- Institute of Pathology, Heidelberg University, 69120 Heidelberg, Germany; (C.H.); (G.S.); (C.Z.); (P.S.); (M.v.W.); (A.S.)
| | - Thomas Muley
- Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (T.M.); (H.W.); (M.E.E.); (F.E.); (P.C.); (M.T.); (C.P.H.); (F.J.F.H.)
- Department of Thoracic Surgery, Thoraxklinik, Heidelberg University, 69126 Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (T.M.); (H.W.); (M.E.E.); (F.E.); (P.C.); (M.T.); (C.P.H.); (F.J.F.H.)
- Department of Thoracic Surgery, Thoraxklinik, Heidelberg University, 69126 Heidelberg, Germany
| | - Martin E. Eichhorn
- Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (T.M.); (H.W.); (M.E.E.); (F.E.); (P.C.); (M.T.); (C.P.H.); (F.J.F.H.)
- Department of Thoracic Surgery, Thoraxklinik, Heidelberg University, 69126 Heidelberg, Germany
| | - Florian Eichhorn
- Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (T.M.); (H.W.); (M.E.E.); (F.E.); (P.C.); (M.T.); (C.P.H.); (F.J.F.H.)
- Department of Thoracic Surgery, Thoraxklinik, Heidelberg University, 69126 Heidelberg, Germany
| | - Joerg Kriegsmann
- Molecular Pathology Trier, 54296 Trier, Germany;
- Danube Private University Krems, 3500 Krems, Austria
| | - Petros Christopolous
- Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (T.M.); (H.W.); (M.E.E.); (F.E.); (P.C.); (M.T.); (C.P.H.); (F.J.F.H.)
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University, 69126 Heidelberg, Germany
| | - Michael Thomas
- Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (T.M.); (H.W.); (M.E.E.); (F.E.); (P.C.); (M.T.); (C.P.H.); (F.J.F.H.)
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University, 69126 Heidelberg, Germany
| | | | - Peter Sinn
- Institute of Pathology, Heidelberg University, 69120 Heidelberg, Germany; (C.H.); (G.S.); (C.Z.); (P.S.); (M.v.W.); (A.S.)
| | - Moritz von Winterfeld
- Institute of Pathology, Heidelberg University, 69120 Heidelberg, Germany; (C.H.); (G.S.); (C.Z.); (P.S.); (M.v.W.); (A.S.)
| | - Claus Peter Heussel
- Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (T.M.); (H.W.); (M.E.E.); (F.E.); (P.C.); (M.T.); (C.P.H.); (F.J.F.H.)
- Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik, Heidelberg University, 69120 Heidelberg, Germany
- Department of Diagnostic and Interventional Radiology, Thoraxklinik, Heidelberg University, 69120 Heidelberg, Germany
| | - Felix J. F. Herth
- Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (T.M.); (H.W.); (M.E.E.); (F.E.); (P.C.); (M.T.); (C.P.H.); (F.J.F.H.)
- Department of Pneumology and Critical Care Medicine, Thoraxklinik, Heidelberg University, 69126 Heidelberg, Germany
| | | | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University, 69120 Heidelberg, Germany; (C.H.); (G.S.); (C.Z.); (P.S.); (M.v.W.); (A.S.)
- Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (T.M.); (H.W.); (M.E.E.); (F.E.); (P.C.); (M.T.); (C.P.H.); (F.J.F.H.)
| | - Katharina Kriegsmann
- Department Hematology, Oncology and Rheumatology, Heidelberg University, 69120 Heidelberg, Germany
- Correspondence: (M.K.); (K.K.); Tel.: +49-6221-56-36930 (M.K.); +49-6221-56-37238 (K.K.)
| |
Collapse
|
680
|
Classification and mutation prediction based on histopathology H&E images in liver cancer using deep learning. NPJ Precis Oncol 2020; 4:14. [PMID: 32550270 PMCID: PMC7280520 DOI: 10.1038/s41698-020-0120-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common subtype of liver cancer, and assessing its histopathological grade requires visual inspection by an experienced pathologist. In this study, the histopathological H&E images from the Genomic Data Commons Databases were used to train a neural network (inception V3) for automatic classification. According to the evaluation of our model by the Matthews correlation coefficient, the performance level was close to the ability of a 5-year experience pathologist, with 96.0% accuracy for benign and malignant classification, and 89.6% accuracy for well, moderate, and poor tumor differentiation. Furthermore, the model was trained to predict the ten most common and prognostic mutated genes in HCC. We found that four of them, including CTNNB1, FMN2, TP53, and ZFX4, could be predicted from histopathology images, with external AUCs from 0.71 to 0.89. The findings demonstrated that convolutional neural networks could be used to assist pathologists in the classification and detection of gene mutation in liver cancer.
Collapse
|
681
|
She Y, Jin Z, Wu J, Deng J, Zhang L, Su H, Jiang G, Liu H, Xie D, Cao N, Ren Y, Chen C. Development and Validation of a Deep Learning Model for Non-Small Cell Lung Cancer Survival. JAMA Netw Open 2020; 3:e205842. [PMID: 32492161 PMCID: PMC7272121 DOI: 10.1001/jamanetworkopen.2020.5842] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IMPORTANCE There is a lack of studies exploring the performance of a deep learning survival neural network in non-small cell lung cancer (NSCLC). OBJECTIVES To compare the performances of DeepSurv, a deep learning survival neural network with a tumor, node, and metastasis staging system in the prediction of survival and test the reliability of individual treatment recommendations provided by the deep learning survival neural network. DESIGN, SETTING, AND PARTICIPANTS In this population-based cohort study, a deep learning-based algorithm was developed and validated using consecutive cases of newly diagnosed stages I to IV NSCLC between January 2010 and December 2015 in a Surveillance, Epidemiology, and End Results database. A total of 127 features, including patient characteristics, tumor stage, and treatment strategies, were assessed for analysis. The algorithm was externally validated on an independent test cohort, comprising 1182 patients with stage I to III NSCLC diagnosed between January 2009 and December 2013 in Shanghai Pulmonary Hospital. Analysis began January 2018 and ended June 2019. MAIN OUTCOMES AND MEASURES The deep learning survival neural network model was compared with the tumor, node, and metastasis staging system for lung cancer-specific survival. The C statistic was used to assess the performance of models. A user-friendly interface was provided to facilitate the survival predictions and treatment recommendations of the deep learning survival neural network model. RESULTS Of 17 322 patients with NSCLC included in the study, 13 361 (77.1%) were white and the median (interquartile range) age was 68 (61-74) years. The majority of tumors were stage I disease (10 273 [59.3%]) and adenocarcinoma (11 985 [69.2%]). The median (interquartile range) follow-up time was 24 (10-43) months. There were 3119 patients who had lung cancer-related death during the follow-up period. The deep learning survival neural network model showed more promising results in the prediction of lung cancer-specific survival than the tumor, node, and metastasis stage on the test data set (C statistic = 0.739 vs 0.706). The population who received the recommended treatments had superior survival rates than those who received treatments not recommended (hazard ratio, 2.99; 95% CI, 2.49-3.59; P < .001), which was verified by propensity score-matched groups. The deep learning survival neural network model visualization was realized by a user-friendly graphic interface. CONCLUSIONS AND RELEVANCE The deep learning survival neural network model shows potential benefits in prognostic evaluation and treatment recommendation with respect to lung cancer-specific survival. This novel analytical approach may provide reliable individual survival information and treatment recommendations.
Collapse
Affiliation(s)
- Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhuochen Jin
- College of Design and Innovation, Tongji University, Shanghai, China
| | - Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiajun Deng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hang Su
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haipeng Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Nan Cao
- College of Design and Innovation, Tongji University, Shanghai, China
- Computer Science, NYU Shanghai, Shanghai, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
682
|
Inge L, Dennis E. Development and applications of computer image analysis algorithms for scoring of PD-L1 immunohistochemistry. ACTA ACUST UNITED AC 2020; 6:2-8. [PMID: 35757235 PMCID: PMC9216464 DOI: 10.1016/j.iotech.2020.04.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors targeting programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) have rapidly become integral to standard-of-care therapy for non-small cell lung cancer and other cancers. Immunohistochemical (IHC) staining of PD-L1 is currently the accepted and approved diagnostic assay for selecting patients for PD-L1/PD-1 axis therapies in certain indications. However, the inherent biological complexity of PD-L1 and the availability of several PD-L1 assays – each with different detection systems, platforms, scoring algorithms and cut-offs – have created challenges to ensure reliable and reproducible results based on subjective visual assessment by pathologists. The increasing adoption of computer technologies into the daily workflow of pathology provides an opportunity to leverage these tools towards improving the clinical value of PD-L1 IHC assays. This review describes several image analysis software programs of computer-aided PD-L1 scoring in the hope of driving further discussion and technological advancement in digital pathology and artificial intelligence approaches, particularly as precision medicine evolves to encompass accurate simultaneous assessment of multiple features of cancer cells and their interactions with the tumor microenvironment.
Collapse
|
683
|
Wang Y, Zhang J, Chen X, Gao L. Circ_0001023 Promotes Proliferation and Metastasis of Gastric Cancer Cells Through miR-409-3p/PHF10 Axis. Onco Targets Ther 2020; 13:4533-4544. [PMID: 32547084 PMCID: PMC7250310 DOI: 10.2147/ott.s244358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
Background Circular RNAs (circRNAs) have been well documented to regulate the gene expression via sponging microRNA (miRNA) in diverse neoplasms including gastric cancer (GC). Methods In the present study, the expressions of circ_0001023, miR-409-3p, and plant homeodomain finger 10 (PHF10) in GC tissues were detected by qRT-PCR. Chi-square test was performed to analyze the associations between circ_0001023 and pathological parameters. Cell Counting Kit-8 assay, colony formation assay, flow cytometry, and transwell assay were adopted to detect the role of circ_0001023/miR-409-3p axis in the proliferation, apoptosis, and migration of GC cells, respectively. The targeting relationship between circ_0001023 and miR-409-3p was investigated by dual-luciferase gene reporter gene assay. Additionally, subcutaneous xenotransplanted tumor model in nude mice was established to detect the function of circ_0001023 on GC growth in vivo. Results Compared with adjacent tissues, the expression of circ_0001023 was significantly upregulated and correlated with lymph node invasion and higher T stage of GC patients. It has also been proved that circ_0001023 could target miR-409-3p. Silencing circ_0001023 can impede the proliferation of GC cells and promote apoptosis, while miR-409-3p inhibitors can partially reverse the biological behavior of GC cells mentioned above. Moreover, the expression of circ_0001023 was reversely associated with miR-409-3p expression but positively correlated with PHF10, a downstream oncogene of miR-409-3p. Conclusion Collectively, it is concluded that circ_0001023 promotes the progression of GC via regulating miR-409-3p/PHF10 axis.
Collapse
Affiliation(s)
- Yongxiang Wang
- Department of Abdominal Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, Zhejiang Province, People's Republic of China
| | - Jianbin Zhang
- Department of Oncology, Zhejiang Provincial People's Hospital, Hangzhou 310022, Zhejiang Province, People's Republic of China.,Department of Oncology, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, People's Republic of China
| | - Xiaochen Chen
- Department of Oncology, Zhejiang Provincial People's Hospital, Hangzhou 310022, Zhejiang Province, People's Republic of China.,Department of Oncology, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, People's Republic of China
| | - Liang Gao
- Department of Oncology, Zhejiang Provincial People's Hospital, Hangzhou 310022, Zhejiang Province, People's Republic of China.,Department of Oncology, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, People's Republic of China
| |
Collapse
|
684
|
Hekler A, Kather JN, Krieghoff-Henning E, Utikal JS, Meier F, Gellrich FF, Upmeier Zu Belzen J, French L, Schlager JG, Ghoreschi K, Wilhelm T, Kutzner H, Berking C, Heppt MV, Haferkamp S, Sondermann W, Schadendorf D, Schilling B, Izar B, Maron R, Schmitt M, Fröhling S, Lipka DB, Brinker TJ. Effects of Label Noise on Deep Learning-Based Skin Cancer Classification. Front Med (Lausanne) 2020; 7:177. [PMID: 32435646 PMCID: PMC7218064 DOI: 10.3389/fmed.2020.00177] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/16/2020] [Indexed: 11/19/2022] Open
Abstract
Recent studies have shown that deep learning is capable of classifying dermatoscopic images at least as well as dermatologists. However, many studies in skin cancer classification utilize non-biopsy-verified training images. This imperfect ground truth introduces a systematic error, but the effects on classifier performance are currently unknown. Here, we systematically examine the effects of label noise by training and evaluating convolutional neural networks (CNN) with 804 images of melanoma and nevi labeled either by dermatologists or by biopsy. The CNNs are evaluated on a test set of 384 images by means of 4-fold cross validation comparing the outputs with either the corresponding dermatological or the biopsy-verified diagnosis. With identical ground truths of training and test labels, high accuracies with 75.03% (95% CI: 74.39–75.66%) for dermatological and 73.80% (95% CI: 73.10–74.51%) for biopsy-verified labels can be achieved. However, if the CNN is trained and tested with different ground truths, accuracy drops significantly to 64.53% (95% CI: 63.12–65.94%, p < 0.01) on a non-biopsy-verified and to 64.24% (95% CI: 62.66–65.83%, p < 0.01) on a biopsy-verified test set. In conclusion, deep learning methods for skin cancer classification are highly sensitive to label noise and future work should use biopsy-verified training images to mitigate this problem.
Collapse
Affiliation(s)
- Achim Hekler
- National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Jakob N Kather
- National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Department of Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Eva Krieghoff-Henning
- National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Jochen S Utikal
- Department of Dermatology, Heidelberg University, Mannheim, Germany.,Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
| | - Friedegund Meier
- Skin Cancer Center at the University Cancer Centre and National Center for Tumor Diseases Dresden, Dresden, Germany.,Department of Dermatology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Frank F Gellrich
- Skin Cancer Center at the University Cancer Centre and National Center for Tumor Diseases Dresden, Dresden, Germany.,Department of Dermatology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Lars French
- Department of Dermatology and Allergology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Justin G Schlager
- Department of Dermatology and Allergology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tabea Wilhelm
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heinz Kutzner
- Dermatopathology Laboratory, Friedrichshafen, Germany
| | - Carola Berking
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Markus V Heppt
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Wiebke Sondermann
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Bastian Schilling
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Benjamin Izar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Roman Maron
- National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Max Schmitt
- National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Stefan Fröhling
- National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel B Lipka
- National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Medicine, Medical Center, Otto-von-Guericke-University, Magdeburg, Germany
| | - Titus J Brinker
- National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
685
|
Woerl AC, Eckstein M, Geiger J, Wagner DC, Daher T, Stenzel P, Fernandez A, Hartmann A, Wand M, Roth W, Foersch S. Deep Learning Predicts Molecular Subtype of Muscle-invasive Bladder Cancer from Conventional Histopathological Slides. Eur Urol 2020; 78:256-264. [PMID: 32354610 DOI: 10.1016/j.eururo.2020.04.023] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/10/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Muscle-invasive bladder cancer (MIBC) is the second most common genitourinary malignancy, and is associated with high morbidity and mortality. Recently, molecular subtypes of MIBC have been identified, which have important clinical implications. OBJECTIVE In the current study, we tried to predict the molecular subtype of MIBC samples from conventional histomorphology alone using deep learning. DESIGN, SETTING, AND PARTICIPANTS Two cohorts of patients with MIBC were used: (1) The Cancer Genome Atlas Urothelial Bladder Carcinoma dataset including 407 patients and (2) our own cohort including 16 patients with treatment-naïve, primary resected MIBC. This resulted in a total of 423 digital whole slide images of tumor tissue to train, validate, and test the deep learning algorithm to predict the molecular subtype. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Various accuracy measurements including the area under the receiver operating characteristic curves were used to evaluate the deep learning model. A sliding window approach to visualize classification was used. Class activation maps were used to identify image features that are most relevant to call a specific class. RESULTS AND LIMITATIONS The deep learning model showed great performance in the prediction of the molecular subtype of MIBC patients from hematoxylin and eosin (HE) slides alone-similar to or better than pathology experts. Using different visualization techniques, we identified new histopathological features that were most relevant to our model. CONCLUSIONS Deep learning can be used to predict important molecular features in MIBC patients from HE slides alone, potentially improving the clinical management of this disease significantly. PATIENT SUMMARY In patients with bladder cancer, a computer program found changes in the appearance of tumor tissue under the microscope and used these to predict genetic alterations. This could potentially benefit patients.
Collapse
Affiliation(s)
- Ann-Christin Woerl
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany; Institute of Computer Science, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Josephine Geiger
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany; Institute of Computer Science, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Daniel C Wagner
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Tamas Daher
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Philipp Stenzel
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Aurélie Fernandez
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wand
- Institute of Computer Science, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
686
|
Shenoy S. Cell plasticity in cancer: A complex interplay of genetic, epigenetic mechanisms and tumor micro-environment. Surg Oncol 2020; 34:154-162. [PMID: 32891322 DOI: 10.1016/j.suronc.2020.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/13/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023]
Abstract
Cell plasticity, also known as lineage plasticity is defined as the ability of a cell to reprogram and change its phenotype identity. Cell plasticity is context dependent and occurs during the development of an embryo, tissue regeneration, wound healing. However when deregulated and aberrant it also contributes to cancer initiation, progression, metastases and resistance to therapies. Tumors cells exhibit varying forms of cell plasticity in each stage of the disease to evade normal regulation as would have occurred in normal cell division and homeostasis. Current evidence demonstrates complex interplay between the genes, epigenes, tumor microenvironment and the EMT in cell reprogramming and cancer cell plasticity. Herein we present experimental evidence and evolving new developments in cell plasticity in cancer cells. Additionally "Deregulated/aberrant/hijacked cell plasticity" could be considered as an additional hallmark of a cancer. In the future, combining the advances in next generation sequencing and single cell RNA techniques with evolving AI (artificial intelligence) technologies such as deep learning techniques may predict the trajectories of cancer cells and assist in navigating through the complex intricacies of the cancers. A durable, precise, personalized oncologic treatment could be a reality.
Collapse
Affiliation(s)
- Santosh Shenoy
- Clinical Associate Professor of Surgery, Department of Surgery, Kansas City VA Medical Center, University of Missouri Kansas City, USA; Cancer Biology and Therapeutics, HMS High-Impact Cancer Research (HI-CR) Program, Harvard Medical School 2018-2019, USA.
| |
Collapse
|
687
|
Computational analysis of pathological images enables a better diagnosis of TFE3 Xp11.2 translocation renal cell carcinoma. Nat Commun 2020; 11:1778. [PMID: 32286325 PMCID: PMC7156652 DOI: 10.1038/s41467-020-15671-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 03/23/2020] [Indexed: 12/17/2022] Open
Abstract
TFE3 Xp11.2 translocation renal cell carcinoma (TFE3-RCC) generally progresses more aggressively compared with other RCC subtypes, but it is challenging to diagnose TFE3-RCC by traditional visual inspection of pathological images. In this study, we collect hematoxylin and eosin- stained histopathology whole-slide images of 74 TFE3-RCC cases (the largest cohort to date) and 74 clear cell RCC cases (ccRCC, the most common RCC subtype) with matched gender and tumor grade. An automatic computational pipeline is implemented to extract image features. Comparative study identifies 52 image features with significant differences between TFE3-RCC and ccRCC. Machine learning models are built to distinguish TFE3-RCC from ccRCC. Tests of the classification models on an external validation set reveal high accuracy with areas under ROC curve ranging from 0.842 to 0.894. Our results suggest that automatically derived image features can capture subtle morphological differences between TFE3-RCC and ccRCC and contribute to a potential guideline for TFE3-RCC diagnosis. Translocation renal cell carcinoma is an aggressive form of renal cancer that is often misdiagnosed to other subtypes. Here the authors demonstrated that by using machine learning and H&E stained whole-slide images, an accurate diagnose of this particular type of renal cancer can be achieved.
Collapse
|
688
|
Les cancers colorectaux héréditaires vus par le pathologiste. Ann Pathol 2020; 40:105-113. [PMID: 32249104 DOI: 10.1016/j.annpat.2020.02.017] [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: 02/04/2020] [Revised: 02/11/2020] [Accepted: 02/17/2020] [Indexed: 11/20/2022]
Abstract
The pathologist's role in the management of hereditary colorectal cancer is important. The pathologist may suspect a familial cancer when particular morphological and/or clinical criteria are present or give a response to a clinical request in the context of a possible hereditary cancer. In this setting, the pathologist's conclusions have necessarily to be integrated to a precise environment, and if needed, followed by an oncogenetic consultation and a germline mutation research. The aim of this article is to present the main aspects of hereditary colon cancers that a pathologist may see, but also to highlight the histopathological characteristics and the place of the pathologist in the management of these different entities.
Collapse
|
689
|
Jiang Y, Yang M, Wang S, Li X, Sun Y. Emerging role of deep learning-based artificial intelligence in tumor pathology. Cancer Commun (Lond) 2020; 40:154-166. [PMID: 32277744 PMCID: PMC7170661 DOI: 10.1002/cac2.12012] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
The development of digital pathology and progression of state-of-the-art algorithms for computer vision have led to increasing interest in the use of artificial intelligence (AI), especially deep learning (DL)-based AI, in tumor pathology. The DL-based algorithms have been developed to conduct all kinds of work involved in tumor pathology, including tumor diagnosis, subtyping, grading, staging, and prognostic prediction, as well as the identification of pathological features, biomarkers and genetic changes. The applications of AI in pathology not only contribute to improve diagnostic accuracy and objectivity but also reduce the workload of pathologists and subsequently enable them to spend additional time on high-level decision-making tasks. In addition, AI is useful for pathologists to meet the requirements of precision oncology. However, there are still some challenges relating to the implementation of AI, including the issues of algorithm validation and interpretability, computing systems, the unbelieving attitude of pathologists, clinicians and patients, as well as regulators and reimbursements. Herein, we present an overview on how AI-based approaches could be integrated into the workflow of pathologists and discuss the challenges and perspectives of the implementation of AI in tumor pathology.
Collapse
Affiliation(s)
- Yahui Jiang
- Department of PathologyKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerNational Clinical Research Center for CancerTianjin Cancer Institute and HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Meng Yang
- Department Epidemiology and BiostatisticsKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerNational Clinical Research Center for CancerTianjin Cancer Institute and HospitalTianjin Medical UniversityTianjin300060P.R. China
| | - Shuhao Wang
- Institute for Interdisciplinary Information SciencesTsinghua UniversityBeijing100084P. R. China
| | - Xiangchun Li
- Department Epidemiology and BiostatisticsKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerNational Clinical Research Center for CancerTianjin Cancer Institute and HospitalTianjin Medical UniversityTianjin300060P.R. China
| | - Yan Sun
- Department of PathologyKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerNational Clinical Research Center for CancerTianjin Cancer Institute and HospitalTianjin Medical UniversityTianjin300060P. R. China
| |
Collapse
|
690
|
Gonçalves WGE, dos Santos MHDP, Lobato FMF, Ribeiro-dos-Santos Â, de Araújo GS. Deep learning in gastric tissue diseases: a systematic review. BMJ Open Gastroenterol 2020; 7:e000371. [PMID: 32337060 PMCID: PMC7170401 DOI: 10.1136/bmjgast-2019-000371] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/14/2020] [Accepted: 02/24/2020] [Indexed: 12/24/2022] Open
Abstract
Background In recent years, deep learning has gained remarkable attention in medical image analysis due to its capacity to provide results comparable to specialists and, in some cases, surpass them. Despite the emergence of deep learning research on gastric tissues diseases, few intensive reviews are addressing this topic. Method We performed a systematic review related to applications of deep learning in gastric tissue disease analysis by digital histology, endoscopy and radiology images. Conclusions This review highlighted the high potential and shortcomings in deep learning research studies applied to gastric cancer, ulcer, gastritis and non-malignant diseases. Our results demonstrate the effectiveness of gastric tissue analysis by deep learning applications. Moreover, we also identified gaps of evaluation metrics, and image collection availability, therefore, impacting experimental reproducibility.
Collapse
Affiliation(s)
- Wanderson Gonçalves e Gonçalves
- Laboratório de Genética Humana e Médica - Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, Brazil
| | | | | | - Ândrea Ribeiro-dos-Santos
- Laboratório de Genética Humana e Médica - Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Gilderlanio Santana de Araújo
- Laboratório de Genética Humana e Médica - Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| |
Collapse
|
691
|
Li L, Feng Q, Wang X. PreMSIm: An R package for predicting microsatellite instability from the expression profiling of a gene panel in cancer. Comput Struct Biotechnol J 2020; 18:668-675. [PMID: 32257050 PMCID: PMC7113609 DOI: 10.1016/j.csbj.2020.03.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 01/10/2023] Open
Abstract
Microsatellite instability (MSI) is a genomic property of the cancers with defective DNA mismatch repair and is a useful marker for cancer diagnosis and treatment in diverse cancer types. In particular, MSI has been associated with the active immune checkpoint blockade therapy response in cancer. Most of computational methods for predicting MSI are based on DNA sequencing data and a few are based on mRNA expression data. Using the RNA-Seq pan-cancer datasets for three cancer cohorts (colon, gastric, and endometrial cancers) from The Cancer Genome Atlas (TCGA) program, we developed an algorithm (PreMSIm) for predicting MSI from the expression profiling of a 15-gene panel in cancer. We demonstrated that PreMSIm had high prediction performance in predicting MSI in most cases using both RNA-Seq and microarray gene expression datasets. Moreover, PreMSIm displayed superior or comparable performance versus other DNA or mRNA-based methods. We conclude that PreMSIm has the potential to provide an alternative approach for identifying MSI in cancer.
Collapse
Key Words
- ACC, adrenocortical carcinoma
- AUC, area under the curve
- Algorithm
- BLCA, bladder urothelial carcinoma
- BRCA, breast invasive carcinoma
- CESC, cervical squamous cell carcinoma and endocervical adenocarcinoma
- CHOL, cholangiocarcinoma
- COAD, colon adenocarcinoma
- CV, cross validation
- Cancer
- Classification
- DLBC, lymphoid neoplasm diffuse large B-cell lymphoma
- ESCA, esophageal carcinoma
- GBM, glioblastoma multiforme
- GEO, Gene Expression Omnibus
- GO, gene ontology
- Gene expression profiling
- HNSC, head and neck squamous cell carcinoma
- KICH, kidney chromophobe
- KIRC, kidney renal clear cell carcinoma
- KIRP, kidney renal papillary cell carcinoma
- LAML, acute myeloid leukemia
- LGG, brain lower grade glioma
- LIHC, liver hepatocellular carcinoma
- LUAD, lung adenocarcinoma
- LUSC, lung squamous cell carcinoma
- MESO, mesothelioma
- MSI, microsatellite instability
- MSS, microsatellite stability
- Machine learning
- Microsatellite instability
- OV, ovarian serous cystadenocarcinoma
- PAAD, pancreatic adenocarcinoma
- PCPG, pheochromocytoma and paraganglioma
- PPI, protein-protein interaction
- PRAD, prostate adenocarcinoma
- READ, rectum adenocarcinoma
- RF, random forest
- ROC, receiver operating characteristic
- SARC, sarcoma
- SKCM, skin cutaneous melanoma
- STAD, stomach adenocarcinoma
- SVM, support vector machine
- TCGA, The Cancer Genome Atlas
- TGCT, testicular germ cell tumors
- THCA, thyroid carcinoma
- THYM, thymoma
- UCEC, uterine corpus endometrial carcinoma
- UCS, uterine carcinosarcoma
- UVM, uveal melanoma
- XGBoost, extreme gradient boosting
- k-NN, k-nearest neighbor
Collapse
Affiliation(s)
- Lin Li
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Qiushi Feng
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
692
|
Refolo MG, Lotesoriere C, Messa C, Caruso MG, D'Alessandro R. Integrated immune gene expression signature and molecular classification in gastric cancer: New insights. J Leukoc Biol 2020; 108:633-646. [PMID: 32170872 DOI: 10.1002/jlb.4mr0120-221r] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/03/2020] [Accepted: 01/24/2020] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is characterized by extreme heterogeneity due to histopathological differences, molecular characteristics, and immune gene expression signature. Until recently, several targeted therapies failed due to this complexity. The recent immunotherapy resulted in more effective and safe approaches in several malignancies. All tumors could be considered potentially immunogenic and the new knowledge regarding the interactions among tumor cells, immune cells, and tumor microenvironment (TME) allowed to reverse possible immune resistance. The immune response is a complex multisteps process that finely regulates the balance between the recognition of non-self and the prevention of autoimmunity. Cancer cells can use these pathways to suppress tumor immunity as a major mechanism of immune resistance. The recent molecular classifications of GCs by The Cancer Genome Atlas (TCGA) and by the Asian Cancer Research (ACRG) networks, together with the identification of multiple biomarkers, open new perspectives for stratification of patients who might benefit from a long-term immune checkpoint therapy. One of the major processes that contribute to an immunosuppressive microenvironment is represented by tumor angiogenesis. The cellular mechanisms inducing both angiogenesis and immunosuppressive responses are often reached by the same cell types and soluble factors, such as vascular endothelial growth factor A (VEGFA). Recent studies point out that combinatorial strategies should be adapted as useful therapeutic approach to reverse the immunosuppressive status of microenvironment occurring in a relevant percentage of gastric tumors.
Collapse
Affiliation(s)
- Maria Grazia Refolo
- Laboratory of Cellular and Molecular Biology, Department of Clinical Pathology, Castellana Grotte, Bari, Italy
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Bari, Italy
| | - Caterina Messa
- Laboratory of Cellular and Molecular Biology, Department of Clinical Pathology, Castellana Grotte, Bari, Italy
| | - Maria Gabriella Caruso
- Ambulatory of Clinical Nutrition, National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Bari, Italy
| | - Rosalba D'Alessandro
- Laboratory of Cellular and Molecular Biology, Department of Clinical Pathology, Castellana Grotte, Bari, Italy
| |
Collapse
|
693
|
Zheng H, Momeni A, Cedoz PL, Vogel H, Gevaert O. Whole slide images reflect DNA methylation patterns of human tumors. NPJ Genom Med 2020; 5:11. [PMID: 32194984 PMCID: PMC7064513 DOI: 10.1038/s41525-020-0120-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
DNA methylation is an important epigenetic mechanism regulating gene expression and its role in carcinogenesis has been extensively studied. High-throughput DNA methylation assays have been used broadly in cancer research. Histopathology images are commonly obtained in cancer treatment, given that tissue sampling remains the clinical gold-standard for diagnosis. In this work, we investigate the interaction between cancer histopathology images and DNA methylation profiles to provide a better understanding of tumor pathobiology at the epigenetic level. We demonstrate that classical machine learning algorithms can associate the DNA methylation profiles of cancer samples with morphometric features extracted from whole slide images. Furthermore, grouping the genes into methylation clusters greatly improves the performance of the models. The well-predicted genes are enriched in key pathways in carcinogenesis including hypoxia in glioma and angiogenesis in renal cell carcinoma. Our results provide new insights into the link between histopathological and molecular data.
Collapse
Affiliation(s)
- Hong Zheng
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, Stanford, USA
| | - Alexandre Momeni
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, Stanford, USA
| | - Pierre-Louis Cedoz
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, Stanford, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University, Stanford, USA
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, Stanford, USA
- Department of Biomedical Data Science, Stanford University, Stanford, USA
| |
Collapse
|
694
|
Huss R, Coupland SE. Software‐assisted decision support in digital histopathology. J Pathol 2020; 250:685-692. [DOI: 10.1002/path.5388] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/09/2020] [Accepted: 01/17/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Ralf Huss
- Institute of Pathology and Molecular Diagnostics University Hospital Augsburg Augsburg Germany
| | - Sarah E Coupland
- Department of Cellular and Molecular Pathology University of Liverpool Liverpool UK
| |
Collapse
|
695
|
Scanning electron microscopy and machine learning reveal heterogeneity in capsular morphotypes of the human pathogen Cryptococcus spp. Sci Rep 2020; 10:2362. [PMID: 32047210 PMCID: PMC7012869 DOI: 10.1038/s41598-020-59276-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/16/2020] [Indexed: 01/09/2023] Open
Abstract
Phenotypic heterogeneity is an important trait for the development and survival of many microorganisms including the yeast Cryptococcus spp., a deadly pathogen spread worldwide. Here, we have applied scanning electron microscopy (SEM) to define four Cryptococcus spp. capsule morphotypes, namely Regular, Spiky, Bald, and Phantom. These morphotypes were persistently observed in varying proportions among yeast isolates. To assess the distribution of such morphotypes we implemented an automated pipeline capable of (1) identifying potentially cell-associated objects in the SEM-derived images; (2) computing object-level features; and (3) classifying these objects into their corresponding classes. The machine learning approach used a Random Forest (RF) classifier whose overall accuracy reached 85% on the test dataset, with per-class specificity above 90%, and sensitivity between 66 and 94%. Additionally, the RF model indicates that structural and texture features, e.g., object area, eccentricity, and contrast, are most relevant for classification. The RF results agree with the observed variation in these features, consistently also with visual inspection of SEM images. Finally, our work introduces morphological variants of Cryptococcus spp. capsule. These can be promptly identified and characterized using computational models so that future work may unveil morphological associations with yeast virulence.
Collapse
|
696
|
Katoh M, Katoh M. Precision medicine for human cancers with Notch signaling dysregulation (Review). Int J Mol Med 2020; 45:279-297. [PMID: 31894255 PMCID: PMC6984804 DOI: 10.3892/ijmm.2019.4418] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
NOTCH1, NOTCH2, NOTCH3 and NOTCH4 are transmembrane receptors that transduce juxtacrine signals of the delta‑like canonical Notch ligand (DLL)1, DLL3, DLL4, jagged canonical Notch ligand (JAG)1 and JAG2. Canonical Notch signaling activates the transcription of BMI1 proto‑oncogene polycomb ring finger, cyclin D1, CD44, cyclin dependent kinase inhibitor 1A, hes family bHLH transcription factor 1, hes related family bHLH transcription factor with YRPW motif 1, MYC, NOTCH3, RE1 silencing transcription factor and transcription factor 7 in a cellular context‑dependent manner, while non‑canonical Notch signaling activates NF‑κB and Rac family small GTPase 1. Notch signaling is aberrantly activated in breast cancer, non‑small‑cell lung cancer and hematological malignancies, such as T‑cell acute lymphoblastic leukemia and diffuse large B‑cell lymphoma. However, Notch signaling is inactivated in small‑cell lung cancer and squamous cell carcinomas. Loss‑of‑function NOTCH1 mutations are early events during esophageal tumorigenesis, whereas gain‑of‑function NOTCH1 mutations are late events during T‑cell leukemogenesis and B‑cell lymphomagenesis. Notch signaling cascades crosstalk with fibroblast growth factor and WNT signaling cascades in the tumor microenvironment to maintain cancer stem cells and remodel the tumor microenvironment. The Notch signaling network exerts oncogenic and tumor‑suppressive effects in a cancer stage‑ or (sub)type‑dependent manner. Small‑molecule γ‑secretase inhibitors (AL101, MRK‑560, nirogacestat and others) and antibody‑based biologics targeting Notch ligands or receptors [ABT‑165, AMG 119, rovalpituzumab tesirine (Rova‑T) and others] have been developed as investigational drugs. The DLL3‑targeting antibody‑drug conjugate (ADC) Rova‑T, and DLL3‑targeting chimeric antigen receptor‑modified T cells (CAR‑Ts), AMG 119, are promising anti‑cancer therapeutics, as are other ADCs or CAR‑Ts targeting tumor necrosis factor receptor superfamily member 17, CD19, CD22, CD30, CD79B, CD205, Claudin 18.2, fibroblast growth factor receptor (FGFR)2, FGFR3, receptor‑type tyrosine‑protein kinase FLT3, HER2, hepatocyte growth factor receptor, NECTIN4, inactive tyrosine‑protein kinase 7, inactive tyrosine‑protein kinase transmembrane receptor ROR1 and tumor‑associated calcium signal transducer 2. ADCs and CAR‑Ts could alter the therapeutic framework for refractory cancers, especially diffuse‑type gastric cancer, ovarian cancer and pancreatic cancer with peritoneal dissemination. Phase III clinical trials of Rova‑T for patients with small‑cell lung cancer and a phase III clinical trial of nirogacestat for patients with desmoid tumors are ongoing. Integration of human intelligence, cognitive computing and explainable artificial intelligence is necessary to construct a Notch‑related knowledge‑base and optimize Notch‑targeted therapy for patients with cancer.
Collapse
Affiliation(s)
| | - Masaru Katoh
- Department of Omics Network, National Cancer Center, Tokyo 104-0045, Japan
| |
Collapse
|
697
|
A High-Throughput Tumor Location System with Deep Learning for Colorectal Cancer Histopathology Image. Artif Intell Med 2020. [DOI: 10.1007/978-3-030-59137-3_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
698
|
Shen Y, Ke J. A Deformable CRF Model for Histopathology Whole-Slide Image Classification. MEDICAL IMAGE COMPUTING AND COMPUTER ASSISTED INTERVENTION – MICCAI 2020 2020. [DOI: 10.1007/978-3-030-59722-1_48] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
699
|
Fridman WH, Miller I, Sautès-Fridman C, Byrne AT. Therapeutic Targeting of the Colorectal Tumor Stroma. Gastroenterology 2020; 158:303-321. [PMID: 31622621 DOI: 10.1053/j.gastro.2019.09.045] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/05/2019] [Accepted: 09/08/2019] [Indexed: 02/07/2023]
Abstract
Colorectal tumors have been classified based on histologic factors, genetic factors, and consensus molecular subtypes, all of which affect the tumor microenvironment. Elements of the tumor microenvironment serve as therapeutic targets and might be used as prognostic factors. For example, immune checkpoint inhibitors are used to treat tumors with microsatellite instability, and anti-angiogenic agents may be used in combination with other drugs to slow or inhibit tumor growth. We review the features of the colorectal tumor stroma that are associated with patient outcomes and discuss potential therapeutic agents that target these features.
Collapse
Affiliation(s)
- Wolf H Fridman
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Inflammation, Complement and Cancer Team, Paris, France.
| | - Ian Miller
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Catherine Sautès-Fridman
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Inflammation, Complement and Cancer Team, Paris, France
| | - Annette T Byrne
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
700
|
Mann SA, Cheng L. Microsatellite instability and mismatch repair deficiency in the era of precision immuno-oncology. Expert Rev Anticancer Ther 2019; 20:1-4. [PMID: 31842633 DOI: 10.1080/14737140.2020.1705789] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Steven A Mann
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|