6,031 research outputs found

    A Survey on Deep Learning in Medical Image Analysis

    Full text link
    Deep learning algorithms, in particular convolutional networks, have rapidly become a methodology of choice for analyzing medical images. This paper reviews the major deep learning concepts pertinent to medical image analysis and summarizes over 300 contributions to the field, most of which appeared in the last year. We survey the use of deep learning for image classification, object detection, segmentation, registration, and other tasks and provide concise overviews of studies per application area. Open challenges and directions for future research are discussed.Comment: Revised survey includes expanded discussion section and reworked introductory section on common deep architectures. Added missed papers from before Feb 1st 201

    The Liver Tumor Segmentation Benchmark (LiTS)

    Get PDF
    In this work, we report the set-up and results of the Liver Tumor Segmentation Benchmark (LiTS), which was organized in conjunction with the IEEE International Symposium on Biomedical Imaging (ISBI) 2017 and the International Conferences on Medical Image Computing and Computer-Assisted Intervention (MICCAI) 2017 and 2018. The image dataset is diverse and contains primary and secondary tumors with varied sizes and appearances with various lesion-to-background levels (hyper-/hypo-dense), created in collaboration with seven hospitals and research institutions. Seventy-five submitted liver and liver tumor segmentation algorithms were trained on a set of 131 computed tomography (CT) volumes and were tested on 70 unseen test images acquired from different patients. We found that not a single algorithm performed best for both liver and liver tumors in the three events. The best liver segmentation algorithm achieved a Dice score of 0.963, whereas, for tumor segmentation, the best algorithms achieved Dices scores of 0.674 (ISBI 2017), 0.702 (MICCAI 2017), and 0.739 (MICCAI 2018). Retrospectively, we performed additional analysis on liver tumor detection and revealed that not all top-performing segmentation algorithms worked well for tumor detection. The best liver tumor detection method achieved a lesion-wise recall of 0.458 (ISBI 2017), 0.515 (MICCAI 2017), and 0.554 (MICCAI 2018), indicating the need for further research. LiTS remains an active benchmark and resource for research, e.g., contributing the liver-related segmentation tasks in http://medicaldecathlon.com/. In addition, both data and online evaluation are accessible via https://competitions.codalab.org/competitions/17094.Bjoern Menze is supported through the DFG funding (SFB 824, subproject B12) and a Helmut-Horten-Professorship for Biomedical Informatics by the Helmut-Horten-Foundation. Florian Kofler is Supported by Deutsche Forschungsgemeinschaft (DFG) through TUM International Graduate School of Science and Engineering (IGSSE), GSC 81. An Tang was supported by the Fonds de recherche du Québec en Santé and Fondation de l’association des radiologistes du Québec (FRQS- ARQ 34939 Clinical Research Scholarship – Junior 2 Salary Award). Hongwei Bran Li is supported by Forschungskredit (Grant NO. FK-21- 125) from University of Zurich.Peer ReviewedArticle signat per 109 autors/es: Patrick Bilic 1,a,b, Patrick Christ 1,a,b, Hongwei Bran Li 1,2,∗,b, Eugene Vorontsov 3,a,b, Avi Ben-Cohen 5,a, Georgios Kaissis 10,12,15,a, Adi Szeskin 18,a, Colin Jacobs 4,a, Gabriel Efrain Humpire Mamani 4,a, Gabriel Chartrand 26,a, Fabian Lohöfer 12,a, Julian Walter Holch 29,30,69,a, Wieland Sommer 32,a, Felix Hofmann 31,32,a, Alexandre Hostettler 36,a, Naama Lev-Cohain 38,a, Michal Drozdzal 34,a, Michal Marianne Amitai 35,a, Refael Vivanti 37,a, Jacob Sosna 38,a, Ivan Ezhov 1, Anjany Sekuboyina 1,2, Fernando Navarro 1,76,78, Florian Kofler 1,13,57,78, Johannes C. Paetzold 15,16, Suprosanna Shit 1, Xiaobin Hu 1, Jana Lipková 17, Markus Rempfler 1, Marie Piraud 57,1, Jan Kirschke 13, Benedikt Wiestler 13, Zhiheng Zhang 14, Christian Hülsemeyer 1, Marcel Beetz 1, Florian Ettlinger 1, Michela Antonelli 9, Woong Bae 73, Míriam Bellver 43, Lei Bi 61, Hao Chen 39, Grzegorz Chlebus 62,64, Erik B. Dam 72, Qi Dou 41, Chi-Wing Fu 41, Bogdan Georgescu 60, Xavier Giró-i-Nieto 45, Felix Gruen 28, Xu Han 77, Pheng-Ann Heng 41, Jürgen Hesser 48,49,50, Jan Hendrik Moltz 62, Christian Igel 72, Fabian Isensee 69,70, Paul Jäger 69,70, Fucang Jia 75, Krishna Chaitanya Kaluva 21, Mahendra Khened 21, Ildoo Kim 73, Jae-Hun Kim 53, Sungwoong Kim 73, Simon Kohl 69, Tomasz Konopczynski 49, Avinash Kori 21, Ganapathy Krishnamurthi 21, Fan Li 22, Hongchao Li 11, Junbo Li 8, Xiaomeng Li 40, John Lowengrub 66,67,68, Jun Ma 54, Klaus Maier-Hein 69,70,7, Kevis-Kokitsi Maninis 44, Hans Meine 62,65, Dorit Merhof 74, Akshay Pai 72, Mathias Perslev 72, Jens Petersen 69, Jordi Pont-Tuset 44, Jin Qi 56, Xiaojuan Qi 40, Oliver Rippel 74, Karsten Roth 47, Ignacio Sarasua 51,12, Andrea Schenk 62,63, Zengming Shen 59,60, Jordi Torres 46,43, Christian Wachinger 51,12,1, Chunliang Wang 42, Leon Weninger 74, Jianrong Wu 25, Daguang Xu 71, Xiaoping Yang 55, Simon Chun-Ho Yu 58, Yading Yuan 52, Miao Yue 20, Liping Zhang 58, Jorge Cardoso 9, Spyridon Bakas 19,23,24, Rickmer Braren 6,12,30,a, Volker Heinemann 33,a, Christopher Pal 3,a, An Tang 27,a, Samuel Kadoury 3,a, Luc Soler 36,a, Bram van Ginneken 4,a, Hayit Greenspan 5,a, Leo Joskowicz 18,a, Bjoern Menze 1,2,a // 1 Department of Informatics, Technical University of Munich, Germany; 2 Department of Quantitative Biomedicine, University of Zurich, Switzerland; 3 Ecole Polytechnique de Montréal, Canada; 4 Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands; 5 Department of Biomedical Engineering, Tel-Aviv University, Israel; 6 German Cancer Consortium (DKTK), Germany; 7 Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; 8 Philips Research China, Philips China Innovation Campus, Shanghai, China; 9 School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK; 10 Institute for AI in Medicine, Technical University of Munich, Germany; 11 Department of Computer Science, Guangdong University of Foreign Studies, China; 12 Institute for diagnostic and interventional radiology, Klinikum rechts der Isar, Technical University of Munich, Germany; 13 Institute for diagnostic and interventional neuroradiology, Klinikum rechts der Isar,Technical University of Munich, Germany; 14 Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, China; 15 Department of Computing, Imperial College London, London, United Kingdom; 16 Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Zentrum München, Neuherberg, Germany; 17 Brigham and Women’s Hospital, Harvard Medical School, USA; 18 School of Computer Science and Engineering, the Hebrew University of Jerusalem, Israel; 19 Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, PA, USA; 20 CGG Services (Singapore) Pte. Ltd., Singapore; 21 Medical Imaging and Reconstruction Lab, Department of Engineering Design, Indian Institute of Technology Madras, India; 22 Sensetime, Shanghai, China; 23 Department of Radiology, Perelman School of Medicine, University of Pennsylvania, USA; 24 Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, PA, USA; 25 Tencent Healthcare (Shenzhen) Co., Ltd, China; 26 The University of Montréal Hospital Research Centre (CRCHUM) Montréal, Québec, Canada; 27 Department of Radiology, Radiation Oncology and Nuclear Medicine, University of Montréal, Canada; 28 Institute of Control Engineering, Technische Universität Braunschweig, Germany; 29 Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; 30 Comprehensive Cancer Center Munich, Munich, Germany; 31 Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Germany; 32 Department of Radiology, University Hospital, LMU Munich, Germany; 33 Department of Hematology/Oncology & Comprehensive Cancer Center Munich, LMU Klinikum Munich, Germany; 34 Polytechnique Montréal, Mila, QC, Canada; 35 Department of Diagnostic Radiology, Sheba Medical Center, Tel Aviv university, Israel; 36 Department of Surgical Data Science, Institut de Recherche contre les Cancers de l’Appareil Digestif (IRCAD), France; 37 Rafael Advanced Defense System, Israel; 38 Department of Radiology, Hadassah University Medical Center, Jerusalem, Israel; 39 Department of Computer Science and Engineering, The Hong Kong University of Science and Technology, China; 40 Department of Electrical and Electronic Engineering, The University of Hong Kong, China; 41 Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China; 42 Department of Biomedical Engineering and Health Systems, KTH Royal Institute of Technology, Sweden; 43 Barcelona Supercomputing Center, Barcelona, Spain; 44 Eidgenössische Technische Hochschule Zurich (ETHZ), Zurich, Switzerland; 45 Signal Theory and Communications Department, Universitat Politecnica de Catalunya, Catalonia, Spain; 46 Universitat Politecnica de Catalunya, Catalonia, Spain; 47 University of Tuebingen, Germany; 48 Mannheim Institute for Intelligent Systems in Medicine, department of Medicine Mannheim, Heidelberg University, Germany; 49 Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Germany; 50 Central Institute for Computer Engineering (ZITI), Heidelberg University, Germany; 51 Department of Child and Adolescent Psychiatry, Ludwig-Maximilians-Universität, Munich, Germany; 52 Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, NY, USA; 53 Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, South Korea; 54 Department of Mathematics, Nanjing University of Science and Technology, China; 55 Department of Mathematics, Nanjing University, China; 56 School of Information and Communication Engineering, University of Electronic Science and Technology of China, China; 57 Helmholtz AI, Helmholtz Zentrum München, Neuherberg, Germany; 58 Department of Imaging and Interventional Radiology, Chinese University of Hong Kong, Hong Kong, China; 59 Beckman Institute, University of Illinois at Urbana-Champaign, USA; 60 Siemens Healthineers, USA; 61 School of Computer Science, the University of Sydney, Australia; 62 Fraunhofer MEVIS, Bremen, Germany; 63 Institute for Diagnostic and Interventional Radiology, Hannover Medical School, Hannover, Germany; 64 Diagnostic Image Analysis Group, Radboud University Medical Center, Nijmegen, The Netherlands; 65 Medical Image Computing Group, FB3, University of Bremen, Germany; 66 Departments of Mathematics, Biomedical Engineering, University of California, Irvine, USA; 67 Center for Complex Biological Systems, University of California, Irvine, USA; 68 Chao Family Comprehensive Cancer Center, University of California, Irvine, USA; 69 Division of Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany; 70 Helmholtz Imaging, Germany; 71 NVIDIA, Santa Clara, CA, USA; 72 Department of Computer Science, University of Copenhagen, Denmark; 73 Kakao Brain, Republic of Korea; 74 Institute of Imaging & Computer Vision, RWTH Aachen University, Germany; 75 Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China; 76 Department of Radiation Oncology and Radiotherapy, Klinikum rechts der Isar, Technical University of Munich, Germany; 77 Department of computer science, UNC Chapel Hill, USA; 78 TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, GermanyPostprint (published version

    The Liver Tumor Segmentation Benchmark (LiTS)

    Full text link
    In this work, we report the set-up and results of the Liver Tumor Segmentation Benchmark (LiTS), which was organized in conjunction with the IEEE International Symposium on Biomedical Imaging (ISBI) 2017 and the International Conferences on Medical Image Computing and Computer-Assisted Intervention (MICCAI) 2017 and 2018. The image dataset is diverse and contains primary and secondary tumors with varied sizes and appearances with various lesion-to-background levels (hyper-/hypo-dense), created in collaboration with seven hospitals and research institutions. Seventy-five submitted liver and liver tumor segmentation algorithms were trained on a set of 131 computed tomography (CT) volumes and were tested on 70 unseen test images acquired from different patients. We found that not a single algorithm performed best for both liver and liver tumors in the three events. The best liver segmentation algorithm achieved a Dice score of 0.963, whereas, for tumor segmentation, the best algorithms achieved Dices scores of 0.674 (ISBI 2017), 0.702 (MICCAI 2017), and 0.739 (MICCAI 2018). Retrospectively, we performed additional analysis on liver tumor detection and revealed that not all top-performing segmentation algorithms worked well for tumor detection. The best liver tumor detection method achieved a lesion-wise recall of 0.458 (ISBI 2017), 0.515 (MICCAI 2017), and 0.554 (MICCAI 2018), indicating the need for further research. LiTS remains an active benchmark and resource for research, e.g., contributing the liver-related segmentation tasks in http://medicaldecathlon.com/. In addition, both data and online evaluation are accessible via https://competitions.codalab.org/competitions/17094

    The Liver Tumor Segmentation Benchmark (LiTS)

    Full text link
    In this work, we report the set-up and results of the Liver Tumor Segmentation Benchmark (LITS) organized in conjunction with the IEEE International Symposium on Biomedical Imaging (ISBI) 2016 and International Conference On Medical Image Computing Computer Assisted Intervention (MICCAI) 2017. Twenty four valid state-of-the-art liver and liver tumor segmentation algorithms were applied to a set of 131 computed tomography (CT) volumes with different types of tumor contrast levels (hyper-/hypo-intense), abnormalities in tissues (metastasectomie) size and varying amount of lesions. The submitted algorithms have been tested on 70 undisclosed volumes. The dataset is created in collaboration with seven hospitals and research institutions and manually reviewed by independent three radiologists. We found that not a single algorithm performed best for liver and tumors. The best liver segmentation algorithm achieved a Dice score of 0.96(MICCAI) whereas for tumor segmentation the best algorithm evaluated at 0.67(ISBI) and 0.70(MICCAI). The LITS image data and manual annotations continue to be publicly available through an online evaluation system as an ongoing benchmarking resource.Comment: conferenc

    Exploring the neuroblastoma DNA methylome: from biology to biomarker

    Get PDF
    Neuroblastoma (NB), a childhood tumor arising from immature sympathetic nervous system cells, is a heterogeneous disease with prognosis ranging from excellent long-term survival to high-risk with fatal outcome. In order to determine the most appropriate treatment modality, patients are stratified into risk groups at the time of diagnosis, based on combinations of clinical and biological parameters, namely age of the patient, tumor stage, histology, grade of differentiation, MYCN oncogene amplification, chromosome 11q aberration and DNA ploidy. However, use of this risk classification system has shown that accurate assessment of NB prognosis remains difficult and that additional prognostic markers are warranted. Therefore, we aimed to identify prognostic tumor DNA methylation biomarkers for NB. To find new biomarkers, we profiled the primary tumor DNA methylome using methyl-CpG-binding domain (MBD) sequencing, i.e. massively parallel sequencing of methylation-enriched DNA fractions, captured using the high affinity of MBD to bind methylated cytosines. As proof of principle, we applied this technology to 8 NB cell lines, and in combination with mRNA expression studies, this led to a first selection of 43 candidate biomarkers. Next, methylation-specific PCR (MSP) assays were designed, to allow candidate-specific methylation analysis in a primary tumor cohort of 89 samples. As such, we identified new prognostic DNA methylation biomarkers, and delineated the technological aspects and data analysis pipeline to set up a more extended biomarker study. In this follow-up study, the DNA methylome of 102 primary tumors, selected for risk classification and survival, was characterized by MBD sequencing. Differential methylation analyses between the prognostic patient groups put forward 78 top-ranking biomarker candidates, which were subsequently tested on two independent cohorts of 132 and 177 samples, adopting the high-throughput MSP pipeline of our pilot study. Multiple individual MSP assays were prognostically validated and through the implementation of a newly developed statistical framework, a robust 58-marker methylation signature predicting overall and event-free survival was established. This study represents the largest DNA methylation (biomarker) study in NB so far. The MBD sequencing data were shared with the research community through the format of a data descriptor. As such, these data are fully available to others, ensuring its reusability for other research purposes. To illustrate how these data can be applied to gain new insights into the NB pathology, we characterized the DNA methylome of stage 4S NB, a special type of NB found in infants with widespread metastases at diagnosis that paradoxically is associated with an excellent outcome due to its remarkable capacity to undergo spontaneous regression. More specifically, we compared promoter methylation levels between stage 4S, stage 1/2 (localized disease with favorable prognosis) and stage 4 (metastatic disease with dismal prognosis) tumors, and showed that specific chromosomal locations are enriched in stage 4S differentially methylated promoters and that specific subtelomeric promoters are hypermethylated in stage 4S. Furthermore, genes involved in important oncogenic pathways, in neural crest development and differentiation, and in epigenetic processes are differentially methylated and expressed in stage 4S. In conclusion, by exploring the DNA methylome of NB, we have not only demonstrated that DNA methylation patterns are intimately related to NB biology, but also found additional clinically relevant prognostic biomarkers

    Machine Learning Approaches to Predict Recurrence of Aggressive Tumors

    Get PDF
    Cancer recurrence is the major cause of cancer mortality. Despite tremendous research efforts, there is a dearth of biomarkers that reliably predict risk of cancer recurrence. Currently available biomarkers and tools in the clinic have limited usefulness to accurately identify patients with a higher risk of recurrence. Consequently, cancer patients suffer either from under- or over- treatment. Recent advances in machine learning and image analysis have facilitated development of techniques that translate digital images of tumors into rich source of new data. Leveraging these computational advances, my work addresses the unmet need to find risk-predictive biomarkers for Triple Negative Breast Cancer (TNBC), Ductal Carcinoma in-situ (DCIS), and Pancreatic Neuroendocrine Tumors (PanNETs). I have developed unique, clinically facile, models that determine the risk of recurrence, either local, invasive, or metastatic in these tumors. All models employ hematoxylin and eosin (H&E) stained digitized images of patient tumor samples as the primary source of data. The TNBC (n=322) models identified unique signatures from a panel of 133 protein biomarkers, relevant to breast cancer, to predict site of metastasis (brain, lung, liver, or bone) for TNBC patients. Even our least significant model (bone metastasis) offered superior prognostic value than clinopathological variables (Hazard Ratio [HR] of 5.123 vs. 1.397 p\u3c0.05). A second model predicted 10-year recurrence risk, in women with DCIS treated with breast conserving surgery, by identifying prognostically relevant features of tumor architecture from digitized H&E slides (n=344), using a novel two-step classification approach. In the validation cohort, our DCIS model provided a significantly higher HR (6.39) versus any clinopathological marker (p\u3c0.05). The third model is a deep-learning based, multi-label (annotation followed by metastasis association), whole slide image analysis pipeline (n=90) that identified a PanNET high risk group with over an 8x higher risk of metastasis (versus the low risk group p\u3c0.05), regardless of cofounding clinical variables. These machine-learning based models may guide treatment decisions and demonstrate proof-of-principle that computational pathology has tremendous clinical utility

    Identification of cancer hallmarks in patients with non-metastatic colon cancer after surgical resection

    Get PDF
    Colon cancer is one of the most common cancers in the world, and the therapeutic workflow is dependent on the TNM staging system and the presence of clinical risk factors. However, in the case of patients with non-metastatic disease, evaluating the benefit of adjuvant chemotherapy is a clinical challenge. Radiomics could be seen as a non-invasive novel imaging biomarker able to outline tumor phenotype and to predict patient prognosis by analyzing preoperative medical images. Radiomics might provide decisional support for oncologists with the goal to reduce the number of arbitrary decisions in the emerging era of personalized medicine. To date, much evidence highlights the strengths of radiomics in cancer workup, but several aspects limit the use of radiomics methods as routine. The study aimed to develop a radiomic model able to identify high-risk colon cancer by analyzing pre-operative CT scans. The study population comprised 148 patients: 108 with non-metastatic colon cancer were retrospectively enrolled from January 2015 to June 2020, and 40 patients were used as the external validation cohort. The population was divided into two groups—High-risk and No-risk—following the presence of at least one high-risk clinical factor. All patients had baseline CT scans, and 3D cancer segmentation was performed on the portal phase by two expert radiologists using open-source software (3DSlicer v4.10.2). Among the 107 radiomic features extracted, stable features were selected to evaluate the inter-class correlation (ICC) (cut-off ICC > 0.8). Stable features were compared between the two groups (T-test or Mann–Whitney), and the significant features were selected for univariate and multivariate logistic regression to build a predictive radiomic model. The radiomic model was then validated with an external cohort. In total, 58/108 were classified as High-risk and 50/108 as No-risk. A total of 35 radiomic features were stable (0.81 ≤ ICC <  0.92). Among these, 28 features were significantly different between the two groups (p < 0.05), and only 9 features were selected to build the radiomic model. The radiomic model yielded an AUC of 0.73 in the internal cohort and 0.75 in the external cohort. In conclusion, the radiomic model could be seen as a performant, non-invasive imaging tool to properly stratify colon cancers with high-risk diseas

    Pan-tumor CAnine cuTaneous Cancer Histology (CATCH) dataset

    Get PDF
    Due to morphological similarities, the differentiation of histologic sections of cutaneous tumors into individual subtypes can be challenging. Recently, deep learning-based approaches have proven their potential for supporting pathologists in this regard. However, many of these supervised algorithms require a large amount of annotated data for robust development. We present a publicly available dataset of 350 whole slide images of seven different canine cutaneous tumors complemented by 12,424 polygon annotations for 13 histologic classes, including seven cutaneous tumor subtypes. In inter-rater experiments, we show a high consistency of the provided labels, especially for tumor annotations. We further validate the dataset by training a deep neural network for the task of tissue segmentation and tumor subtype classification. We achieve a class-averaged Jaccard coefficient of 0.7047, and 0.9044 for tumor in particular. For classification, we achieve a slide-level accuracy of 0.9857. Since canine cutaneous tumors possess various histologic homologies to human tumors the added value of this dataset is not limited to veterinary pathology but extends to more general fields of application
    • …
    corecore