1
|
Tarakçı EA, Çeliker M, Birinci M, Yemiş T, Gül O, Oğuz EF, Solak M, Kaba E, Çeliker FB, Özergin Coşkun Z, Alkan A, Erdivanlı ÖÇ. Novel Preprocessing-Based Sequence for Comparative MR Cervical Lymph Node Segmentation. J Clin Med 2025; 14:1802. [PMID: 40142614 PMCID: PMC11943128 DOI: 10.3390/jcm14061802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Background and Objective: This study aims to utilize deep learning methods for the automatic segmentation of cervical lymph nodes in magnetic resonance images (MRIs), enhancing the speed and accuracy of diagnosing pathological masses in the neck and improving patient treatment processes. Materials and Methods: This study included 1346 MRI slices from 64 patients undergoing cervical lymph node dissection, biopsy, and preoperative contrast-enhanced neck MRI. A preprocessing model was used to crop and highlight lymph nodes, along with a method for automatic re-cropping. Two datasets were created from the cropped images-one with augmentation and one without-divided into 90% training and 10% validation sets. After preprocessing, the ResNet-50 images in the DeepLabv3+ encoder block were automatically segmented. Results: According to the results of the validation set, the mean IoU values for the DWI, T2, T1, T1+C, and ADC sequences in the dataset without augmentation created for cervical lymph node segmentation were 0.89, 0.88, 0.81, 0.85, and 0.80, respectively. In the augmented dataset, the average IoU values for all sequences were 0.91, 0.89, 0.85, 0.88, and 0.84. The DWI sequence showed the highest performance in the datasets with and without augmentation. Conclusions: Our preprocessing-based deep learning architectures successfully segmented cervical lymph nodes with high accuracy. This study is the first to explore automatic segmentation of the cervical lymph nodes using comprehensive neck MRI sequences. The proposed model can streamline the detection process, reducing the need for radiology expertise. Additionally, it offers a promising alternative to manual segmentation in radiotherapy, potentially enhancing treatment effectiveness.
Collapse
Affiliation(s)
- Elif Ayten Tarakçı
- Department of Otorhinolaryngology, Medicine Faculty, Recep Tayyip Erdoğan University, Rize 53000, Turkey; (E.A.T.); (M.B.); (T.Y.); (Z.Ö.C.); (Ö.Ç.E.)
| | - Metin Çeliker
- Department of Otorhinolaryngology, Medicine Faculty, Recep Tayyip Erdoğan University, Rize 53000, Turkey; (E.A.T.); (M.B.); (T.Y.); (Z.Ö.C.); (Ö.Ç.E.)
| | - Mehmet Birinci
- Department of Otorhinolaryngology, Medicine Faculty, Recep Tayyip Erdoğan University, Rize 53000, Turkey; (E.A.T.); (M.B.); (T.Y.); (Z.Ö.C.); (Ö.Ç.E.)
| | - Tuğba Yemiş
- Department of Otorhinolaryngology, Medicine Faculty, Recep Tayyip Erdoğan University, Rize 53000, Turkey; (E.A.T.); (M.B.); (T.Y.); (Z.Ö.C.); (Ö.Ç.E.)
| | - Oğuz Gül
- Department of Otorhinolaryngology, Akçaabat Haçkalı Baba State Hospital, Trabzon 61310, Turkey;
| | - Enes Faruk Oğuz
- Department of Biomedical Device Technology, Hassa Vocational School, Hatay Mustafa Kemal University, Hatay 31000, Turkey;
| | - Merve Solak
- Department of Radiolagy, Medicine Faculty, Recep Tayyip Erdoğan University, Rize 53000, Turkey; (M.S.); (E.K.); (F.B.Ç.)
| | - Esat Kaba
- Department of Radiolagy, Medicine Faculty, Recep Tayyip Erdoğan University, Rize 53000, Turkey; (M.S.); (E.K.); (F.B.Ç.)
| | - Fatma Beyazal Çeliker
- Department of Radiolagy, Medicine Faculty, Recep Tayyip Erdoğan University, Rize 53000, Turkey; (M.S.); (E.K.); (F.B.Ç.)
| | - Zerrin Özergin Coşkun
- Department of Otorhinolaryngology, Medicine Faculty, Recep Tayyip Erdoğan University, Rize 53000, Turkey; (E.A.T.); (M.B.); (T.Y.); (Z.Ö.C.); (Ö.Ç.E.)
| | - Ahmet Alkan
- Department of Electrical and Electronics Engineering, Kahramanmaraş Sütçü İmam University, Kahramanmaraş 46000, Turkey;
| | - Özlem Çelebi Erdivanlı
- Department of Otorhinolaryngology, Medicine Faculty, Recep Tayyip Erdoğan University, Rize 53000, Turkey; (E.A.T.); (M.B.); (T.Y.); (Z.Ö.C.); (Ö.Ç.E.)
| |
Collapse
|
2
|
Pan Y, Shi L, Liu Y, Chen JC, Qiu J. Multi-omics models for predicting prognosis in non-small cell lung cancer patients following chemotherapy and radiotherapy: A multi-center study. Radiother Oncol 2025; 204:110715. [PMID: 39800269 DOI: 10.1016/j.radonc.2025.110715] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 12/18/2024] [Accepted: 01/04/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND AND PURPOSE Quantifying tumor heterogeneity from various dimensions is crucial for precise treatment. This study aimed to develop and validate multi-omics models based on the computed tomography images, pathological images, dose and clinical information to predict treatment response and overall survival of non-small cell lung cancer (NSCLC) patients undergoing chemotherapy and radiotherapy. MATERIALS AND METHODS This retrospective study included 220 NSCLC patients from three centers. Following feature extraction and selection, single-omics and multi-omics models were built for treatment response and overall survival prediction. The performance of treatment response models was evaluated using the area under the curve (AUC) and box plots. For overall survival analysis, the model's evaluation included AUC, concordance index (C-index), Kaplan-Meier curves, and calibration curves. Shapley values were used to assess the contribution of different features to multi-omics models. RESULTS Multi-omics models consistently exhibited superior discriminative ability compared to single-omics models in predicting both treatment response and overall survival. For treatment response, the three all-modality models achieved AUC values of 0.87, 0.91, and 0.82 in the external validation set, respectively. In overall survival analysis, the three all-modality models demonstrated AUC values and C-index of 0.73/0.72, 0.80/0.77, 0.79/0.78 in the external validation set, respectively. CONCLUSION Multi-omics prediction models demonstrated superior predictive ability with robustness and interpretability. By predicting treatment response and overall survival in NSCLC patients, these models have the potential to assist clinician optimizing treatment plans, supporting individualized treatment strategies, improving the tumor control probability and prolonging the patients' survival.
Collapse
Affiliation(s)
- Yuteng Pan
- School of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Liting Shi
- School of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yuan Liu
- School of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Jyh-Cheng Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming Chiao-Tung University, Taipei, Taiwan; Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Jianfeng Qiu
- School of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China.
| |
Collapse
|
3
|
Hassan MH, Reiter E, Razzaq M. Automatic ovarian follicle detection using object detection models. Sci Rep 2024; 14:31856. [PMID: 39738599 PMCID: PMC11685387 DOI: 10.1038/s41598-024-82904-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025] Open
Abstract
Ovaries are of paramount importance in reproduction as they produce female gametes through a complex developmental process known as folliculogenesis. In the prospect of better understanding the mechanisms of folliculogenesis and of developing novel pharmacological approaches to control it, it is important to accurately and quantitatively assess the later stages of ovarian folliculogenesis (i.e. the formation of antral follicles and corpus lutea). Manual counting from histological sections is commonly employed to determine the number of these follicular structures, however it is a laborious and error prone task. In this work, we show the benefits of deep learning models for counting antral follicles and corpus lutea in ovarian histology sections. Here, we use various backbone architectures to build two one-stage object detection models, i.e. YOLO and RetinaNet. We employ transfer learning, early stopping, and data augmentation approaches to improve the generalizability of the object detectors. Furthermore, we use sampling strategy to mitigate the foreground-foreground class imbalance and focal loss to reduce the imbalance between the foreground-background classes. Our models were trained and validated using a dataset containing only 1000 images. With RetinaNet, we achieved a mean average precision of 83% whereas with YOLO of 75% on the testing dataset. Our results demonstrate that deep learning methods are useful to speed up the follicle counting process and improve accuracy by correcting manual counting errors.
Collapse
Affiliation(s)
- Maya Haj Hassan
- INRAE, CNRS, Université de Tours, PRC, Nouzilly, 37380, France
| | - Eric Reiter
- INRAE, CNRS, Université de Tours, PRC, Nouzilly, 37380, France
- Université Paris-Saclay, Inria, Inria Saclay-Île-de-France, Palaiseau, 91120, France
| | - Misbah Razzaq
- INRAE, CNRS, Université de Tours, PRC, Nouzilly, 37380, France.
| |
Collapse
|
4
|
Abe M, Kanavati F, Tsuneki M. Evaluation of a Deep Learning Model for Metastatic Squamous Cell Carcinoma Prediction From Whole Slide Images. Arch Pathol Lab Med 2024; 148:1344-1351. [PMID: 38387604 DOI: 10.5858/arpa.2023-0406-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 02/24/2024]
Abstract
CONTEXT.— Squamous cell carcinoma (SCC) is a histologic type of cancer that exhibits various degrees of keratinization. Identifying lymph node metastasis in SCC is crucial for prognosis and treatment strategies. Although artificial intelligence (AI) has shown promise in cancer prediction, applications specifically targeting SCC are limited. OBJECTIVE.— To design and validate a deep learning model tailored to predict metastatic SCC in radical lymph node dissection specimens using whole slide images (WSIs). DESIGN.— Using the EfficientNetB1 architecture, a model was trained on 6587 WSIs (2413 SCC and 4174 nonneoplastic) from several hospitals, encompassing esophagus, head and neck, lung, and skin specimens. The training exclusively relied on WSI-level labels without annotations. We evaluated the model on a test set consisting of 541 WSIs (41 SCC and 500 nonneoplastic) of radical lymph node dissection specimens. RESULTS.— The model exhibited high performance, with receiver operating characteristic curve areas under the curve between 0.880 and 0.987 in detecting SCC metastases in lymph nodes. Although true positives and negatives were accurately identified, certain limitations were observed. These included false positives due to germinal centers, dust cell aggregations, and specimen-handling artifacts, as well as false negatives due to poor differentiation. CONCLUSIONS.— The developed artificial intelligence model presents significant potential in enhancing SCC lymph node detection, offering workload reduction for pathologists and increasing diagnostic efficiency. Continuous refinement is needed to overcome existing challenges, making the model more robust and clinically relevant.
Collapse
Affiliation(s)
- Makoto Abe
- From the Department of Pathology, Tochigi Cancer Center, Tochigi, Japan (Abe)
| | - Fahdi Kanavati
- Medmain Research, Medmain Inc, Fukuoka, Japan (Kanavati, Tsuneki)
| | - Masayuki Tsuneki
- Medmain Research, Medmain Inc, Fukuoka, Japan (Kanavati, Tsuneki)
| |
Collapse
|
5
|
Budginaite E, Magee DR, Kloft M, Woodruff HC, Grabsch HI. Computational methods for metastasis detection in lymph nodes and characterization of the metastasis-free lymph node microarchitecture: A systematic-narrative hybrid review. J Pathol Inform 2024; 15:100367. [PMID: 38455864 PMCID: PMC10918266 DOI: 10.1016/j.jpi.2024.100367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 03/09/2024] Open
Abstract
Background Histological examination of tumor draining lymph nodes (LNs) plays a vital role in cancer staging and prognostication. However, as soon as a LN is classed as metastasis-free, no further investigation will be performed and thus, potentially clinically relevant information detectable in tumor-free LNs is currently not captured. Objective To systematically study and critically assess methods for the analysis of digitized histological LN images described in published research. Methods A systematic search was conducted in several public databases up to December 2023 using relevant search terms. Studies using brightfield light microscopy images of hematoxylin and eosin or immunohistochemically stained LN tissue sections aiming to detect and/or segment LNs, their compartments or metastatic tumor using artificial intelligence (AI) were included. Dataset, AI methodology, cancer type, and study objective were compared between articles. Results A total of 7201 articles were collected and 73 articles remained for detailed analyses after article screening. Of the remaining articles, 86% aimed at LN metastasis identification, 8% aimed at LN compartment segmentation, and remaining focused on LN contouring. Furthermore, 78% of articles used patch classification and 22% used pixel segmentation models for analyses. Five out of six studies (83%) of metastasis-free LNs were performed on publicly unavailable datasets, making quantitative article comparison impossible. Conclusions Multi-scale models mimicking multiple microscopy zooms show promise for computational LN analysis. Large-scale datasets are needed to establish the clinical relevance of analyzing metastasis-free LN in detail. Further research is needed to identify clinically interpretable metrics for LN compartment characterization.
Collapse
Affiliation(s)
- Elzbieta Budginaite
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | - Maximilian Kloft
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Internal Medicine, Justus-Liebig-University, Giessen, Germany
| | - Henry C. Woodruff
- Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Heike I. Grabsch
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, UK
| |
Collapse
|
6
|
Shahadat N, Lama R, Nguyen A. Lung and Colon Cancer Detection Using a Deep AI Model. Cancers (Basel) 2024; 16:3879. [PMID: 39594834 PMCID: PMC11592951 DOI: 10.3390/cancers16223879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Lung and colon cancers are among the leading causes of cancer-related mortality worldwide. Early and accurate detection of these cancers is crucial for effective treatment and improved patient outcomes. False or incorrect detection is harmful. Accurately detecting cancer in a patient's tissue is crucial to their effective treatment. While analyzing tissue samples is complicated and time-consuming, deep learning techniques have made it possible to complete this process more efficiently and accurately. As a result, researchers can study more patients in a shorter amount of time and at a lower cost. Much research has been conducted to investigate deep learning models that require great computational ability and resources. However, none of these have had a 100% accurate detection rate for these life-threatening malignancies. Misclassified or falsely detecting cancer can have very harmful consequences. This research proposes a new lightweight, parameter-efficient, and mobile-embedded deep learning model based on a 1D convolutional neural network with squeeze-and-excitation layers for efficient lung and colon cancer detection. This proposed model diagnoses and classifies lung squamous cell carcinomas and adenocarcinoma of the lung and colon from digital pathology images. Extensive experiment demonstrates that our proposed model achieves 100% accuracy for detecting lung, colon, and lung and colon cancers from the histopathological (LC25000) lung and colon datasets, which is considered the best accuracy for around 0.35 million trainable parameters and around 6.4 million flops. Compared with the existing results, our proposed architecture shows state-of-the-art performance in lung, colon, and lung and colon cancer detection.
Collapse
Affiliation(s)
- Nazmul Shahadat
- Department of Computer and Data Sciences, Truman State University, Kirksville, MO 63501, USA; (R.L.); (A.N.)
| | | | | |
Collapse
|
7
|
Caranfil E, Lami K, Uegami W, Fukuoka J. Artificial Intelligence and Lung Pathology. Adv Anat Pathol 2024; 31:344-351. [PMID: 38780094 DOI: 10.1097/pap.0000000000000448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
This manuscript provides a comprehensive overview of the application of artificial intelligence (AI) in lung pathology, particularly in the diagnosis of lung cancer. It discusses various AI models designed to support pathologists and clinicians. AI models supporting pathologists are to standardize diagnosis, score PD-L1 status, supporting tumor cellularity count, and indicating explainability for pathologic judgements. Several models predict outcomes beyond pathologic diagnosis and predict clinical outcomes like patients' survival and molecular alterations. The manuscript emphasizes the potential of AI to enhance accuracy and efficiency in pathology, while also addressing the challenges and future directions for integrating AI into clinical practice.
Collapse
Affiliation(s)
- Emanuel Caranfil
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
| | - Kris Lami
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
| | - Wataru Uegami
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
- Department of Pathology, Kameda Medical Center, Kamogawa, Japan
| | - Junya Fukuoka
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
- Department of Pathology, Kameda Medical Center, Kamogawa, Japan
| |
Collapse
|
8
|
Yin Y, Wang Y, Yu X, Li Y, Zhao Y, Liu Z. Overactivation of XBP1 in plasma cells implies worse survival through innate immunity in esophageal squamous cell carcinoma. Cancer Lett 2024; 597:217045. [PMID: 38871246 DOI: 10.1016/j.canlet.2024.217045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
To maintain protein homeostasis, X-box binding protein 1 (XBP1) undergoes splicing following the activation of the unfolded protein response (UPR) in response to endoplasmic reticulum (ER) stress. Although targeting ER stress represents a promising therapeutic strategy, a comprehensive understanding of XBP1 at the cellular level and the link between XBP1 and the innate nervous system is lacking. Here, TCGA pancancer datasets from 33 cancer types, scRNA pancancer datasets from 454 patients and bulk RNA-seq datasets from 155 paired esophageal squamous cell carcinoma (ESCC) patients were analyzed. To cope with ER stress, plasma cells tend to activate XBP1 after undergoing bacterial infection and inflammatory signaling from the innate immune system. Patients with high XBP1 expression in their plasma cells have a higher tumor grade and worse survival. However, activation of the innate immune system with increased XBP1 expression in plasma cells correlates with an increased lymphocyte ratio, indicative of a more robust immune response. Moreover, XBP1 activation appears to initiate leukocyte migration at the transcriptional level. Our study revealed that the XBP1-induced UPR could mediate the crosstalk between optimal acquired humoral immune responses and innate immunity in ESCC.
Collapse
Affiliation(s)
- Yin Yin
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuhao Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiao Yu
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yang Li
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yahui Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
9
|
Guo W, Lu T, Song Y, Li A, Feng X, Han D, Cao Y, Sun D, Gong X, Li C, Jin R, Du H, Chen K, Xiang J, Hang J, Chen G, Li H. Lymph node metastasis in early invasive lung adenocarcinoma: Prediction model establishment and validation based on genomic profiling and clinicopathologic characteristics. Cancer Med 2024; 13:e70039. [PMID: 39046176 PMCID: PMC11267562 DOI: 10.1002/cam4.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/22/2024] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND The presence of lymph node (LN) metastasis directly affects the treatment strategy for lung adenocarcinoma (LUAD). Next-generation sequencing (NGS) has been widely used in patients with advanced LUAD to identify targeted genes, while early detection of pathologic LN metastasis using NGS has not been assessed. METHODS Clinicopathologic features and molecular characteristics of 224 patients from Ruijin Hospital were analyzed to detect factors associated with LN metastases. Another 140 patients from Huashan Hospital were set as a test cohort. RESULTS Twenty-four out of 224 patients were found to have lymph node metastases (10.7%). Pathologic LN-positive tumors showed higher mutant allele tumor heterogeneity (p < 0.05), higher tumor mutation burden (p < 0.001), as well as more frequent KEAP1 (p = 0.001), STK11 (p = 0.004), KRAS (p = 0.007), CTNNB1 (p = 0.017), TP53, and ARID2 mutations (both p = 0.02); whereas low frequency of EGFR mutation (p = 0.005). A predictive nomogram involving male sex, solid tumor morphology, higher T stage, EGFR wild-type, and TP53, STK11, CDKN2A, KEAP1, ARID2, KRAS, SDHA, SPEN, CTNNB1, DICER1 mutations showed outstanding efficiency in both the training cohort (AUC = 0.819) and the test cohort (AUC = 0.780). CONCLUSION This study suggests that the integration of genomic profiling and clinical features identifies early-invasive LUAD patients at higher risk of LN metastasis. Improved identification of LN metastasis is beneficial for the optimization of the patient's therapy decisions.
Collapse
Affiliation(s)
- Wei Guo
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tong Lu
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yang Song
- Department of Thoracic SurgeryHuashan Hospital, Fudan UniversityShanghaiChina
| | - Anqi Li
- Department of PathologyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xijia Feng
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Dingpei Han
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuqin Cao
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Debin Sun
- Genecast Biotechnology Co., LtdWuxiChina
| | | | - Chengqiang Li
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Runsen Jin
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hailei Du
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Kai Chen
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jie Xiang
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Junbiao Hang
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Gang Chen
- Department of Thoracic SurgeryHuashan Hospital, Fudan UniversityShanghaiChina
| | - Hecheng Li
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
10
|
Pan Y, Dai H, Wang S, Wang L, Li Q, Wang W, Li J, Qi D, Yang Z, Jia J, Wang Y, Fang Q, Li L, Zhou W, Song Z, Zou S. Clinically Applicable Pan-Origin Cancer Detection for Lymph Nodes via Artificial Intelligence-Based Pathology. Pathobiology 2024; 91:345-358. [PMID: 38718783 DOI: 10.1159/000539010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/09/2024] [Indexed: 06/13/2024] Open
Abstract
INTRODUCTION Lymph node metastasis is one of the most common ways of tumour metastasis. The presence or absence of lymph node involvement influences the cancer's stage, therapy, and prognosis. The integration of artificial intelligence systems in the histopathological diagnosis of lymph nodes after surgery is urgent. METHODS Here, we propose a pan-origin lymph node cancer metastasis detection system. The system is trained by over 700 whole-slide images (WSIs) and is composed of two deep learning models to locate the lymph nodes and detect cancers. RESULTS It achieved an area under the receiver operating characteristic curve (AUC) of 0.958, with a 95.2% sensitivity and 72.2% specificity, on 1,402 WSIs from 49 organs at the National Cancer Center, China. Moreover, we demonstrated that the system could perform robustly with 1,051 WSIs from 52 organs from another medical centre, with an AUC of 0.925. CONCLUSION Our research represents a step forward in a pan-origin lymph node metastasis detection system, providing accurate pathological guidance by reducing the probability of missed diagnosis in routine clinical practice.
Collapse
Affiliation(s)
- Yi Pan
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,
| | - Hongtian Dai
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuhao Wang
- Thorough Lab, Thorough Future, Beijing, China
| | - Lang Wang
- Thorough Lab, Thorough Future, Beijing, China
| | - Qiting Li
- R&D Department, China Academy of Launch Vehicle Technology, Beijing, China
| | - Wenmiao Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangtao Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Qi
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaoyang Yang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Jia
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaxi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Fang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weixun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Zhigang Song
- Department of Pathology, The Chinese PLA General Hospital, Beijing, China
| | - Shuangmei Zou
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Wang W, Jiang F, Wu WQ, Zhu XL, Wang HX, Zhang L, Fan ZY. Identification of lymph node adulteration in minced pork by comprehensive metabolomics and lipidomics approach based on UPLC/LTQ-Orbitrap MS. J Food Sci 2024; 89:2249-2260. [PMID: 38477648 DOI: 10.1111/1750-3841.17005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/09/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024]
Abstract
The deliberate pork adulteration with lymph nodes is a common adulteration phenomenon, and it poses a serious threat to public health and food safety. An untargeted metabolomics and lipidomics approach based on ultrahigh performance liquid chromatography coupled with linear ion trap quadrupole-Orbitrap high resolution mass spectrometry (MS) was used to distinguish lymph nodes from minced pork. The principal component analysis and orthogonal projection to latent structures discriminant analysis models were established with the good of fitness and predictivity. The results showed that there were significant differences in metabolites and lipids between lymph nodes and pork. A total of 16 significantly differentiated metabolites were identified, of which 1-palmitoylglycerophosphocholine, 12,13-dihydroxy-9-octadecenoic acid, and prostaglandin E2 (PGE2) were positively correlated with lymph node content and were identified as potential markers of lymph nodes. These three markers were combined to create a binary logistic regression model, and a combined-factor exceeding 0.75 was ultimately identified as a marker for pork adulteration with lymph nodes. The desorption electrospray ionization-MS images showed that PGE2 had a higher relative abundance in the lymph node region than in adjacent non-lymph node regions, indicating that PGE2 was a marker that contributed significantly for identifying lymph nodes adulteration into pork. Our results provide a theoretical basis for identifying lymph node adulteration, which will contribute to combating fraud in the meat industry.
Collapse
Affiliation(s)
- Wei Wang
- Hubei Provincial Institute for Food Supervision and Test, Wuhan, China
- Key Laboratory of Detection Technology of Focus Chemical Hazards in Animal-derived Food for State Market Regulation, Wuhan, China
- Hubei Provincial Engineering and Technology Research Center for Food Quality and Safety Test, Wuhan, China
| | - Feng Jiang
- Hubei Provincial Institute for Food Supervision and Test, Wuhan, China
- Key Laboratory of Detection Technology of Focus Chemical Hazards in Animal-derived Food for State Market Regulation, Wuhan, China
- Hubei Provincial Engineering and Technology Research Center for Food Quality and Safety Test, Wuhan, China
| | - Wan-Qin Wu
- Hubei Provincial Institute for Food Supervision and Test, Wuhan, China
- Key Laboratory of Detection Technology of Focus Chemical Hazards in Animal-derived Food for State Market Regulation, Wuhan, China
- Hubei Provincial Engineering and Technology Research Center for Food Quality and Safety Test, Wuhan, China
| | - Xiao-Ling Zhu
- Hubei Provincial Institute for Food Supervision and Test, Wuhan, China
- Key Laboratory of Detection Technology of Focus Chemical Hazards in Animal-derived Food for State Market Regulation, Wuhan, China
- Hubei Provincial Engineering and Technology Research Center for Food Quality and Safety Test, Wuhan, China
| | - Hui-Xia Wang
- Hubei Provincial Institute for Food Supervision and Test, Wuhan, China
- Key Laboratory of Detection Technology of Focus Chemical Hazards in Animal-derived Food for State Market Regulation, Wuhan, China
- Hubei Provincial Engineering and Technology Research Center for Food Quality and Safety Test, Wuhan, China
| | - Li Zhang
- Hubei Provincial Institute for Food Supervision and Test, Wuhan, China
- Key Laboratory of Detection Technology of Focus Chemical Hazards in Animal-derived Food for State Market Regulation, Wuhan, China
- Hubei Provincial Engineering and Technology Research Center for Food Quality and Safety Test, Wuhan, China
| | - Zhi-Yong Fan
- Hubei Provincial Institute for Food Supervision and Test, Wuhan, China
- Key Laboratory of Detection Technology of Focus Chemical Hazards in Animal-derived Food for State Market Regulation, Wuhan, China
- Hubei Provincial Engineering and Technology Research Center for Food Quality and Safety Test, Wuhan, China
| |
Collapse
|
12
|
Lami K, Ota N, Yamaoka S, Bychkov A, Matsumoto K, Uegami W, Munkhdelger J, Seki K, Sukhbaatar O, Attanoos R, Berezowska S, Brcic L, Cavazza A, English JC, Fabro AT, Ishida K, Kashima Y, Kitamura Y, Larsen BT, Marchevsky AM, Miyazaki T, Morimoto S, Ozasa M, Roden AC, Schneider F, Smith ML, Tabata K, Takano AM, Tanaka T, Tsuchiya T, Nagayasu T, Sakanashi H, Fukuoka J. Standardized Classification of Lung Adenocarcinoma Subtypes and Improvement of Grading Assessment Through Deep Learning. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:2066-2079. [PMID: 37544502 DOI: 10.1016/j.ajpath.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/04/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023]
Abstract
The histopathologic distinction of lung adenocarcinoma (LADC) subtypes is subject to high interobserver variability, which can compromise the optimal assessment of patient prognosis. Therefore, this study developed convolutional neural networks capable of distinguishing LADC subtypes and predicting disease-specific survival, according to the recently established LADC tumor grades. Consensus LADC histopathologic images were obtained from 17 expert pulmonary pathologists and one pathologist in training. Two deep learning models (AI-1 and AI-2) were trained to predict eight different LADC classes. Furthermore, the trained models were tested on an independent cohort of 133 patients. The models achieved high precision, recall, and F1 scores exceeding 0.90 for most of the LADC classes. Clear stratification of the three LADC grades was reached in predicting the disease-specific survival by the two models, with both Kaplan-Meier curves showing significance (P = 0.0017 and 0.0003). Moreover, both trained models showed high stability in the segmentation of each pair of predicted grades with low variation in the hazard ratio across 200 bootstrapped samples. These findings indicate that the trained convolutional neural networks improve the diagnostic accuracy of the pathologist and refine LADC grade assessment. Thus, the trained models are promising tools that may assist in the routine evaluation of LADC subtypes and grades in clinical practice.
Collapse
Affiliation(s)
- Kris Lami
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Noriaki Ota
- Systems Research & Development Center, Technology Bureau, NS Solutions Corp., Yokohama, Japan
| | - Shinsuke Yamaoka
- Systems Research & Development Center, Technology Bureau, NS Solutions Corp., Yokohama, Japan
| | - Andrey Bychkov
- Department of Pathology, Kameda Medical Center, Kamogawa, Japan
| | - Keitaro Matsumoto
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Wataru Uegami
- Department of Pathology, Kameda Medical Center, Kamogawa, Japan
| | | | - Kurumi Seki
- Department of Pathology, Kameda Medical Center, Kamogawa, Japan
| | | | - Richard Attanoos
- Department of Cellular Pathology, Cardiff University, Cardiff, United Kingdom
| | - Sabina Berezowska
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Alberto Cavazza
- Unit of Pathologic Anatomy, Azienda USL/IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - John C English
- Department of Pathology, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Alexandre Todorovic Fabro
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kaori Ishida
- Department of Pathology, Kansai Medical University, Hirakata City, Japan
| | - Yukio Kashima
- Department of Pathology, Hyogo Prefectural Awaji Medical Center, Sumoto City, Japan
| | - Yuka Kitamura
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; N Lab Co. Ltd., Nagasaki, Japan
| | - Brandon T Larsen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona
| | | | - Takuro Miyazaki
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shimpei Morimoto
- Innovation Platform & Office for Precision Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mutsumi Ozasa
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Frank Schneider
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Maxwell L Smith
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona
| | - Kazuhiro Tabata
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Angela M Takano
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Tomonori Tanaka
- Department of Diagnostic Pathology, Kobe University Hospital, Kobe, Japan
| | - Tomoshi Tsuchiya
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takeshi Nagayasu
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hidenori Sakanashi
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Junya Fukuoka
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Department of Pathology, Kameda Medical Center, Kamogawa, Japan.
| |
Collapse
|
13
|
Nibid L, Greco C, Cordelli E, Sabarese G, Fiore M, Liu CZ, Ippolito E, Sicilia R, Miele M, Tortora M, Taffon C, Rakaee M, Soda P, Ramella S, Perrone G. Deep pathomics: A new image-based tool for predicting response to treatment in stage III non-small cell lung cancer. PLoS One 2023; 18:e0294259. [PMID: 38015944 PMCID: PMC10684067 DOI: 10.1371/journal.pone.0294259] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023] Open
Abstract
Despite the advantages offered by personalized treatments, there is presently no way to predict response to chemoradiotherapy in patients with non-small cell lung cancer (NSCLC). In this exploratory study, we investigated the application of deep learning techniques to histological tissue slides (deep pathomics), with the aim of predicting the response to therapy in stage III NSCLC. We evaluated 35 digitalized tissue slides (biopsies or surgical specimens) obtained from patients with stage IIIA or IIIB NSCLC. Patients were classified as responders (12/35, 34.7%) or non-responders (23/35, 65.7%) based on the target volume reduction shown on weekly CT scans performed during chemoradiation treatment. Digital tissue slides were tested by five pre-trained convolutional neural networks (CNNs)-AlexNet, VGG, MobileNet, GoogLeNet, and ResNet-using a leave-two patient-out cross validation approach, and we evaluated the networks' performances. GoogLeNet was globally found to be the best CNN, correctly classifying 8/12 responders and 10/11 non-responders. Moreover, Deep-Pathomics was found to be highly specific (TNr: 90.1) and quite sensitive (TPr: 0.75). Our data showed that AI could surpass the capabilities of all presently available diagnostic systems, supplying additional information beyond that currently obtainable in clinical practice. The ability to predict a patient's response to treatment could guide the development of new and more effective therapeutic AI-based approaches and could therefore be considered an effective and innovative step forward in personalised medicine.
Collapse
Affiliation(s)
- Lorenzo Nibid
- Research Unit of Anatomical Pathology, Department of of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Carlo Greco
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Ermanno Cordelli
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Giovanna Sabarese
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Michele Fiore
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Charles Z. Liu
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Edy Ippolito
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Rosa Sicilia
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Marianna Miele
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Matteo Tortora
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Taffon
- Research Unit of Anatomical Pathology, Department of of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Mehrdad Rakaee
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Paolo Soda
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Sara Ramella
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Giuseppe Perrone
- Research Unit of Anatomical Pathology, Department of of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| |
Collapse
|
14
|
Terada K, Yoshizawa A, Liu X, Ito H, Hamaji M, Menju T, Date H, Bise R, Haga H. Deep Learning for Predicting Effect of Neoadjuvant Therapies in Non-Small Cell Lung Carcinomas With Histologic Images. Mod Pathol 2023; 36:100302. [PMID: 37580019 DOI: 10.1016/j.modpat.2023.100302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/23/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023]
Abstract
Neoadjuvant therapies are used for locally advanced non-small cell lung carcinomas, whereby pathologists histologically evaluate the effect using resected specimens. Major pathological response (MPR) has recently been used for treatment evaluation and as an economical survival surrogate; however, interobserver variability and poor reproducibility are often noted. The aim of this study was to develop a deep learning (DL) model to predict MPR from hematoxylin and eosin-stained tissue images and to validate its utility for clinical use. We collected data on 125 primary non-small cell lung carcinoma cases that were resected after neoadjuvant therapy. The cases were randomly divided into 55 for training/validation and 70 for testing. A total of 261 hematoxylin and eosin-stained slides were obtained from the maximum tumor beds, and whole slide images were prepared. We used a multiscale patch model that can adaptively weight multiple convolutional neural networks trained with different field-of-view images. We performed 3-fold cross-validation to evaluate the model. During testing, we compared the percentages of viable tumor evaluated by annotator pathologists (reviewed data), those evaluated by nonannotator pathologists (primary data), and those predicted by the DL-based model using 2-class confusion matrices and receiver operating characteristic curves and performed a survival analysis between MPR-achieved and non-MPR cases. In cross-validation, accuracy and mean F1 score were 0.859 and 0.805, respectively. During testing, accuracy and mean F1 score with reviewed data and those with primary data were 0.986, 0.985, 0.943, and 0.943, respectively. The areas under the receiver operating characteristic curve with reviewed and primary data were 0.999 and 0.978, respectively. The disease-free survival of MPR-achieved cases with reviewed and primary data was significantly better than that of the non-MPR cases (P<.001 and P=.001), and that predicted by the DL-based model was almost identical (P=.005). The DL model may support pathologist evaluations and can offer accurate determinations of MPR in patients.
Collapse
Affiliation(s)
- Kazuhiro Terada
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan.
| | - Xiaoqing Liu
- Department of Advanced Information Technology, Faculty of Information Science and Electrical Engineering, Kyushu University, Fukuoka, Japan
| | - Hiroaki Ito
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Masatsugu Hamaji
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Ryoma Bise
- Department of Advanced Information Technology, Faculty of Information Science and Electrical Engineering, Kyushu University, Fukuoka, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
15
|
Liu Q, Jiang N, Hao Y, Hao C, Wang W, Bian T, Wang X, Li H, zhang Y, Kang Y, Xie F, Li Y, Jiang X, Feng Y, Mao Z, Wang Q, Gao Q, Zhang W, Cui B, Dong T. Identification of lymph node metastasis in pre-operation cervical cancer patients by weakly supervised deep learning from histopathological whole-slide biopsy images. Cancer Med 2023; 12:17952-17966. [PMID: 37559500 PMCID: PMC10523985 DOI: 10.1002/cam4.6437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Lymph node metastasis (LNM) significantly impacts the prognosis of individuals diagnosed with cervical cancer, as it is closely linked to disease recurrence and mortality, thereby impacting therapeutic schedule choices for patients. However, accurately predicting LNM prior to treatment remains challenging. Consequently, this study seeks to utilize digital pathological features extracted from histopathological slides of primary cervical cancer patients to preoperatively predict the presence of LNM. METHODS A deep learning (DL) model was trained using the Vision transformer (ViT) and recurrent neural network (RNN) frameworks to predict LNM. This prediction was based on the analysis of 554 histopathological whole-slide images (WSIs) obtained from Qilu Hospital of Shandong University. To validate the model's performance, an external test was conducted using 336 WSIs from four other hospitals. Additionally, the efficiency of the DL model was evaluated using 190 cervical biopsies WSIs in a prospective set. RESULTS In the internal test set, our DL model achieved an area under the curve (AUC) of 0.919, with sensitivity and specificity values of 0.923 and 0.905, respectively, and an accuracy (ACC) of 0.909. The performance of the DL model remained strong in the external test set. In the prospective cohort, the AUC was 0.91, and the ACC was 0.895. Additionally, the DL model exhibited higher accuracy compared to imaging examination in the evaluation of LNM. By utilizing the transformer visualization method, we generated a heatmap that illustrates the local pathological features in primary lesions relevant to LNM. CONCLUSION DL-based image analysis has demonstrated efficiency in predicting LNM in early operable cervical cancer through the utilization of biopsies WSI. This approach has the potential to enhance therapeutic decision-making for patients diagnosed with cervical cancer.
Collapse
Affiliation(s)
- Qingqing Liu
- Cheeloo College of MedicineShandong UniversityJinan CityChina
| | - Nan Jiang
- Cheeloo College of MedicineShandong UniversityJinan CityChina
| | - Yiping Hao
- Cheeloo College of MedicineShandong UniversityJinan CityChina
| | - Chunyan Hao
- Department of Pathology, School of Basic Medical Science, Cheeloo College of MedicineShandong UniversityJinan CityChina
- Department of PathologyQilu Hospital of Shandong UniversityJinan CityChina
| | - Wei Wang
- Department of PathologyAffiliated Hospital of Jining Medical UniversityJining CityChina
| | - Tingting Bian
- Department of Medical ImagingAffiliated Hospital of Jining Medical UniversityJining CityChina
| | - Xiaohong Wang
- Department of Obstetrics and GynecologyJinan People's HospitalJinan CityChina
| | - Hua Li
- Department of Obstetrics and GynecologyTai'an City Central HospitalTai'an CityChina
| | - Yan zhang
- Department of Obstetrics and GynecologyWeifang People's HospitalWeifang CityChina
| | - Yanjun Kang
- Department of Obstetrics and GynecologyWomen and Children's Hospital, Qingdao UniversityQingdao CityChina
| | - Fengxiang Xie
- Department of PathologyKingMed DiagnosticsJinan CityChina
| | - Yawen Li
- Department of PathologyQilu Hospital of Shandong UniversityJinan CityChina
| | - XuJi Jiang
- Cheeloo College of MedicineShandong UniversityJinan CityChina
| | - Yuan Feng
- Cheeloo College of MedicineShandong UniversityJinan CityChina
| | - Zhonghao Mao
- Cheeloo College of MedicineShandong UniversityJinan CityChina
| | - Qi Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Qianfoshan HospitalShandong UniversityJinan CityChina
| | - Qun Gao
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdao CityChina
| | - Wenjing Zhang
- Department of Obstetrics and GynecologyQilu Hospital of Shandong UniversityJinan CityChina
| | - Baoxia Cui
- Department of Obstetrics and GynecologyQilu Hospital of Shandong UniversityJinan CityChina
| | - Taotao Dong
- Department of Obstetrics and GynecologyQilu Hospital of Shandong UniversityJinan CityChina
| |
Collapse
|
16
|
Matsushima J, Sato T, Ohnishi T, Yoshimura Y, Mizutani H, Koto S, Ikeda JI, Kano M, Matsubara H, Hayashi H. The Use of Deep Learning-Based Computer Diagnostic Algorithm for Detection of Lymph Node Metastases of Gastric Adenocarcinoma. Int J Surg Pathol 2023; 31:975-981. [PMID: 35898183 DOI: 10.1177/10668969221113475] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Objectives. The diversifying modalities of treatment for gastric cancer raise urgent demands for the rapid and precise diagnosis of metastases in regional lymph nodes, thereby significantly impact the workload of pathologists. Meanwhile, the recent advent of whole-slide scanners and deep-learning techniques have enabled the computer-assisted analysis of histopathological images, which could help to alleviate this impact. Thus, we developed a deep learning-based diagnostic algorithm to detect lymph node metastases of gastric adenocarcinoma and evaluated its performance. Methods. We randomly selected 20 patients with gastric adenocarcinoma who underwent surgery as definitive treatment and were found to be node metastasis-positive. HEMATOXYLIN-eosin (HE) stained glass slides, including a total of 51 metastasis-positive nodes, were retrieved from the specimens of these cases. Other slides with 776 metastasis-negative nodes were also retrieved from other twenty cases with the same disease that were diagnosed as metastasis-negative by the final pathological examinations. All glass slides were digitized using a whole-slide scanner. A deep-learning algorithm to detect metastases was developed using the data in which metastasis-positive parts of the images were annotated by a well-trained pathologist, and its performance in detecting metastases was evaluated. Results. Cross-validation analysis indicated an area of 0.9994 under the receiver operating characteristic curve. Free-response receiver operating characteristic curve (FROC) analysis indicated a sensitivity of 1.00 with three false positives. Further evaluation using an independent dataset also showed similar level of accuracies. Conclusion. This deep learning-based diagnosis-aid system is a promising tool that can assist pathologists involved in gastric cancer care and reduce their workload.
Collapse
Affiliation(s)
- Jun Matsushima
- Department of Pathology, Saitama Medical Center, Dokkyo Medical University, Saitama, Japan
- Center for Frontier Medical Engineering, Chiba University, Chiba, Japan
- Department of Diagnostic Pathology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tamotsu Sato
- Toshiba Digital Solutions Corporation, Kanagawa, Japan
| | - Takashi Ohnishi
- Center for Frontier Medical Engineering, Chiba University, Chiba, Japan
| | | | | | | | - Jun-Ichiro Ikeda
- Department of Diagnostic Pathology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masayuki Kano
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hideki Hayashi
- Center for Frontier Medical Engineering, Chiba University, Chiba, Japan
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
17
|
Davri A, Birbas E, Kanavos T, Ntritsos G, Giannakeas N, Tzallas AT, Batistatou A. Deep Learning for Lung Cancer Diagnosis, Prognosis and Prediction Using Histological and Cytological Images: A Systematic Review. Cancers (Basel) 2023; 15:3981. [PMID: 37568797 PMCID: PMC10417369 DOI: 10.3390/cancers15153981] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer is one of the deadliest cancers worldwide, with a high incidence rate, especially in tobacco smokers. Lung cancer accurate diagnosis is based on distinct histological patterns combined with molecular data for personalized treatment. Precise lung cancer classification from a single H&E slide can be challenging for a pathologist, requiring most of the time additional histochemical and special immunohistochemical stains for the final pathology report. According to WHO, small biopsy and cytology specimens are the available materials for about 70% of lung cancer patients with advanced-stage unresectable disease. Thus, the limited available diagnostic material necessitates its optimal management and processing for the completion of diagnosis and predictive testing according to the published guidelines. During the new era of Digital Pathology, Deep Learning offers the potential for lung cancer interpretation to assist pathologists' routine practice. Herein, we systematically review the current Artificial Intelligence-based approaches using histological and cytological images of lung cancer. Most of the published literature centered on the distinction between lung adenocarcinoma, lung squamous cell carcinoma, and small cell lung carcinoma, reflecting the realistic pathologist's routine. Furthermore, several studies developed algorithms for lung adenocarcinoma predominant architectural pattern determination, prognosis prediction, mutational status characterization, and PD-L1 expression status estimation.
Collapse
Affiliation(s)
- Athena Davri
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece;
| | - Effrosyni Birbas
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.B.); (T.K.)
| | - Theofilos Kanavos
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.B.); (T.K.)
| | - Georgios Ntritsos
- Department of Hygiene and Epidemiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
- Department of Informatics and Telecommunications, University of Ioannina, 47100 Arta, Greece;
| | - Nikolaos Giannakeas
- Department of Informatics and Telecommunications, University of Ioannina, 47100 Arta, Greece;
| | - Alexandros T. Tzallas
- Department of Informatics and Telecommunications, University of Ioannina, 47100 Arta, Greece;
| | - Anna Batistatou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece;
| |
Collapse
|
18
|
Verghese G, Li M, Liu F, Lohan A, Kurian NC, Meena S, Gazinska P, Shah A, Oozeer A, Chan T, Opdam M, Linn S, Gillett C, Alberts E, Hardiman T, Jones S, Thavaraj S, Jones JL, Salgado R, Pinder SE, Rane S, Sethi A, Grigoriadis A. Multiscale deep learning framework captures systemic immune features in lymph nodes predictive of triple negative breast cancer outcome in large-scale studies. J Pathol 2023; 260:376-389. [PMID: 37230111 PMCID: PMC10720675 DOI: 10.1002/path.6088] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/27/2023] [Accepted: 04/11/2023] [Indexed: 05/27/2023]
Abstract
The suggestion that the systemic immune response in lymph nodes (LNs) conveys prognostic value for triple-negative breast cancer (TNBC) patients has not previously been investigated in large cohorts. We used a deep learning (DL) framework to quantify morphological features in haematoxylin and eosin-stained LNs on digitised whole slide images. From 345 breast cancer patients, 5,228 axillary LNs, cancer-free and involved, were assessed. Generalisable multiscale DL frameworks were developed to capture and quantify germinal centres (GCs) and sinuses. Cox regression proportional hazard models tested the association between smuLymphNet-captured GC and sinus quantifications and distant metastasis-free survival (DMFS). smuLymphNet achieved a Dice coefficient of 0.86 and 0.74 for capturing GCs and sinuses, respectively, and was comparable to an interpathologist Dice coefficient of 0.66 (GC) and 0.60 (sinus). smuLymphNet-captured sinuses were increased in LNs harbouring GCs (p < 0.001). smuLymphNet-captured GCs retained clinical relevance in LN-positive TNBC patients whose cancer-free LNs had on average ≥2 GCs, had longer DMFS (hazard ratio [HR] = 0.28, p = 0.02) and extended GCs' prognostic value to LN-negative TNBC patients (HR = 0.14, p = 0.002). Enlarged smuLymphNet-captured sinuses in involved LNs were associated with superior DMFS in LN-positive TNBC patients in a cohort from Guy's Hospital (multivariate HR = 0.39, p = 0.039) and with distant recurrence-free survival in 95 LN-positive TNBC patients of the Dutch-N4plus trial (HR = 0.44, p = 0.024). Heuristic scoring of subcapsular sinuses in LNs of LN-positive Tianjin TNBC patients (n = 85) cross-validated the association of enlarged sinuses with shorter DMFS (involved LNs: HR = 0.33, p = 0.029 and cancer-free LNs: HR = 0.21 p = 0.01). Morphological LN features reflective of cancer-associated responses are robustly quantifiable by smuLymphNet. Our findings further strengthen the value of assessment of LN properties beyond the detection of metastatic deposits for prognostication of TNBC patients. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Gregory Verghese
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- Breast Cancer Now Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Mengyuan Li
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Fangfang Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of EducationKey Laboratory of Cancer Prevention and TherapyTianjinPR China
| | - Amit Lohan
- Department of Electrical EngineeringIndian Institute of Technology BombayMumbaiIndia
| | - Nikhil Cherian Kurian
- Department of Electrical EngineeringIndian Institute of Technology BombayMumbaiIndia
| | - Swati Meena
- Department of Electrical EngineeringIndian Institute of Technology BombayMumbaiIndia
| | - Patrycja Gazinska
- Breast Cancer Now Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- Biobank Research GroupLukasiewicz Research Network, PORT Polish Center for Technology DevelopmentWroclawPoland
| | - Aekta Shah
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- Department of PathologyTata Memorial Centre, Tata Memorial Hospital, Homi Bhabha National InstituteMumbaiIndia
| | - Aasiyah Oozeer
- King's Health Partners Cancer Biobank, King's College LondonLondonUK
| | - Terry Chan
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Mark Opdam
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Sabine Linn
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Medical OncologyThe Netherlands Cancer Institute, Antoni van LeeuwenhoekAmsterdamThe Netherlands
- Department of PathologyUniversity Medical CentreUtrechtThe Netherlands
| | - Cheryl Gillett
- King's Health Partners Cancer Biobank, King's College LondonLondonUK
| | - Elena Alberts
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Thomas Hardiman
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Samantha Jones
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of LondonLondonUK
| | - Selvam Thavaraj
- Faculty of Dentistry, Oral & Craniofacial ScienceKing's College LondonLondonUK
- Head and Neck PathologyGuy's & St Thomas' NHS Foundation TrustLondonUK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of LondonLondonUK
| | - Roberto Salgado
- Department of PathologyGZA‐ZNA HospitalsAntwerpBelgium
- Division of ResearchPeter Mac Callum Cancer CentreMelbourneAustralia
| | - Sarah E Pinder
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Swapnil Rane
- Department of PathologyTata Memorial Centre‐ACTREC, HBNIMumbaiIndia
| | - Amit Sethi
- Department of Electrical EngineeringIndian Institute of Technology BombayMumbaiIndia
| | - Anita Grigoriadis
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- Breast Cancer Now Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
| |
Collapse
|
19
|
Lee M. Recent Advancements in Deep Learning Using Whole Slide Imaging for Cancer Prognosis. Bioengineering (Basel) 2023; 10:897. [PMID: 37627783 PMCID: PMC10451210 DOI: 10.3390/bioengineering10080897] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023] Open
Abstract
This review furnishes an exhaustive analysis of the latest advancements in deep learning techniques applied to whole slide images (WSIs) in the context of cancer prognosis, focusing specifically on publications from 2019 through 2023. The swiftly maturing field of deep learning, in combination with the burgeoning availability of WSIs, manifests significant potential in revolutionizing the predictive modeling of cancer prognosis. In light of the swift evolution and profound complexity of the field, it is essential to systematically review contemporary methodologies and critically appraise their ramifications. This review elucidates the prevailing landscape of this intersection, cataloging major developments, evaluating their strengths and weaknesses, and providing discerning insights into prospective directions. In this paper, a comprehensive overview of the field aims to be presented, which can serve as a critical resource for researchers and clinicians, ultimately enhancing the quality of cancer care outcomes. This review's findings accentuate the need for ongoing scrutiny of recent studies in this rapidly progressing field to discern patterns, understand breakthroughs, and navigate future research trajectories.
Collapse
Affiliation(s)
- Minhyeok Lee
- School of Electrical and Electronics Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
20
|
Chen S, Xiang J, Wang X, Zhang J, Yang S, Yang W, Zheng J, Han X. Deep learning-based pathology signature could reveal lymph node status and act as a novel prognostic marker across multiple cancer types. Br J Cancer 2023; 129:46-53. [PMID: 37137998 PMCID: PMC10307798 DOI: 10.1038/s41416-023-02262-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Identifying lymph node metastasis (LNM) relies mainly on indirect radiology. Current studies omitted the quantified associations with traits beyond cancer types, failing to provide generalisation performance across various tumour types. METHODS 4400 whole slide images across 11 cancer types were collected for training, cross-verification, and external validation of the pan-cancer lymph node metastasis (PC-LNM) model. We proposed an attention-based weakly supervised neural network based on self-supervised cancer-invariant features for the prediction task. RESULTS PC-LNM achieved a test area under the curve (AUC) of 0.732 (95% confidence interval: 0.717-0.746, P < 0.0001) in fivefold cross-validation of multiple cancer types, which also demonstrated good generalisation in the external validation cohort with AUC of 0.699 (95% confidence interval: 0.658-0.737, P < 0.0001). The interpretability results derived from PC-LNM revealed that the regions with the highest attention scores identified by the model generally correspond to tumours with poorly differentiated morphologies. PC-LNM achieved superior performance over previously reported methods and could also act as an independent prognostic factor for patients across multiple tumour types. DISCUSSION We presented an automated pan-cancer model for predicting the LNM status from primary tumour histology, which could act as a novel prognostic marker across multiple cancer types.
Collapse
Affiliation(s)
- Siteng Chen
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200135, Shanghai, China
| | | | - Xiyue Wang
- College of Computer Science, Sichuan University, 610065, Chengdu, China
| | - Jun Zhang
- Tencent AI Lab, 518057, Shenzhen, China.
| | - Sen Yang
- Tencent AI Lab, 518057, Shenzhen, China
| | - Wei Yang
- Tencent AI Lab, 518057, Shenzhen, China
| | - Junhua Zheng
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200135, Shanghai, China.
| | - Xiao Han
- Tencent AI Lab, 518057, Shenzhen, China.
| |
Collapse
|
21
|
Caldonazzi N, Rizzo PC, Eccher A, Girolami I, Fanelli GN, Naccarato AG, Bonizzi G, Fusco N, d’Amati G, Scarpa A, Pantanowitz L, Marletta S. Value of Artificial Intelligence in Evaluating Lymph Node Metastases. Cancers (Basel) 2023; 15:2491. [PMID: 37173958 PMCID: PMC10177013 DOI: 10.3390/cancers15092491] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
One of the most relevant prognostic factors in cancer staging is the presence of lymph node (LN) metastasis. Evaluating lymph nodes for the presence of metastatic cancerous cells can be a lengthy, monotonous, and error-prone process. Owing to digital pathology, artificial intelligence (AI) applied to whole slide images (WSIs) of lymph nodes can be exploited for the automatic detection of metastatic tissue. The aim of this study was to review the literature regarding the implementation of AI as a tool for the detection of metastases in LNs in WSIs. A systematic literature search was conducted in PubMed and Embase databases. Studies involving the application of AI techniques to automatically analyze LN status were included. Of 4584 retrieved articles, 23 were included. Relevant articles were labeled into three categories based upon the accuracy of AI in evaluating LNs. Published data overall indicate that the application of AI in detecting LN metastases is promising and can be proficiently employed in daily pathology practice.
Collapse
Affiliation(s)
- Nicolò Caldonazzi
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (N.C.); (P.C.R.); (A.S.); (S.M.)
| | - Paola Chiara Rizzo
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (N.C.); (P.C.R.); (A.S.); (S.M.)
| | - Albino Eccher
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, 37126 Verona, Italy
| | - Ilaria Girolami
- Department of Pathology, Lehrkrankenhaus der Paracelsus Medizinischen Privatuniversität, Provincial Hospital of Bolzano (SABES-ASDAA), 39100 Bolzano-Bozen, Italy;
| | - Giuseppe Nicolò Fanelli
- Division of Pathology, Department of Translational Research, New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.N.F.); (A.G.N.)
| | - Antonio Giuseppe Naccarato
- Division of Pathology, Department of Translational Research, New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.N.F.); (A.G.N.)
| | - Giuseppina Bonizzi
- Division of Pathology, IEO, Europefan Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy; (G.B.); (N.F.)
| | - Nicola Fusco
- Division of Pathology, IEO, Europefan Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy; (G.B.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Giulia d’Amati
- Department of Radiology, Oncology and Pathology, Sapienza, University of Rome, 00185 Rome, Italy;
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (N.C.); (P.C.R.); (A.S.); (S.M.)
| | - Liron Pantanowitz
- Department of Pathology, University of Michigan, Ann Arbor, MI 48104, USA;
| | - Stefano Marletta
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (N.C.); (P.C.R.); (A.S.); (S.M.)
- Department of Pathology, Pederzoli Hospital, 37019 Peschiera del Garda, Italy
| |
Collapse
|
22
|
Modak S, Abdel-Raheem E, Rueda L. Applications of Deep Learning in Disease Diagnosis of Chest Radiographs: A Survey on Materials and Methods. BIOMEDICAL ENGINEERING ADVANCES 2023. [DOI: 10.1016/j.bea.2023.100076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
23
|
Zhu L, Shi H, Wei H, Wang C, Shi S, Zhang F, Yan R, Liu Y, He T, Wang L, Cheng J, Duan H, Du H, Meng F, Zhao W, Gu X, Guo L, Ni Y, He Y, Guan T, Han A. An accurate prediction of the origin for bone metastatic cancer using deep learning on digital pathological images. EBioMedicine 2022; 87:104426. [PMID: 36577348 PMCID: PMC9803701 DOI: 10.1016/j.ebiom.2022.104426] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Determining the origin of bone metastatic cancer (OBMC) is of great significance to clinical therapeutics. It is challenging for pathologists to determine the OBMC with limited clinical information and bone biopsy. METHODS We designed a regional multiple-instance learning algorithm to predict the OBMC based on hematoxylin-eosin (H&E) staining slides alone. We collected 1041 cases from eight different hospitals and labeled 26,431 regions of interest to train the model. The performance of the model was assessed by ten-fold cross validation and external validation. Under the guidance of top3 predictions, we conducted an IHC test on 175 cases of unknown origins to compare the consistency of the results predicted by the model and indicated by the IHC markers. We also applied the model to identify whether there was tumor or not in a region, as well as distinguishing squamous cell carcinoma, adenocarcinoma, and neuroendocrine tumor. FINDINGS In the within-cohort, our model achieved a top1-accuracy of 91.35% and a top3-accuracy of 97.75%. In the external cohort, our model displayed a good generalizability with a top3-accuracy of 97.44%. The top1 consistency between the results of the model and the immunohistochemistry markers was 83.90% and the top3 consistency was 94.33%. The model obtained an accuracy of 98.98% to identify whether there was tumor or not and an accuracy of 93.85% to differentiate three types of cancers. INTERPRETATION Our model demonstrated good performance to predict the OBMC from routine histology and had great potential for assisting pathologists with determining the OBMC accurately. FUNDING National Science Foundation of China (61875102 and 61975089), Natural Science Foundation of Guangdong province (2021A15-15012379 and 2022A1515 012550), Science and Technology Research Program of Shenzhen City (JCYJ20200109110606054 and WDZC20200821141349001), and Tsinghua University Spring Breeze Fund (2020Z99CFZ023).
Collapse
Affiliation(s)
- Lianghui Zhu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Huijuan Shi
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huiting Wei
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chengjiang Wang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Shanshan Shi
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Fenfen Zhang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Renao Yan
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Yiqing Liu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Tingting He
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Liyuan Wang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junru Cheng
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Hufei Duan
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Hong Du
- Department of Pathology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Fengjiao Meng
- Department of Pathology, Zhongshan People's Hospital, Zhongshan, China
| | - Wenli Zhao
- Department of Pathology, The First People's Hospital of Huizhou, Huizhou, China
| | - Xia Gu
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Linlang Guo
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yingpeng Ni
- Department of Pathology, Jieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat-Sen University), Jieyang, China
| | - Yonghong He
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China,Corresponding author.
| | - Tian Guan
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China,Corresponding author.
| | - Anjia Han
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China,Corresponding author.
| |
Collapse
|
24
|
Sakamoto T, Furukawa T, Pham HHN, Kuroda K, Tabata K, Kashima Y, Okoshi EN, Morimoto S, Bychkov A, Fukuoka J. A collaborative workflow between pathologists and deep learning for the evaluation of tumour cellularity in lung adenocarcinoma. Histopathology 2022; 81:758-769. [PMID: 35989443 PMCID: PMC9826135 DOI: 10.1111/his.14779] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 01/11/2023]
Abstract
AIMS The reporting of tumour cellularity in cancer samples has become a mandatory task for pathologists. However, the estimation of tumour cellularity is often inaccurate. Therefore, we propose a collaborative workflow between pathologists and artificial intelligence (AI) models to evaluate tumour cellularity in lung cancer samples and propose a protocol to apply it to routine practice. METHODS AND RESULTS We developed a quantitative model of lung adenocarcinoma that was validated and tested on 50 cases, and a collaborative workflow where pathologists could access the AI results and adjust their original tumour cellularity scores (adjusted-score) that we tested on 151 cases. The adjusted-score was validated by comparing them with a ground truth established by manual annotation of haematoxylin and eosin slides with reference to immunostains with thyroid transcription factor-1 and napsin A. For training, validation, testing the AI and testing the collaborative workflow, we used 40, 10, 50 and 151 whole slide images of lung adenocarcinoma, respectively. The sensitivity and specificity of tumour segmentation were 97 and 87%, respectively, and the accuracy of nuclei recognition was 99%. One pathologist's visually estimated scores were compared to the adjusted-score, and the pathologist's scores were altered in 87% of cases. Comparison with the ground truth revealed that the adjusted-score was more precise than the pathologists' scores (P < 0.05). CONCLUSION We proposed a collaborative workflow between AI and pathologists as a model to improve daily practice and enhance the prediction of tumour cellularity for genetic tests.
Collapse
Affiliation(s)
- Taro Sakamoto
- Department of PathologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Tomoi Furukawa
- Department of PathologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Hoa H N Pham
- Department of PathologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Kishio Kuroda
- Department of PathologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan,Department of Pathology, Kameda Medical CenterKamogawaJapan
| | - Kazuhiro Tabata
- Department of PathologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Yukio Kashima
- Department of Pathology, Awaji Medical CenterSumotoJapan
| | - Ethan N Okoshi
- Department of PathologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Shimpei Morimoto
- Innovation Platform and Office for Precision Medicine (iPOP), Graduate School of Biomedical SciencesNagasaki UniversityNagasakiJapan
| | - Andrey Bychkov
- Department of PathologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan,Department of Pathology, Kameda Medical CenterKamogawaJapan
| | - Junya Fukuoka
- Department of PathologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan,Department of Pathology, Kameda Medical CenterKamogawaJapan
| |
Collapse
|
25
|
Wang L. Deep Learning Techniques to Diagnose Lung Cancer. Cancers (Basel) 2022; 14:5569. [PMID: 36428662 PMCID: PMC9688236 DOI: 10.3390/cancers14225569] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022] Open
Abstract
Medical imaging tools are essential in early-stage lung cancer diagnostics and the monitoring of lung cancer during treatment. Various medical imaging modalities, such as chest X-ray, magnetic resonance imaging, positron emission tomography, computed tomography, and molecular imaging techniques, have been extensively studied for lung cancer detection. These techniques have some limitations, including not classifying cancer images automatically, which is unsuitable for patients with other pathologies. It is urgently necessary to develop a sensitive and accurate approach to the early diagnosis of lung cancer. Deep learning is one of the fastest-growing topics in medical imaging, with rapidly emerging applications spanning medical image-based and textural data modalities. With the help of deep learning-based medical imaging tools, clinicians can detect and classify lung nodules more accurately and quickly. This paper presents the recent development of deep learning-based imaging techniques for early lung cancer detection.
Collapse
Affiliation(s)
- Lulu Wang
- Biomedical Device Innovation Center, Shenzhen Technology University, Shenzhen 518118, China
| |
Collapse
|
26
|
Reshma IA, Franchet C, Gaspard M, Ionescu RT, Mothe J, Cussat-Blanc S, Luga H, Brousset P. Finding a Suitable Class Distribution for Building Histological Images Datasets Used in Deep Model Training-The Case of Cancer Detection. J Digit Imaging 2022; 35:1326-1349. [PMID: 35445341 PMCID: PMC9582112 DOI: 10.1007/s10278-022-00618-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/15/2022] [Accepted: 03/09/2022] [Indexed: 11/26/2022] Open
Abstract
The class distribution of a training dataset is an important factor which influences the performance of a deep learning-based system. Understanding the optimal class distribution is therefore crucial when building a new training set which may be costly to annotate. This is the case for histological images used in cancer diagnosis where image annotation requires domain experts. In this paper, we tackle the problem of finding the optimal class distribution of a training set to be able to train an optimal model that detects cancer in histological images. We formulate several hypotheses which are then tested in scores of experiments with hundreds of trials. The experiments have been designed to account for both segmentation and classification frameworks with various class distributions in the training set, such as natural, balanced, over-represented cancer, and over-represented non-cancer. In the case of cancer detection, the experiments show several important results: (a) the natural class distribution produces more accurate results than the artificially generated balanced distribution; (b) the over-representation of non-cancer/negative classes (healthy tissue and/or background classes) compared to cancer/positive classes reduces the number of samples which are falsely predicted as cancer (false positive); (c) the least expensive to annotate non-ROI (non-region-of-interest) data can be useful in compensating for the performance loss in the system due to a shortage of expensive to annotate ROI data; (d) the multi-label examples are more useful than the single-label ones to train a segmentation model; and (e) when the classification model is tuned with a balanced validation set, it is less affected than the segmentation model by the class distribution of the training set.
Collapse
Affiliation(s)
| | - Camille Franchet
- Department of Pathology, University Cancer Institute of Toulouse-Oncopole, Toulouse, France
| | - Margot Gaspard
- Department of Pathology, University Cancer Institute of Toulouse-Oncopole, Toulouse, France
| | | | - Josiane Mothe
- IRIT, UMR5505 CNRS, Université de Toulouse, Toulouse, France
| | - Sylvain Cussat-Blanc
- IRIT, UMR5505 CNRS, Université de Toulouse, Toulouse, France
- Artificial and Natural Intelligence Toulouse Institute, Toulouse, France
| | - Hervé Luga
- IRIT, UMR5505 CNRS, Université de Toulouse, Toulouse, France
| | - Pierre Brousset
- Department of Pathology, University Cancer Institute of Toulouse-Oncopole, Toulouse, France
- INSERM UMR 1037 Cancer Research Centre of Toulouse (CRCT), Université Toulouse III Paul-Sabatier, CNRS ERL 5294, Toulouse, France
- Laboratoire d’Excellence TOUCAN, Toulouse, France
| |
Collapse
|
27
|
Xu Z, Peng J, Zeng X, Xu H, Chen Q. High-Accuracy Oral Squamous Cell Carcinoma Auxiliary Diagnosis System Based on EfficientNet. Front Oncol 2022; 12:894978. [PMID: 35875067 PMCID: PMC9302026 DOI: 10.3389/fonc.2022.894978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
It is important to diagnose the grade of oral squamous cell carcinoma (OSCC), but the current evaluation of the biopsy slide still mainly depends on the manual operation of pathologists. The workload of manual evaluation is large, and the results are greatly affected by the subjectivity of the pathologists. In recent years, with the development and application of deep learning, automatic evaluation of biopsy slides is gradually being applied to medical diagnoses, and it has shown good results. Therefore, a new OSCC auxiliary diagnostic system was proposed to automatically and accurately evaluate the patients’ tissue slides. This is the first study that compared the effects of different resolutions on the results. The OSCC tissue slides from The Cancer Genome Atlas (TCGA, n=697) and our independent datasets (n=337) were used for model training and verification. In the test dataset of tiles, the accuracy was 93.1% at 20x resolution (n=306,134), which was higher than that at 10x (n=154,148, accuracy=90.9%) and at 40x (n=890,681, accuracy=89.3%). The accuracy of the new system based on EfficientNet, which was used to evaluate the tumor grade of the biopsy slide, reached 98.1% [95% confidence interval (CI): 97.1% to 99.1%], and the area under the receiver operating characteristic curve (AUROC) reached 0.998 (95%CI: 0.995 to 1.000) in the TCGA dataset. When verifying the model on the independent image dataset, the accuracy still reached 91.4% (95% CI: 88.4% to 94.4%, at 20x) and the AUROC reached 0.992 (95%CI: 0.982 to 1.000). It may benefit oral pathologists by reducing certain repetitive and time-consuming tasks, improving the efficiency of diagnosis, and facilitating the further development of computational histopathology.
Collapse
Affiliation(s)
- Ziang Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiakuan Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Xin Zeng, ; Hao Xu,
| | - Hao Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Xin Zeng, ; Hao Xu,
| | - Qianming Chen
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, China
| |
Collapse
|
28
|
Viswanathan VS, Toro P, Corredor G, Mukhopadhyay S, Madabhushi A. The state of the art for artificial intelligence in lung digital pathology. J Pathol 2022; 257:413-429. [PMID: 35579955 PMCID: PMC9254900 DOI: 10.1002/path.5966] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/26/2022] [Accepted: 05/15/2022] [Indexed: 12/03/2022]
Abstract
Lung diseases carry a significant burden of morbidity and mortality worldwide. The advent of digital pathology (DP) and an increase in computational power have led to the development of artificial intelligence (AI)-based tools that can assist pathologists and pulmonologists in improving clinical workflow and patient management. While previous works have explored the advances in computational approaches for breast, prostate, and head and neck cancers, there has been a growing interest in applying these technologies to lung diseases as well. The application of AI tools on radiology images for better characterization of indeterminate lung nodules, fibrotic lung disease, and lung cancer risk stratification has been well documented. In this article, we discuss methodologies used to build AI tools in lung DP, describing the various hand-crafted and deep learning-based unsupervised feature approaches. Next, we review AI tools across a wide spectrum of lung diseases including cancer, tuberculosis, idiopathic pulmonary fibrosis, and COVID-19. We discuss the utility of novel imaging biomarkers for different types of clinical problems including quantification of biomarkers like PD-L1, lung disease diagnosis, risk stratification, and prediction of response to treatments such as immune checkpoint inhibitors. We also look briefly at some emerging applications of AI tools in lung DP such as multimodal data analysis, 3D pathology, and transplant rejection. Lastly, we discuss the future of DP-based AI tools, describing the challenges with regulatory approval, developing reimbursement models, planning clinical deployment, and addressing AI biases. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Paula Toro
- Department of PathologyCleveland ClinicClevelandOHUSA
| | - Germán Corredor
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| | | | - Anant Madabhushi
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| |
Collapse
|
29
|
Fine-Tuned DenseNet-169 for Breast Cancer Metastasis Prediction Using FastAI and 1-Cycle Policy. SENSORS 2022; 22:s22082988. [PMID: 35458972 PMCID: PMC9025766 DOI: 10.3390/s22082988] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 12/02/2022]
Abstract
Lymph node metastasis in breast cancer may be accurately predicted using a DenseNet-169 model. However, the current system for identifying metastases in a lymph node is manual and tedious. A pathologist well-versed with the process of detection and characterization of lymph nodes goes through hours investigating histological slides. Furthermore, because of the massive size of most whole-slide images (WSI), it is wise to divide a slide into batches of small image patches and apply methods independently on each patch. The present work introduces a novel method for the automated diagnosis and detection of metastases from whole slide images using the Fast AI framework and the 1-cycle policy. Additionally, it compares this new approach to previous methods. The proposed model has surpassed other state-of-art methods with more than 97.4% accuracy. In addition, a mobile application is developed for prompt and quick response. It collects user information and models to diagnose metastases present in the early stages of cancer. These results indicate that the suggested model may assist general practitioners in accurately analyzing breast cancer situations, hence preventing future complications and mortality. With digital image processing, histopathologic interpretation and diagnostic accuracy have improved considerably.
Collapse
|
30
|
Budak C, Mençik V. Detection of ring cell cancer in histopathological images with region of interest determined by SLIC superpixels method. Neural Comput Appl 2022. [DOI: 10.1007/s00521-022-07183-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
31
|
Zaizen Y, Kanahori Y, Ishijima S, Kitamura Y, Yoon HS, Ozasa M, Mukae H, Bychkov A, Hoshino T, Fukuoka J. Deep-Learning-Aided Detection of Mycobacteria in Pathology Specimens Increases the Sensitivity in Early Diagnosis of Pulmonary Tuberculosis Compared with Bacteriology Tests. Diagnostics (Basel) 2022; 12:diagnostics12030709. [PMID: 35328262 PMCID: PMC8946921 DOI: 10.3390/diagnostics12030709] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 01/27/2023] Open
Abstract
The histopathological diagnosis of mycobacterial infection may be improved by a comprehensive analysis using artificial intelligence. Two autopsy cases of pulmonary tuberculosis, and forty biopsy cases of undetected acid-fast bacilli (AFB) were used to train AI (convolutional neural network), and construct an AI to support AFB detection. Forty-two patients underwent bronchoscopy, and were evaluated using AI-supported pathology to detect AFB. The AI-supported pathology diagnosis was compared with bacteriology diagnosis from bronchial lavage fluid and the final definitive diagnosis of mycobacteriosis. Among the 16 patients with mycobacteriosis, bacteriology was positive in 9 patients (56%). Two patients (13%) were positive for AFB without AI assistance, whereas AI-supported pathology identified eleven positive patients (69%). When limited to tuberculosis, AI-supported pathology had significantly higher sensitivity compared with bacteriology (86% vs. 29%, p = 0.046). Seven patients diagnosed with mycobacteriosis had no consolidation or cavitary shadows in computed tomography; the sensitivity of bacteriology and AI-supported pathology was 29% and 86%, respectively (p = 0.046). The specificity of AI-supported pathology was 100% in this study. AI-supported pathology may be more sensitive than bacteriological tests for detecting AFB in samples collected via bronchoscopy.
Collapse
Affiliation(s)
- Yoshiaki Zaizen
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.Z.); (Y.K.); (S.I.); (Y.K.); (H.-S.Y.); (M.O.)
- Division of Respirology, Neurology and Rheumatology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka 830-0011, Japan;
| | - Yuki Kanahori
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.Z.); (Y.K.); (S.I.); (Y.K.); (H.-S.Y.); (M.O.)
| | - Sousuke Ishijima
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.Z.); (Y.K.); (S.I.); (Y.K.); (H.-S.Y.); (M.O.)
| | - Yuka Kitamura
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.Z.); (Y.K.); (S.I.); (Y.K.); (H.-S.Y.); (M.O.)
- N Lab Co. Ltd., 1-43-403 Dejima, Nagasaki 850-0862, Japan
| | - Han-Seung Yoon
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.Z.); (Y.K.); (S.I.); (Y.K.); (H.-S.Y.); (M.O.)
| | - Mutsumi Ozasa
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.Z.); (Y.K.); (S.I.); (Y.K.); (H.-S.Y.); (M.O.)
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan;
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan;
| | - Andrey Bychkov
- Department of Pathology, Kameda Medical Center, 929 Higashi-cho, Kamogawa, Chiba 296-8602, Japan;
| | - Tomoaki Hoshino
- Division of Respirology, Neurology and Rheumatology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka 830-0011, Japan;
| | - Junya Fukuoka
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.Z.); (Y.K.); (S.I.); (Y.K.); (H.-S.Y.); (M.O.)
- Department of Pathology, Kameda Medical Center, 929 Higashi-cho, Kamogawa, Chiba 296-8602, Japan;
- Correspondence: ; Tel.: +81-95-819-7055; Fax: +81-95-819-7056
| |
Collapse
|
32
|
Zhao Y, Hu B, Wang Y, Yin X, Jiang Y, Zhu X. Identification of gastric cancer with convolutional neural networks: a systematic review. MULTIMEDIA TOOLS AND APPLICATIONS 2022; 81:11717-11736. [PMID: 35221775 PMCID: PMC8856868 DOI: 10.1007/s11042-022-12258-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/20/2021] [Accepted: 01/14/2022] [Indexed: 06/14/2023]
Abstract
The identification of diseases is inseparable from artificial intelligence. As an important branch of artificial intelligence, convolutional neural networks play an important role in the identification of gastric cancer. We conducted a systematic review to summarize the current applications of convolutional neural networks in the gastric cancer identification. The original articles published in Embase, Cochrane Library, PubMed and Web of Science database were systematically retrieved according to relevant keywords. Data were extracted from published papers. A total of 27 articles were retrieved for the identification of gastric cancer using medical images. Among them, 19 articles were applied in endoscopic images and 8 articles were applied in pathological images. 16 studies explored the performance of gastric cancer detection, 7 studies explored the performance of gastric cancer classification, 2 studies reported the performance of gastric cancer segmentation and 2 studies analyzed the performance of gastric cancer delineating margins. The convolutional neural network structures involved in the research included AlexNet, ResNet, VGG, Inception, DenseNet and Deeplab, etc. The accuracy of studies was 77.3 - 98.7%. Good performances of the systems based on convolutional neural networks have been showed in the identification of gastric cancer. Artificial intelligence is expected to provide more accurate information and efficient judgments for doctors to diagnose diseases in clinical work.
Collapse
Affiliation(s)
- Yuxue Zhao
- School of Nursing, Department of Medicine, Qingdao University, No. 15, Ningde Road, Shinan District, Qingdao, 266073 China
| | - Bo Hu
- Department of Thoracic Surgery, Qingdao Municipal Hospital, Qingdao, China
| | - Ying Wang
- School of Nursing, Department of Medicine, Qingdao University, No. 15, Ningde Road, Shinan District, Qingdao, 266073 China
| | - Xiaomeng Yin
- Pediatrics Intensive Care Unit, Qingdao Municipal Hospital, Qingdao, China
| | - Yuanyuan Jiang
- International Medical Services, Qilu Hospital of Shandong University, Jinan, China
| | - Xiuli Zhu
- School of Nursing, Department of Medicine, Qingdao University, No. 15, Ningde Road, Shinan District, Qingdao, 266073 China
| |
Collapse
|
33
|
Construction of a Diagnostic Model for Lymph Node Metastasis of the Papillary Thyroid Carcinoma Using Preoperative Ultrasound Features and Imaging Omics. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:1872412. [PMID: 35178222 PMCID: PMC8846989 DOI: 10.1155/2022/1872412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/14/2021] [Accepted: 01/07/2022] [Indexed: 11/17/2022]
Abstract
In this paper, we mainly adopted 337 patients who had undergone the surgery on lymph node metastasis of papillary thyroid carcinoma (PTC) as the sample population. In order to provide clinical reference for the intelligent decision-making in treatment plan and improvement of prognosis, we utilized ultrasound features and imaging features to construct five early diagnosis models for patients based on the ultrasound features, imaging features, and combined features. The model integrated with broad learning system (BLS) showed the best performance, with the area under the curve (AUC) of 0.857 (95% confidence interval (CI): 0.811–0.902)) and the accuracy of 0.805 (95% CI: 0.759–0.850). For demographic and clinical features, the prediction effect was also good, with the AUC more than 0.700.
Collapse
|
34
|
Tang H, Li G, Liu C, Huang D, Zhang X, Qiu Y, Liu Y. Diagnosis of lymph node metastasis in head and neck squamous cell carcinoma using deep learning. Laryngoscope Investig Otolaryngol 2022; 7:161-169. [PMID: 35155794 PMCID: PMC8823170 DOI: 10.1002/lio2.742] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/04/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND To build an automatic pathological diagnosis model to assess the lymph node metastasis status of head and neck squamous cell carcinoma (HNSCC) based on deep learning algorithms. STUDY DESIGN A retrospective study. METHODS A diagnostic model integrating two-step deep learning networks was trained to analyze the metastasis status in 85 images of HNSCC lymph nodes. The diagnostic model was tested in a test set of 21 images with metastasis and 29 images without metastasis. All images were scanned from HNSCC lymph node sections stained with hematoxylin-eosin (HE). RESULTS In the test set, the overall accuracy, sensitivity, and specificity of the diagnostic model reached 86%, 100%, and 75.9%, respectively. CONCLUSIONS Our two-step diagnostic model can be used to automatically assess the status of HNSCC lymph node metastasis with high sensitivity. LEVEL OF EVIDENCE NA.
Collapse
Affiliation(s)
- Haosheng Tang
- Department of Otolaryngology‐Head and Neck Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Otolaryngology Major Disease Research Key Laboratory of Hunan ProvinceChangshaHunanChina
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan ProvinceChangshaHunanChina
| | - Guo Li
- Department of Otolaryngology‐Head and Neck Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Otolaryngology Major Disease Research Key Laboratory of Hunan ProvinceChangshaHunanChina
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan ProvinceChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaHunanChina
| | - Chao Liu
- Department of Otolaryngology‐Head and Neck Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Otolaryngology Major Disease Research Key Laboratory of Hunan ProvinceChangshaHunanChina
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan ProvinceChangshaHunanChina
| | - Donghai Huang
- Department of Otolaryngology‐Head and Neck Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Otolaryngology Major Disease Research Key Laboratory of Hunan ProvinceChangshaHunanChina
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan ProvinceChangshaHunanChina
| | - Xin Zhang
- Department of Otolaryngology‐Head and Neck Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Otolaryngology Major Disease Research Key Laboratory of Hunan ProvinceChangshaHunanChina
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan ProvinceChangshaHunanChina
| | - Yuanzheng Qiu
- Department of Otolaryngology‐Head and Neck Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Otolaryngology Major Disease Research Key Laboratory of Hunan ProvinceChangshaHunanChina
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan ProvinceChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaHunanChina
| | - Yong Liu
- Department of Otolaryngology‐Head and Neck Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Otolaryngology Major Disease Research Key Laboratory of Hunan ProvinceChangshaHunanChina
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan ProvinceChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaHunanChina
| |
Collapse
|
35
|
AIM in Respiratory Disorders. Artif Intell Med 2022. [DOI: 10.1007/978-3-030-64573-1_178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
36
|
Carrillo-Perez F, Morales JC, Castillo-Secilla D, Molina-Castro Y, Guillén A, Rojas I, Herrera LJ. Non-small-cell lung cancer classification via RNA-Seq and histology imaging probability fusion. BMC Bioinformatics 2021; 22:454. [PMID: 34551733 PMCID: PMC8456075 DOI: 10.1186/s12859-021-04376-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 09/11/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Adenocarcinoma and squamous cell carcinoma are the two most prevalent lung cancer types, and their distinction requires different screenings, such as the visual inspection of histology slides by an expert pathologist, the analysis of gene expression or computer tomography scans, among others. In recent years, there has been an increasing gathering of biological data for decision support systems in the diagnosis (e.g. histology imaging, next-generation sequencing technologies data, clinical information, etc.). Using all these sources to design integrative classification approaches may improve the final diagnosis of a patient, in the same way that doctors can use multiple types of screenings to reach a final decision on the diagnosis. In this work, we present a late fusion classification model using histology and RNA-Seq data for adenocarcinoma, squamous-cell carcinoma and healthy lung tissue. RESULTS The classification model improves results over using each source of information separately, being able to reduce the diagnosis error rate up to a 64% over the isolate histology classifier and a 24% over the isolate gene expression classifier, reaching a mean F1-Score of 95.19% and a mean AUC of 0.991. CONCLUSIONS These findings suggest that a classification model using a late fusion methodology can considerably help clinicians in the diagnosis between the aforementioned lung cancer cancer subtypes over using each source of information separately. This approach can also be applied to any cancer type or disease with heterogeneous sources of information.
Collapse
Affiliation(s)
- Francisco Carrillo-Perez
- Department of Computer Architecture and Technology, University of Granada. C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18014, Granada, Spain.
| | - Juan Carlos Morales
- Department of Computer Architecture and Technology, University of Granada. C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18014, Granada, Spain
| | - Daniel Castillo-Secilla
- Department of Computer Architecture and Technology, University of Granada. C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18014, Granada, Spain
| | - Yésica Molina-Castro
- Department of Computer Architecture and Technology, University of Granada. C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18014, Granada, Spain
| | - Alberto Guillén
- Department of Computer Architecture and Technology, University of Granada. C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18014, Granada, Spain
| | - Ignacio Rojas
- Department of Computer Architecture and Technology, University of Granada. C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18014, Granada, Spain
| | - Luis Javier Herrera
- Department of Computer Architecture and Technology, University of Granada. C.I.T.I.C., Periodista Rafael Gómez Montero, 2, 18014, Granada, Spain
| |
Collapse
|
37
|
Calderaro J, Kather JN. Artificial intelligence-based pathology for gastrointestinal and hepatobiliary cancers. Gut 2021; 70:1183-1193. [PMID: 33214163 DOI: 10.1136/gutjnl-2020-322880] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/03/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022]
Abstract
Artificial intelligence (AI) can extract complex information from visual data. Histopathology images of gastrointestinal (GI) and liver cancer contain a very high amount of information which human observers can only partially make sense of. Complementing human observers, AI allows an in-depth analysis of digitised histological slides of GI and liver cancer and offers a wide range of clinically relevant applications. First, AI can automatically detect tumour tissue, easing the exponentially increasing workload on pathologists. In addition, and possibly exceeding pathologist's capacities, AI can capture prognostically relevant tissue features and thus predict clinical outcome across GI and liver cancer types. Finally, AI has demonstrated its capacity to infer molecular and genetic alterations of cancer tissues from histological digital slides. These are likely only the first of many AI applications that will have important clinical implications. Thus, pathologists and clinicians alike should be aware of the principles of AI-based pathology and its ability to solve clinically relevant problems, along with its limitations and biases.
Collapse
Affiliation(s)
- Julien Calderaro
- U955, INSERM, Créteil, France .,Pathology, Hopital Henri Mondor, Creteil, Île-de-France, France
| | - Jakob Nikolas Kather
- Applied Tumor Immunity, Deutsches Krebsforschungszentrum, Heidelberg, BW, Germany.,Department of Medicine III, University Hospital RWTH, Aachen, Germany
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Pathology is the cornerstone of cancer care. Pathomics, which represents the use of artificial intelligence in digital pathology, is an emerging and promising field that will revolutionize medical and surgical pathology in the coming years. This review provides an overview of pathomics, its current and future applications and its most relevant applications in Head and Neck cancer care. RECENT FINDINGS The number of studies investigating the use of artificial intelligence in pathology is rapidly growing, especially as the utilization of deep learning has shown great potential with Whole Slide Images. Even though numerous steps still remain before its clinical use, Pathomics has been used for varied applications comprising of computer-assisted diagnosis, molecular anomalies prediction, tumor microenvironment and biomarker identification as well as prognosis evaluation. The majority of studies were performed on the most frequent cancers, notably breast, prostate, and lung. Interesting results were also found in Head and Neck cancers. SUMMARY Even if its use in Head and Neck cancer care is still low, Pathomics is a powerful tool to improve diagnosis, identify prognostic factors and new biomarkers. Important challenges lie ahead before its use in a clinical practice, notably the lack of information on how AI makes its decisions, the slow deployment of digital pathology, and the need for extensively validated data in order to obtain authorities approval. Regardless, pathomics will most likely improve pathology in general, including Head and Neck cancer care in the coming years.
Collapse
|
39
|
Ran J, Cao R, Cai J, Yu T, Zhao D, Wang Z. Development and Validation of a Nomogram for Preoperative Prediction of Lymph Node Metastasis in Lung Adenocarcinoma Based on Radiomics Signature and Deep Learning Signature. Front Oncol 2021; 11:585942. [PMID: 33968715 PMCID: PMC8101496 DOI: 10.3389/fonc.2021.585942] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The preoperative LN (lymph node) status of patients with LUAD (lung adenocarcinoma) is a key factor for determining if systemic nodal dissection is required, which is usually confirmed after surgery. This study aimed to develop and validate a nomogram for preoperative prediction of LN metastasis in LUAD based on a radiomics signature and deep learning signature. MATERIALS AND METHODS This retrospective study included a training cohort of 200 patients, an internal validation cohort of 40 patients, and an external validation cohort of 60 patients. Radiomics features were extracted from conventional CT (computed tomography) images. T-test and Extra-trees were performed for feature selection, and the selected features were combined using logistic regression to build the radiomics signature. The features and weights of the last fully connected layer of a CNN (convolutional neural network) were combined to obtain a deep learning signature. By incorporating clinical risk factors, the prediction model was developed using a multivariable logistic regression analysis, based on which the nomogram was developed. The calibration, discrimination and clinical values of the nomogram were evaluated. RESULTS Multivariate logistic regression analysis showed that the radiomics signature, deep learning signature, and CT-reported LN status were independent predictors. The prediction model developed by all the independent predictors showed good discrimination (C-index, 0.820; 95% CI, 0.762 to 0.879) and calibration (Hosmer-Lemeshow test, P=0.193) capabilities for the training cohort. Additionally, the model achieved satisfactory discrimination (C-index, 0.861; 95% CI, 0.769 to 0.954) and calibration (Hosmer-Lemeshow test, P=0.775) when applied to the external validation cohort. An analysis of the decision curve showed that the nomogram had potential for clinical application. CONCLUSIONS This study presents a prediction model based on radiomics signature, deep learning signature, and CT-reported LN status that can be used to predict preoperative LN metastasis in patients with LUAD.
Collapse
Affiliation(s)
- Jia Ran
- Engineering Research Center of Molecular & Neuro-imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, China
| | - Ran Cao
- Engineering Research Center of Molecular & Neuro-imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, China
| | - Jiumei Cai
- Department of Medical Imaging, Cancer Hospital of China Medical University, Shenyang, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Shenyang, China
- Department of Medical Imaging, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Dan Zhao
- Department of Medical Imaging, Cancer Hospital of China Medical University, Shenyang, China
- Department of Medical Imaging, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Zhongliang Wang
- Engineering Research Center of Molecular & Neuro-imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, China
| |
Collapse
|
40
|
Yang H, Chen L, Cheng Z, Yang M, Wang J, Lin C, Wang Y, Huang L, Chen Y, Peng S, Ke Z, Li W. Deep learning-based six-type classifier for lung cancer and mimics from histopathological whole slide images: a retrospective study. BMC Med 2021; 19:80. [PMID: 33775248 PMCID: PMC8006383 DOI: 10.1186/s12916-021-01953-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/26/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Targeted therapy and immunotherapy put forward higher demands for accurate lung cancer classification, as well as benign versus malignant disease discrimination. Digital whole slide images (WSIs) witnessed the transition from traditional histopathology to computational approaches, arousing a hype of deep learning methods for histopathological analysis. We aimed at exploring the potential of deep learning models in the identification of lung cancer subtypes and cancer mimics from WSIs. METHODS We initially obtained 741 WSIs from the First Affiliated Hospital of Sun Yat-sen University (SYSUFH) for the deep learning model development, optimization, and verification. Additional 318 WSIs from SYSUFH, 212 from Shenzhen People's Hospital, and 422 from The Cancer Genome Atlas were further collected for multi-centre verification. EfficientNet-B5- and ResNet-50-based deep learning methods were developed and compared using the metrics of recall, precision, F1-score, and areas under the curve (AUCs). A threshold-based tumour-first aggregation approach was proposed and implemented for the label inferencing of WSIs with complex tissue components. Four pathologists of different levels from SYSUFH reviewed all the testing slides blindly, and the diagnosing results were used for quantitative comparisons with the best performing deep learning model. RESULTS We developed the first deep learning-based six-type classifier for histopathological WSI classification of lung adenocarcinoma, lung squamous cell carcinoma, small cell lung carcinoma, pulmonary tuberculosis, organizing pneumonia, and normal lung. The EfficientNet-B5-based model outperformed ResNet-50 and was selected as the backbone in the classifier. Tested on 1067 slides from four cohorts of different medical centres, AUCs of 0.970, 0.918, 0.963, and 0.978 were achieved, respectively. The classifier achieved high consistence to the ground truth and attending pathologists with high intraclass correlation coefficients over 0.873. CONCLUSIONS Multi-cohort testing demonstrated our six-type classifier achieved consistent and comparable performance to experienced pathologists and gained advantages over other existing computational methods. The visualization of prediction heatmap improved the model interpretability intuitively. The classifier with the threshold-based tumour-first label inferencing method exhibited excellent accuracy and feasibility in classifying lung cancers and confused nonneoplastic tissues, indicating that deep learning can resolve complex multi-class tissue classification that conforms to real-world histopathological scenarios.
Collapse
Affiliation(s)
- Huan Yang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Lili Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhiqiang Cheng
- Department of Pathology, Shenzhen People's Hospital, Shenzhen, 518020, China
| | - Minglei Yang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jianbo Wang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chenghao Lin
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuefeng Wang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Leilei Huang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yangshan Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Sui Peng
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Molecular Diagnosis Center or Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China. .,Center for Precision Medicine, Sun Yat-sen University, Guangzhou, 510080, China. .,Molecular Diagnosis Center or Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Weizhong Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China. .,Center for Precision Medicine, Sun Yat-sen University, Guangzhou, 510080, China. .,Key Laboratory of Tropical Disease Control (Ministry of Education), Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
41
|
Das N, Topalovic M, Janssens W. AIM in Respiratory Disorders. Artif Intell Med 2021. [DOI: 10.1007/978-3-030-58080-3_178-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
42
|
Xing F, Zhang X, Cornish TC. Artificial intelligence for pathology. Artif Intell Med 2021. [DOI: 10.1016/b978-0-12-821259-2.00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
43
|
Schömig-Markiefka B, Pryalukhin A, Hulla W, Bychkov A, Fukuoka J, Madabhushi A, Achter V, Nieroda L, Büttner R, Quaas A, Tolkach Y. Quality control stress test for deep learning-based diagnostic model in digital pathology. Mod Pathol 2021; 34:2098-2108. [PMID: 34168282 PMCID: PMC8592835 DOI: 10.1038/s41379-021-00859-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/06/2021] [Accepted: 06/06/2021] [Indexed: 11/23/2022]
Abstract
Digital pathology provides a possibility for computational analysis of histological slides and automatization of routine pathological tasks. Histological slides are very heterogeneous concerning staining, sections' thickness, and artifacts arising during tissue processing, cutting, staining, and digitization. In this study, we digitally reproduce major types of artifacts. Using six datasets from four different institutions digitized by different scanner systems, we systematically explore artifacts' influence on the accuracy of the pre-trained, validated, deep learning-based model for prostate cancer detection in histological slides. We provide evidence that any histological artifact dependent on severity can lead to a substantial loss in model performance. Strategies for the prevention of diagnostic model accuracy losses in the context of artifacts are warranted. Stress-testing of diagnostic models using synthetically generated artifacts might be an essential step during clinical validation of deep learning-based algorithms.
Collapse
Affiliation(s)
- Birgid Schömig-Markiefka
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Alexey Pryalukhin
- Institute of Pathology, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
| | - Wolfgang Hulla
- Institute of Pathology, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
| | - Andrey Bychkov
- grid.174567.60000 0000 8902 2273Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan ,grid.414927.d0000 0004 0378 2140Department of Pathology, Kameda Medical Center, Kamogawa, Japan
| | - Junya Fukuoka
- grid.174567.60000 0000 8902 2273Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan ,grid.414927.d0000 0004 0378 2140Department of Pathology, Kameda Medical Center, Kamogawa, Japan
| | - Anant Madabhushi
- grid.67105.350000 0001 2164 3847Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH USA ,grid.410349.b0000 0004 5912 6484Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH USA
| | - Viktor Achter
- grid.6190.e0000 0000 8580 3777Regional computing center (RRZK), University of Cologne, Cologne, Germany
| | - Lech Nieroda
- grid.6190.e0000 0000 8580 3777Regional computing center (RRZK), University of Cologne, Cologne, Germany
| | - Reinhard Büttner
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Alexander Quaas
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Yuri Tolkach
- Institute of Pathology, University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
44
|
Li F, Shi JX, Yan L, Wang YG, Zhang XD, Jiang MS, Wu ZZ, Zhou KQ. Lesion-aware convolutional neural network for chest radiograph classification. Clin Radiol 2020; 76:155.e1-155.e14. [PMID: 33077154 DOI: 10.1016/j.crad.2020.08.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 08/18/2020] [Indexed: 01/18/2023]
Abstract
AIM To investigate the performance of a deep-learning approach termed lesion-aware convolutional neural network (LACNN) to identify 14 different thoracic diseases on chest X-rays (CXRs). MATERIALS AND METHODS In total, 10,738 CXRs of 3,526 patients were collected retrospectively. Of these, 1,937 CXRs of 598 patients were selected for training and optimising the lesion-detection network (LDN) of LACNN. The remaining 8,801 CXRs from 2,928 patients were used to train and test the classification network of LACNN. The discriminative performance of the deep-learning approach was compared with that obtained by the radiologists. In addition, its generalisation was validated on the independent public dataset, ChestX-ray14. The decision-making process of the model was visualised by occlusion testing, and the effect of the integration of CXRs and non-image data on model performance was also investigated. In a systematic evaluation, F1 score, sensitivity, specificity, and area under the receiver operating characteristic curve (AUC) metrics were calculated. RESULTS The model generated statistically significantly higher AUC performance compared with radiologists on atelectasis, mass, and nodule, with AUC values of 0.831 (95% confidence interval [CI]: 0.807-0.855), 0.959 (95% CI: 0.944-0.974), and 0.928 (95% CI: 0.906-0.950), respectively. For the other 11 pathologies, there were no statistically significant differences. The average time to complete each CXR classification in the testing dataset was substantially longer for the radiologists (∼35 seconds) than for the LACNN (∼0.197 seconds). In the ChestX-ray14 dataset, the present model also showed competitive performance in comparison with other state-of-the-art deep-learning approaches. Model performance was slightly improved when introducing non-image data. CONCLUSION The proposed LACNN achieved radiologist-level performance in identifying thoracic diseases on CXRs, and could potentially expand patient access to CXR diagnostics.
Collapse
Affiliation(s)
- F Li
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - J-X Shi
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - L Yan
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Y-G Wang
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - X-D Zhang
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - M-S Jiang
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, China.
| | - Z-Z Wu
- Department of Precision Mechanical Engineering, Shanghai University, Shanghai, China
| | - K-Q Zhou
- Liver Cancer Institute, Zhongshan Hospital, Shanghai, China
| |
Collapse
|
45
|
Sakamoto T, Furukawa T, Lami K, Pham HHN, Uegami W, Kuroda K, Kawai M, Sakanashi H, Cooper LAD, Bychkov A, Fukuoka J. A narrative review of digital pathology and artificial intelligence: focusing on lung cancer. Transl Lung Cancer Res 2020; 9:2255-2276. [PMID: 33209648 PMCID: PMC7653145 DOI: 10.21037/tlcr-20-591] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The emergence of whole slide imaging technology allows for pathology diagnosis on a computer screen. The applications of digital pathology are expanding, from supporting remote institutes suffering from a shortage of pathologists to routine use in daily diagnosis including that of lung cancer. Through practice and research large archival databases of digital pathology images have been developed that will facilitate the development of artificial intelligence (AI) methods for image analysis. Currently, several AI applications have been reported in the field of lung cancer; these include the segmentation of carcinoma foci, detection of lymph node metastasis, counting of tumor cells, and prediction of gene mutations. Although the integration of AI algorithms into clinical practice remains a significant challenge, we have implemented tumor cell count for genetic analysis, a helpful application for routine use. Our experience suggests that pathologists often overestimate the contents of tumor cells, and the use of AI-based analysis increases the accuracy and makes the tasks less tedious. However, there are several difficulties encountered in the practical use of AI in clinical diagnosis. These include the lack of sufficient annotated data for the development and validation of AI systems, the explainability of black box AI models, such as those based on deep learning that offer the most promising performance, and the difficulty in defining the ground truth data for training and validation owing to inherent ambiguity in most applications. All of these together present significant challenges in the development and clinical translation of AI methods in the practice of pathology. Additional research on these problems will help in resolving the barriers to the clinical use of AI. Helping pathologists in developing knowledge of the working and limitations of AI will benefit the use of AI in both diagnostics and research.
Collapse
Affiliation(s)
- Taro Sakamoto
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomoi Furukawa
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kris Lami
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hoa Hoang Ngoc Pham
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Wataru Uegami
- Department of Pathology, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Kishio Kuroda
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masataka Kawai
- Department of Pathology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Hidenori Sakanashi
- Configurable Learning Mechanism Research Team, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | | | - Andrey Bychkov
- Department of Pathology, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Junya Fukuoka
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Pathology, Kameda Medical Center, Kamogawa, Chiba, Japan
| |
Collapse
|
46
|
Gonem S, Janssens W, Das N, Topalovic M. Applications of artificial intelligence and machine learning in respiratory medicine. Thorax 2020; 75:695-701. [PMID: 32409611 DOI: 10.1136/thoraxjnl-2020-214556] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023]
Abstract
The past 5 years have seen an explosion of interest in the use of artificial intelligence (AI) and machine learning techniques in medicine. This has been driven by the development of deep neural networks (DNNs)-complex networks residing in silico but loosely modelled on the human brain-that can process complex input data such as a chest radiograph image and output a classification such as 'normal' or 'abnormal'. DNNs are 'trained' using large banks of images or other input data that have been assigned the correct labels. DNNs have shown the potential to equal or even surpass the accuracy of human experts in pattern recognition tasks such as interpreting medical images or biosignals. Within respiratory medicine, the main applications of AI and machine learning thus far have been the interpretation of thoracic imaging, lung pathology slides and physiological data such as pulmonary function tests. This article surveys progress in this area over the past 5 years, as well as highlighting the current limitations of AI and machine learning and the potential for future developments.
Collapse
Affiliation(s)
- Sherif Gonem
- Department of Respiratory Medicine, Nottingham University Hospitals NHS Trust, Nottingham, UK .,Division of Respiratory Medicine, University of Nottingham, Nottingham, UK
| | - Wim Janssens
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.,Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Nilakash Das
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Marko Topalovic
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.,ArtiQ NV, Leuven, Belgium
| |
Collapse
|
47
|
Rogers W, Thulasi Seetha S, Refaee TAG, Lieverse RIY, Granzier RWY, Ibrahim A, Keek SA, Sanduleanu S, Primakov SP, Beuque MPL, Marcus D, van der Wiel AMA, Zerka F, Oberije CJG, van Timmeren JE, Woodruff HC, Lambin P. Radiomics: from qualitative to quantitative imaging. Br J Radiol 2020; 93:20190948. [PMID: 32101448 DOI: 10.1259/bjr.20190948] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Historically, medical imaging has been a qualitative or semi-quantitative modality. It is difficult to quantify what can be seen in an image, and to turn it into valuable predictive outcomes. As a result of advances in both computational hardware and machine learning algorithms, computers are making great strides in obtaining quantitative information from imaging and correlating it with outcomes. Radiomics, in its two forms "handcrafted and deep," is an emerging field that translates medical images into quantitative data to yield biological information and enable radiologic phenotypic profiling for diagnosis, theragnosis, decision support, and monitoring. Handcrafted radiomics is a multistage process in which features based on shape, pixel intensities, and texture are extracted from radiographs. Within this review, we describe the steps: starting with quantitative imaging data, how it can be extracted, how to correlate it with clinical and biological outcomes, resulting in models that can be used to make predictions, such as survival, or for detection and classification used in diagnostics. The application of deep learning, the second arm of radiomics, and its place in the radiomics workflow is discussed, along with its advantages and disadvantages. To better illustrate the technologies being used, we provide real-world clinical applications of radiomics in oncology, showcasing research on the applications of radiomics, as well as covering its limitations and its future direction.
Collapse
Affiliation(s)
- William Rogers
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Thoracic Oncology, IRCCS Foundation National Cancer Institute, Milan, Italy
| | - Sithin Thulasi Seetha
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Thoracic Oncology, IRCCS Foundation National Cancer Institute, Milan, Italy
| | - Turkey A G Refaee
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Diagnostic Radiology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Relinde I Y Lieverse
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Renée W Y Granzier
- Department of Radiology and Nuclear Imaging, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Surgery, Maastricht University Medical Centre, Grow-School for Oncology and Developmental Biology, Maastricht, The Netherlands
| | - Abdalla Ibrahim
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Radiology and Nuclear Imaging, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Nuclear Medicine and Comprehensive diagnostic center Aachen (CDCA), University Hospital RWTH Aachen University, Aachen, Germany.,Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, Hospital Center Universitaire De Liege, Liege, Belgium
| | - Simon A Keek
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Sebastian Sanduleanu
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Sergey P Primakov
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Manon P L Beuque
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Damiënne Marcus
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Alexander M A van der Wiel
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Fadila Zerka
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Cary J G Oberije
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Janita E van Timmeren
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Radiation Oncology, University Hospital Zürich, Zürich, Switzerland.,University of Zürich, Zürich, Switzerland
| | - Henry C Woodruff
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Radiology and Nuclear Imaging, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Philippe Lambin
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Radiology and Nuclear Imaging, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|