1
|
Ren S, Li J, Dorado J, Sierra A, González-Díaz H, Duardo A, Shen B. From molecular mechanisms of prostate cancer to translational applications: based on multi-omics fusion analysis and intelligent medicine. Health Inf Sci Syst 2024; 12:6. [PMID: 38125666 PMCID: PMC10728428 DOI: 10.1007/s13755-023-00264-5] [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: 08/24/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Prostate cancer is the most common cancer in men worldwide and has a high mortality rate. The complex and heterogeneous development of prostate cancer has become a core obstacle in the treatment of prostate cancer. Simultaneously, the issues of overtreatment in early-stage diagnosis, oligometastasis and dormant tumor recognition, as well as personalized drug utilization, are also specific concerns that require attention in the clinical management of prostate cancer. Some typical genetic mutations have been proved to be associated with prostate cancer's initiation and progression. However, single-omic studies usually are not able to explain the causal relationship between molecular alterations and clinical phenotypes. Exploration from a systems genetics perspective is also lacking in this field, that is, the impact of gene network, the environmental factors, and even lifestyle behaviors on disease progression. At the meantime, current trend emphasizes the utilization of artificial intelligence (AI) and machine learning techniques to process extensive multidimensional data, including multi-omics. These technologies unveil the potential patterns, correlations, and insights related to diseases, thereby aiding the interpretable clinical decision making and applications, namely intelligent medicine. Therefore, there is a pressing need to integrate multidimensional data for identification of molecular subtypes, prediction of cancer progression and aggressiveness, along with perosonalized treatment performing. In this review, we systematically elaborated the landscape from molecular mechanism discovery of prostate cancer to clinical translational applications. We discussed the molecular profiles and clinical manifestations of prostate cancer heterogeneity, the identification of different states of prostate cancer, as well as corresponding precision medicine practices. Taking multi-omics fusion, systems genetics, and intelligence medicine as the main perspectives, the current research results and knowledge-driven research path of prostate cancer were summarized.
Collapse
Affiliation(s)
- Shumin Ren
- Department of Urology and Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041 China
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
| | - Jiakun Li
- Department of Urology and Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Julián Dorado
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
| | - Alejandro Sierra
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
- IKERDATA S.L., ZITEK, University of Basque Country UPVEHU, Rectorate Building, 48940 Leioa, Spain
| | - Humbert González-Díaz
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
- IKERDATA S.L., ZITEK, University of Basque Country UPVEHU, Rectorate Building, 48940 Leioa, Spain
| | - Aliuska Duardo
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
- IKERDATA S.L., ZITEK, University of Basque Country UPVEHU, Rectorate Building, 48940 Leioa, Spain
| | - Bairong Shen
- Department of Urology and Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041 China
| |
Collapse
|
2
|
Maeda K, Ogawa T, Kayama T, Sasaki T, Tainaka K, Murakami M, Haseyama M. Trial Analysis of Brain Activity Information for the Presymptomatic Disease Detection of Rheumatoid Arthritis. Bioengineering (Basel) 2024; 11:523. [PMID: 38927759 PMCID: PMC11200460 DOI: 10.3390/bioengineering11060523] [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: 03/21/2024] [Revised: 04/26/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
This study presents a trial analysis that uses brain activity information obtained from mice to detect rheumatoid arthritis (RA) in its presymptomatic stages. Specifically, we confirmed that F759 mice, serving as a mouse model of RA that is dependent on the inflammatory cytokine IL-6, and healthy wild-type mice can be classified on the basis of brain activity information. We clarified which brain regions are useful for the presymptomatic detection of RA. We introduced a matrix completion-based approach to handle missing brain activity information to perform the aforementioned analysis. In addition, we implemented a canonical correlation-based method capable of analyzing the relationship between various types of brain activity information. This method allowed us to accurately classify F759 and wild-type mice, thereby identifying essential features, including crucial brain regions, for the presymptomatic detection of RA. Our experiment obtained brain activity information from 15 F759 and 10 wild-type mice and analyzed the acquired data. By employing four types of classifiers, our experimental results show that the thalamus and periaqueductal gray are effective for the classification task. Furthermore, we confirmed that classification performance was maximized when seven brain regions were used, excluding the electromyogram and nucleus accumbens.
Collapse
Affiliation(s)
- Keisuke Maeda
- Data-Driven Interdisciplinary Research Emergence Department, Hokkaido University, N-13, W-10, Kita-ku, Sapporo 060-0813, Japan;
| | - Takahiro Ogawa
- Faculty of Information Science and Technology, Hokkaido University, N-14, W-9, Kita-ku, Sapporo 060-0814, Japan;
| | - Tasuku Kayama
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai 980-8578, Japan; (T.K.); (T.S.)
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai 980-8578, Japan; (T.K.); (T.S.)
- Department of Neuropharmacology, Tohoku University School of Medicine, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8585, Japan;
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-0815, Japan;
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, Myodaiji, Okazaki 444-8585, Japan
- Group of Quantum Immunology, National Institute for Quantum and Radiological Science and Technology (QST), 4-9-1 Anagawa, Inage 263-8555, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo 001-0021, Japan
| | - Miki Haseyama
- Faculty of Information Science and Technology, Hokkaido University, N-14, W-9, Kita-ku, Sapporo 060-0814, Japan;
| |
Collapse
|
3
|
Hiremath A, Corredor G, Li L, Leo P, Magi-Galluzzi C, Elliott R, Purysko A, Shiradkar R, Madabhushi A. An integrated radiology-pathology machine learning classifier for outcome prediction following radical prostatectomy: Preliminary findings. Heliyon 2024; 10:e29602. [PMID: 38665576 PMCID: PMC11044050 DOI: 10.1016/j.heliyon.2024.e29602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Objectives To evaluate the added benefit of integrating features from pre-treatment MRI (radiomics) and digitized post-surgical pathology slides (pathomics) in prostate cancer (PCa) patients for prognosticating outcomes post radical-prostatectomy (RP) including a) rising prostate specific antigen (PSA), and b) extraprostatic-extension (EPE). Methods Multi-institutional data (N = 58) of PCa patients who underwent pre-treatment 3-T MRI prior to RP were included in this retrospective study. Radiomic and pathomic features were extracted from PCa regions on MRI and RP specimens delineated by expert clinicians. On training set (D1, N = 44), Cox Proportional-Hazards models MR, MP and MRaP were trained using radiomics, pathomics, and their combination, respectively, to prognosticate rising PSA (PSA > 0.03 ng/mL). Top features from MRaP were used to train a model to predict EPE on D1 and test on external dataset (D2, N = 14). C-index, Kalplan-Meier curves were used for survival analysis, and area under ROC (AUC) was used for EPE. MRaP was compared with the existing post-treatment risk-calculator, CAPRA (MC). Results Patients had median follow-up of 34 months. MRaP (c-index = 0.685 ± 0.05) significantly outperformed MR (c-index = 0.646 ± 0.05), MP (c-index = 0.631 ± 0.06) and MC (c-index = 0.601 ± 0.071) (p < 0.0001). Cross-validated Kaplan-Meier curves showed significant separation among risk groups for rising PSA for MRaP (p < 0.005, Hazard Ratio (HR) = 11.36) as compared to MR (p = 0.64, HR = 1.33), MP (p = 0.19, HR = 2.82) and MC (p = 0.10, HR = 3.05). Integrated radio-pathomic model MRaP (AUC = 0.80) outperformed MR (AUC = 0.57) and MP (AUC = 0.76) in predicting EPE on external-data (D2). Conclusions Results from this preliminary study suggest that a combination of radiomic and pathomic features can better predict post-surgical outcomes (rising PSA and EPE) compared to either of them individually as well as extant prognostic nomogram (CAPRA).
Collapse
Affiliation(s)
| | - Germán Corredor
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Lin Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Patrick Leo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | | | - Robin Elliott
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Andrei Purysko
- Department of Radiology and Nuclear Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Rakesh Shiradkar
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Atlanta Veterans Administration Medical Center, Atlanta, GA, USA
| |
Collapse
|
4
|
Affaticati F, Bartholomeus E, Mullan K, Damme PV, Beutels P, Ogunjimi B, Laukens K, Meysman P. Multi-View Learning to Unravel the Different Levels Underlying Hepatitis B Vaccine Response. Vaccines (Basel) 2023; 11:1236. [PMID: 37515051 PMCID: PMC10384938 DOI: 10.3390/vaccines11071236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
The immune system acts as an intricate apparatus that is dedicated to mounting a defense and ensures host survival from microbial threats. To engage this faceted immune response and provide protection against infectious diseases, vaccinations are a critical tool to be developed. However, vaccine responses are governed by levels that, when interrogated, separately only explain a fraction of the immune reaction. To address this knowledge gap, we conducted a feasibility study to determine if multi-view modeling could aid in gaining actionable insights on response markers shared across populations, capture the immune system's diversity, and disentangle confounders. We thus sought to assess this multi-view modeling capacity on the responsiveness to the Hepatitis B virus (HBV) vaccination. Seroconversion to vaccine-induced antibodies against the HBV surface antigen (anti-HBs) in early converters (n = 21; <2 months) and late converters (n = 9; <6 months) and was defined based on the anti-HBs titers (>10IU/L). The multi-view data encompassed bulk RNA-seq, CD4+ T-cell parameters (including T-cell receptor data), flow cytometry data, and clinical metadata (including age and gender). The modeling included testing single-view and multi-view joint dimensionality reductions. Multi-view joint dimensionality reduction outperformed single-view methods in terms of the area under the curve and balanced accuracy, confirming the increase in predictive power to be gained. The interpretation of these findings showed that age, gender, inflammation-related gene sets, and pre-existing vaccine-specific T-cells could be associated with vaccination responsiveness. This multi-view dimensionality reduction approach complements clinical seroconversion and all single modalities. Importantly, this modeling could identify what features could predict HBV vaccine response. This methodology could be extended to other vaccination trials to identify the key features regulating responsiveness.
Collapse
Affiliation(s)
- Fabio Affaticati
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| | - Esther Bartholomeus
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
- Centre for Health Economics Research & Modeling Infectious Diseases (CHERMID), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, 2610 Antwerp, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp (VAXINFECTIO), 2610 Antwerp, Belgium
| | - Kerry Mullan
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| | - Pierre Van Damme
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
- Centre for the Evaluation of Vaccination (CEV), Vaccine and Infectious Disease Institute, University of Antwerp, 2610 Antwerp, Belgium
| | - Philippe Beutels
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
- Centre for Health Economics Research & Modeling Infectious Diseases (CHERMID), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, 2610 Antwerp, Belgium
| | - Benson Ogunjimi
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
- Centre for Health Economics Research & Modeling Infectious Diseases (CHERMID), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, 2610 Antwerp, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp (VAXINFECTIO), 2610 Antwerp, Belgium
- Department of Paediatrics, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Kris Laukens
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| | - Pieter Meysman
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| |
Collapse
|
5
|
Chervenkov L, Sirakov N, Kostov G, Velikova T, Hadjidekov G. Future of prostate imaging: Artificial intelligence in assessing prostatic magnetic resonance imaging. World J Radiol 2023; 15:136-145. [PMID: 37275303 PMCID: PMC10236970 DOI: 10.4329/wjr.v15.i5.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/21/2023] [Accepted: 04/10/2023] [Indexed: 05/23/2023] Open
Abstract
Prostate cancer (Pca; adenocarcinoma) is one of the most common cancers in adult males and one of the leading causes of death in both men and women. The diagnosis of Pca requires substantial experience, and even then the lesions can be difficult to detect. Moreover, although the diagnostic approach for this disease has improved significantly with the advent of multiparametric magnetic resonance, that technology has certain unresolved limitations. In recent years artificial intelligence (AI) has been introduced to the field of radiology, providing new software solutions for prostate diagnostics. Precise mapping of the prostate has become possible through AI and this has greatly improved the accuracy of biopsy. AI has also allowed for certain suspicious lesions to be attributed to a given group according to the Prostate Imaging-Reporting & Data System classification. Finally, AI has facilitated the combination of data obtained from clinical, laboratory (prostate-specific antigen), imaging (magnetic resonance), and biopsy examinations, and in this way new regularities can be found which at the moment remain hidden. Further evolution of AI in this field is inevitable and it is almost certain to significantly expand the efficacy, accuracy and efficiency of diagnosis and treatment of Pca.
Collapse
Affiliation(s)
- Lyubomir Chervenkov
- Department of Diagnostic Imaging, Medical University Plovdiv, Plovdiv 4000, Bulgaria
- Research Complex for Translational Neuroscience, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, Plovdiv 4002, Bulgaria
| | - Nikolay Sirakov
- Research Complex for Translational Neuroscience, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, Plovdiv 4002, Bulgaria
- Department of Diagnostic Imaging, Dental Allergology and Physiotherapy, Faculty of Dental Medicine, Medical University Plovdiv, Plovdiv 4000, Bulgaria
| | - Gancho Kostov
- Department of Special Surgery, Medical University Plovdiv, Plovdiv 4000, Bulgaria
| | - Tsvetelina Velikova
- Department of Clinical Immunology, University Hospital Lozenetz, Sofia 1407, Bulgaria
- Department of Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - George Hadjidekov
- Department of Radiology, University Hospital Lozenetz, Sofia 1407, Bulgaria
- Department of Physics, Biophysics and Radiology, Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| |
Collapse
|
6
|
Huang W, Tan K, Zhang Z, Hu J, Dong S. A Review of Fusion Methods for Omics and Imaging Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:74-93. [PMID: 35044920 DOI: 10.1109/tcbb.2022.3143900] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The development of omics data and biomedical images has greatly advanced the progress of precision medicine in diagnosis, treatment, and prognosis. The fusion of omics and imaging data, i.e., omics-imaging fusion, offers a new strategy for understanding complex diseases. However, due to a variety of issues such as the limited number of samples, high dimensionality of features, and heterogeneity of different data types, efficiently learning complementary or associated discriminative fusion information from omics and imaging data remains a challenge. Recently, numerous machine learning methods have been proposed to alleviate these problems. In this review, from the perspective of fusion levels and fusion methods, we first provide an overview of preprocessing and feature extraction methods for omics and imaging data, and comprehensively analyze and summarize the basic forms and variations of commonly used and newly emerging fusion methods, along with their advantages, disadvantages and the applicable scope. We then describe public datasets and compare experimental results of various fusion methods on the ADNI and TCGA datasets. Finally, we discuss future prospects and highlight remaining challenges in the field.
Collapse
|
7
|
Guo YB, Zheng ZX, Kong LJ, Guo W, Yan ZM, Cui LZ, Xiao-Fang Wang A. A Novel Multi-view Bi-clustering method for identifying abnormal Co-occurrence medical visit behaviors. Methods 2022; 207:65-73. [PMID: 36122881 DOI: 10.1016/j.ymeth.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/05/2022] [Accepted: 09/12/2022] [Indexed: 10/31/2022] Open
Abstract
Abnormal co-occurrence medical visit behavior is a form of medical insurance fraud. Specifically, an organized gang of fraudsters hold multiple medical insurance cards and purchase similar drugs frequently at the same time and the same location in order to siphon off medical insurance funds. Conventional identification methods to identify such behaviors rely mainly on manual auditing, making it difficult to satisfy the needs of identifying the small number of fraudulent behaviors in the large-scale medical data. On the other hand, the existing single-view bi-clustering algorithms only consider the features of the time-location dimension while neglecting the similarities in prescriptions and neglecting the fact that fraudsters may belong to multiple gangs. Therefore, in this paper, we present a multi-view bi-clustering method for identifying abnormal co-occurrence medical visit behavioral patterns, which performs cluster analysis simultaneously on the large-scale, complex and diverse visiting record dimension and prescription dimension to identify bi-clusters with similar time-location features. The proposed method constructs a matrix view of patients and visit records as well as a matrix view of patients and prescriptions, while decomposing multiple data matrices into sparse row and column vectors to obtain a consistent patient population across views. Subsequently the proposed method identifies the corresponding abnormal co-occurrence medical visit behavior and may greatly facilitate the safe operations and the sustainability of medical insurance funds. The experimental results show that our proposed method leads to more efficient and more accurate identifications of abnormal co-occurrence medical visit behavior, demonstrating its high efficiency and effectiveness.
Collapse
Affiliation(s)
- Yu-Bing Guo
- Research Center of Software and Data Engineering, School of Software, Shandong University, Jinan, China
| | - Zi-Xin Zheng
- Research Center of Software and Data Engineering, School of Software, Shandong University, Jinan, China
| | - Lan-Ju Kong
- Research Center of Software and Data Engineering, School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China.
| | - Wei Guo
- Research Center of Software and Data Engineering, School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China
| | - Zhong-Min Yan
- Research Center of Software and Data Engineering, School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China
| | - Li-Zhen Cui
- Research Center of Software and Data Engineering, School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China
| | - And Xiao-Fang Wang
- Shandong Provincial Key Laboratory of Network Based Intelligent Computing, University of Jinan, Jinan, China
| |
Collapse
|
8
|
Sutera P, Deek MP, Van der Eecken K, Wyatt AW, Kishan AU, Molitoris JK, Ferris MJ, Minhaj Siddiqui M, Rana Z, Mishra MV, Kwok Y, Davicioni E, Spratt DE, Ost P, Feng FY, Tran PT. Genomic biomarkers to guide precision radiotherapy in prostate cancer. Prostate 2022; 82 Suppl 1:S73-S85. [PMID: 35657158 PMCID: PMC9202472 DOI: 10.1002/pros.24373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/30/2022] [Accepted: 04/29/2022] [Indexed: 11/08/2022]
Abstract
Our ability to prognosticate the clinical course of patients with cancer has historically been limited to clinical, histopathological, and radiographic features. It has long been clear however, that these data alone do not adequately capture the heterogeneity and breadth of disease trajectories experienced by patients. The advent of efficient genomic sequencing has led to a revolution in cancer care as we try to understand and personalize treatment specific to patient clinico-genomic phenotypes. Within prostate cancer, emerging evidence suggests that tumor genomics (e.g., DNA, RNA, and epigenetics) can be utilized to inform clinical decision making. In addition to providing discriminatory information about prognosis, it is likely tumor genomics also hold a key in predicting response to oncologic therapies which could be used to further tailor treatment recommendations. Herein we review select literature surrounding the use of tumor genomics within the management of prostate cancer, specifically leaning toward analytically validated and clinically tested genomic biomarkers utilized in radiotherapy and/or adjunctive therapies given with radiotherapy.
Collapse
Affiliation(s)
- Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew P. Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Kim Van der Eecken
- Department of Pathology, Ghent University Hospital, Cancer Research Institute (CRIG), Ghent, Belgium
| | - Alexander W. Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amar U. Kishan
- Department of Radiation Oncology, UCLA, Los Angeles, CA, USA
| | - Jason K. Molitoris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew J. Ferris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - M. Minhaj Siddiqui
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zaker Rana
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark V. Mishra
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Young Kwok
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Daniel E. Spratt
- Department of Radiation Oncology, University Hospitals, Cleveland, OH, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, Antwerp, Belgium and Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Felix Y. Feng
- Departments of Radiation Oncology, Medicine and Urology, UCSF, San Francisco, CA, USA
| | - Phuoc T. Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Neonatal encephalopathy prediction of poor outcome with diffusion-weighted imaging connectome and fixel-based analysis. Pediatr Res 2022; 91:1505-1515. [PMID: 33966055 PMCID: PMC9053106 DOI: 10.1038/s41390-021-01550-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/01/2021] [Accepted: 04/08/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Better biomarkers of eventual outcome are needed for neonatal encephalopathy. To identify the most potent neonatal imaging marker associated with 2-year outcomes, we retrospectively performed diffusion-weighted imaging connectome (DWIC) and fixel-based analysis (FBA) on magnetic resonance imaging (MRI) obtained in the first 4 weeks of life in term neonatal encephalopathy newborns. METHODS Diffusion tractography was available in 15 out of 24 babies with MRI, five each with normal, abnormal motor outcome, or death. All 15 except one underwent hypothermia as initial treatment. In abnormal motor and death groups, DWIC found 19 white matter pathways with severely disrupted fiber orientation distributions. RESULTS Using random forest classification, these disruptions predicted the follow-up outcomes with 89-99% accuracy. These pathways showed reduced integrity in abnormal motor and death vs. normal tone groups (p < 10-6). Using ranked supervised multi-view canonical correlation and depicting just three of the five dimensions of the analysis, the abnormal motor and death were clearly differentiated from each other and the normal tone group. CONCLUSIONS This study suggests that a machine-learning model for prediction using early DWIC and FBA could be a possible way of developing biomarkers in large MRI datasets having clinical outcomes. IMPACT Early connectome and FBA of clinically acquired DWI provide a new noninvasive imaging tool to predict the long-term motor outcomes after birth, based on the severity of white matter injury. Disrupted white matter connectivity as a novel neonatal marker achieves high accuracy of 89-99% to predict 2-year motor outcomes using conventional machine-learning classification. The proposed neonatal marker may allow better prognostication that is important to elucidate neural repair mechanisms and evaluate treatment modalities in neonatal encephalopathy.
Collapse
|
10
|
Xie W, Reder NP, Koyuncu C, Leo P, Hawley S, Huang H, Mao C, Postupna N, Kang S, Serafin R, Gao G, Han Q, Bishop KW, Barner LA, Fu P, Wright JL, Keene CD, Vaughan JC, Janowczyk A, Glaser AK, Madabhushi A, True LD, Liu JTC. Prostate Cancer Risk Stratification via Nondestructive 3D Pathology with Deep Learning-Assisted Gland Analysis. Cancer Res 2022; 82:334-345. [PMID: 34853071 PMCID: PMC8803395 DOI: 10.1158/0008-5472.can-21-2843] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/19/2021] [Accepted: 11/24/2021] [Indexed: 01/07/2023]
Abstract
Prostate cancer treatment planning is largely dependent upon examination of core-needle biopsies. The microscopic architecture of the prostate glands forms the basis for prognostic grading by pathologists. Interpretation of these convoluted three-dimensional (3D) glandular structures via visual inspection of a limited number of two-dimensional (2D) histology sections is often unreliable, which contributes to the under- and overtreatment of patients. To improve risk assessment and treatment decisions, we have developed a workflow for nondestructive 3D pathology and computational analysis of whole prostate biopsies labeled with a rapid and inexpensive fluorescent analogue of standard hematoxylin and eosin (H&E) staining. This analysis is based on interpretable glandular features and is facilitated by the development of image translation-assisted segmentation in 3D (ITAS3D). ITAS3D is a generalizable deep learning-based strategy that enables tissue microstructures to be volumetrically segmented in an annotation-free and objective (biomarker-based) manner without requiring immunolabeling. As a preliminary demonstration of the translational value of a computational 3D versus a computational 2D pathology approach, we imaged 300 ex vivo biopsies extracted from 50 archived radical prostatectomy specimens, of which, 118 biopsies contained cancer. The 3D glandular features in cancer biopsies were superior to corresponding 2D features for risk stratification of patients with low- to intermediate-risk prostate cancer based on their clinical biochemical recurrence outcomes. The results of this study support the use of computational 3D pathology for guiding the clinical management of prostate cancer. SIGNIFICANCE: An end-to-end pipeline for deep learning-assisted computational 3D histology analysis of whole prostate biopsies shows that nondestructive 3D pathology has the potential to enable superior prognostic stratification of patients with prostate cancer.
Collapse
Affiliation(s)
- Weisi Xie
- Department of Mechanical Engineering, University of Washington, Seattle, Washington
| | - Nicholas P Reder
- Department of Mechanical Engineering, University of Washington, Seattle, Washington
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington
| | - Can Koyuncu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Patrick Leo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | | | - Hongyi Huang
- Department of Mechanical Engineering, University of Washington, Seattle, Washington
| | - Chenyi Mao
- Department of Chemistry, University of Washington, Seattle, Washington
| | - Nadia Postupna
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington
| | - Soyoung Kang
- Department of Mechanical Engineering, University of Washington, Seattle, Washington
| | - Robert Serafin
- Department of Mechanical Engineering, University of Washington, Seattle, Washington
| | - Gan Gao
- Department of Mechanical Engineering, University of Washington, Seattle, Washington
| | - Qinghua Han
- Department of Bioengineering, University of Washington, Seattle, Washington
| | - Kevin W Bishop
- Department of Mechanical Engineering, University of Washington, Seattle, Washington
- Department of Bioengineering, University of Washington, Seattle, Washington
| | - Lindsey A Barner
- Department of Mechanical Engineering, University of Washington, Seattle, Washington
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Jonathan L Wright
- Department of Urology, University of Washington, Seattle, Washington
| | - C Dirk Keene
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, Washington
- Department of Physiology & Biophysics, Seattle, Washington
| | - Andrew Janowczyk
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
- Department of Oncology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Adam K Glaser
- Department of Mechanical Engineering, University of Washington, Seattle, Washington
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
- Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, Ohio
| | - Lawrence D True
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington
- Department of Urology, University of Washington, Seattle, Washington
| | - Jonathan T C Liu
- Department of Mechanical Engineering, University of Washington, Seattle, Washington.
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington
- Department of Bioengineering, University of Washington, Seattle, Washington
| |
Collapse
|
11
|
Mata LA, Retamero JA, Gupta RT, García Figueras R, Luna A. Artificial Intelligence-assisted Prostate Cancer Diagnosis: Radiologic-Pathologic Correlation. Radiographics 2021; 41:1676-1697. [PMID: 34597215 DOI: 10.1148/rg.2021210020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The classic prostate cancer (PCa) diagnostic pathway that is based on prostate-specific antigen (PSA) levels and the findings of digital rectal examination followed by systematic biopsy has shown multiple limitations. The use of multiparametric MRI (mpMRI) is now widely accepted in men with clinical suspicion for PCa. In addition, clinical information, PSA density, risk calculators, and genomic and other "omics" biomarkers are being used to improve risk stratification. On the basis of mpMRI and MRI-targeted biopsies (MRI-TBx), new diagnostic pathways have been established, aiming to improve the limitations of the classic diagnostic approach. However, these pathways still show limitations associated with mpMRI and MRI-TBx. Definitive PCa diagnosis is made on the basis of histopathologic Gleason grading, which has demonstrated an excellent correlation with clinical outcomes. However, Gleason grading is done subjectively by pathologists and involves poor reproducibility, and PCa may have a heterogeneous distribution of histologic patterns. Thus, important discrepancies persist between biopsy tumor grading and final whole-organ pathologic assessment after radical prostatectomy. PCa offers a unique opportunity to establish a real radiologic-pathologic correlation, as whole-mount radical prostatectomy specimens permit a complete spatial relationship with mpMRI. Artificial intelligence is increasingly being applied to radiologic and pathologic images to improve clinical accuracy and efficiency in PCa diagnosis. This review delineates current PCa diagnostic pathways, with a focus on the role of mpMRI, MRI-TBx, and pathologic analysis. An overview of the expected improvements in PCa diagnosis derived from the use of artificial intelligence, integrated radiologic-pathologic systems, and decision support tools for multidisciplinary teams is provided. An invited commentary by Purysko is available online. Online supplemental material is available for this article. ©RSNA, 2021.
Collapse
Affiliation(s)
- Lidia Alcalá Mata
- From the Department of Radiology, Clínica Las Nieves, HT Médica, Calle Carmelo Torres Núm 2, 23007 Jaén, Spain (L.A.M., A.L.); Paige.AI, New York, NY (J.A.R.); Department of Radiology, Duke University Medical Center, Durham, NC (R.T.G.); and Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain (R.G.F.)
| | - Juan Antonio Retamero
- From the Department of Radiology, Clínica Las Nieves, HT Médica, Calle Carmelo Torres Núm 2, 23007 Jaén, Spain (L.A.M., A.L.); Paige.AI, New York, NY (J.A.R.); Department of Radiology, Duke University Medical Center, Durham, NC (R.T.G.); and Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain (R.G.F.)
| | - Rajan T Gupta
- From the Department of Radiology, Clínica Las Nieves, HT Médica, Calle Carmelo Torres Núm 2, 23007 Jaén, Spain (L.A.M., A.L.); Paige.AI, New York, NY (J.A.R.); Department of Radiology, Duke University Medical Center, Durham, NC (R.T.G.); and Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain (R.G.F.)
| | - Roberto García Figueras
- From the Department of Radiology, Clínica Las Nieves, HT Médica, Calle Carmelo Torres Núm 2, 23007 Jaén, Spain (L.A.M., A.L.); Paige.AI, New York, NY (J.A.R.); Department of Radiology, Duke University Medical Center, Durham, NC (R.T.G.); and Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain (R.G.F.)
| | - Antonio Luna
- From the Department of Radiology, Clínica Las Nieves, HT Médica, Calle Carmelo Torres Núm 2, 23007 Jaén, Spain (L.A.M., A.L.); Paige.AI, New York, NY (J.A.R.); Department of Radiology, Duke University Medical Center, Durham, NC (R.T.G.); and Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain (R.G.F.)
| |
Collapse
|
12
|
Leo P, Janowczyk A, Elliott R, Janaki N, Bera K, Shiradkar R, Farré X, Fu P, El-Fahmawi A, Shahait M, Kim J, Lee D, Yamoah K, Rebbeck TR, Khani F, Robinson BD, Eklund L, Jambor I, Merisaari H, Ettala O, Taimen P, Aronen HJ, Boström PJ, Tewari A, Magi-Galluzzi C, Klein E, Purysko A, Nc Shih N, Feldman M, Gupta S, Lal P, Madabhushi A. Computer extracted gland features from H&E predicts prostate cancer recurrence comparably to a genomic companion diagnostic test: a large multi-site study. NPJ Precis Oncol 2021; 5:35. [PMID: 33941830 PMCID: PMC8093226 DOI: 10.1038/s41698-021-00174-3] [Citation(s) in RCA: 12] [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: 08/20/2020] [Accepted: 04/05/2021] [Indexed: 01/04/2023] Open
Abstract
Existing tools for post-radical prostatectomy (RP) prostate cancer biochemical recurrence (BCR) prognosis rely on human pathologist-derived parameters such as tumor grade, with the resulting inter-reviewer variability. Genomic companion diagnostic tests such as Decipher tend to be tissue destructive, expensive, and not routinely available in most centers. We present a tissue non-destructive method for automated BCR prognosis, termed "Histotyping", that employs computational image analysis of morphologic patterns of prostate tissue from a single, routinely acquired hematoxylin and eosin slide. Patients from two institutions (n = 214) were used to train Histotyping for identifying high-risk patients based on six features of glandular morphology extracted from RP specimens. Histotyping was validated for post-RP BCR prognosis on a separate set of n = 675 patients from five institutions and compared against Decipher on n = 167 patients. Histotyping was prognostic of BCR in the validation set (p < 0.001, univariable hazard ratio [HR] = 2.83, 95% confidence interval [CI]: 2.03-3.93, concordance index [c-index] = 0.68, median years-to-BCR: 1.7). Histotyping was also prognostic in clinically stratified subsets, such as patients with Gleason grade group 3 (HR = 4.09) and negative surgical margins (HR = 3.26). Histotyping was prognostic independent of grade group, margin status, pathological stage, and preoperative prostate-specific antigen (PSA) (multivariable p < 0.001, HR = 2.09, 95% CI: 1.40-3.10, n = 648). The combination of Histotyping, grade group, and preoperative PSA outperformed Decipher (c-index = 0.75 vs. 0.70, n = 167). These results suggest that a prognostic classifier for prostate cancer based on digital images could serve as an alternative or complement to molecular-based companion diagnostic tests.
Collapse
Grants
- National Cancer Institute under award numbers 1U24CA199374-01, R01CA249992-01A1 R01CA202752-01A1 R01CA208236-01A1 R01CA216579-01A1 R01CA220581-01A1 1U01CA239055-01 1U01CA248226-01 1U54CA254566-01 National Heart, Lung and Blood Institute 1R01HL15127701A1, National Institute for Biomedical Imaging and Bioengineering 1R43EB028736-01, National Center for Research Resources 1 C06 RR12463-01, VA Merit Review Award IBX004121A from the United States Department of Veterans Affairs Biomedical Laboratory Research and Development Service, the Office of the Assistant Secretary of Defense for Health Affairs, through the Breast Cancer Research Program (W81XWH-19-1-0668), the Prostate Cancer Research Program (W81XWH-15-1-0558, W81XWH-20-1-0851), the Lung Cancer Research Program (W81XWH-18-1-0440, W81XWH-20-1-0595), the Peer Reviewed Cancer Research Program (W81XWH-18-1-0404), the Kidney Precision Medicine Project Glue Grant, the Ohio Third Frontier Technology Validation Fund, the Clinical and Translational Science Collaborative of Cleveland (UL1TR0002548) from the National Center for Advancing Translational Sciences (NCATS) component of the National Institutes of Health and NIH roadmap for Medical Research, The Wallace H. Coulter Foundation Program in the Department of Biomedical Engineering at Case Western Reserve University,
- Sigrid Jusélius Foundation The Finnish Cancer Foundation
- Department of Defense Prostate Cancer Disparity Award (W81XWH-19-1-0720),
- National Science Foundation Graduate Research Fellowship Program (CON501692)
Collapse
Affiliation(s)
- Patrick Leo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew Janowczyk
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Oncology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Robin Elliott
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Nafiseh Janaki
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Kaustav Bera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Rakesh Shiradkar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Xavier Farré
- Public Health Agency of Catalonia, Lleida, Catalonia, Spain
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Ayah El-Fahmawi
- Department of Urology, Penn Presbyterian Medical Center, Philadelphia, PA, USA
| | - Mohammed Shahait
- Department of Urology, Penn Presbyterian Medical Center, Philadelphia, PA, USA
| | - Jessica Kim
- Department of Urology, Penn Presbyterian Medical Center, Philadelphia, PA, USA
| | - David Lee
- Department of Urology, Penn Presbyterian Medical Center, Philadelphia, PA, USA
| | - Kosj Yamoah
- Moffitt Cancer Center, Department of Radiation Oncology, University of South Florida, Tampa, FL, USA
| | - Timothy R Rebbeck
- T.H. Chan School of Public Health and Dana Farber Cancer Institute, Harvard University, Boston, MA, USA
| | - Francesca Khani
- Departments of Pathology and Laboratory Medicine and Urology, Weill Cornell Medicine, New York, NY, USA
| | - Brian D Robinson
- Departments of Pathology and Laboratory Medicine and Urology, Weill Cornell Medicine, New York, NY, USA
| | - Lauri Eklund
- Department of Pathology, University of Turku, Institute of Biomedicine and Turku University Hospital, Turku, Finland
| | - Ivan Jambor
- Department of Pathology, University of Turku, Institute of Biomedicine and Turku University Hospital, Turku, Finland
- Department of Diagnostic Radiology, University of Turku, Turku, Finland
| | - Harri Merisaari
- Department of Pathology, University of Turku, Institute of Biomedicine and Turku University Hospital, Turku, Finland
| | - Otto Ettala
- Department of Urology, University of Turku, Institute of Biomedicine and Turku University Hospital, Turku, Finland
| | - Pekka Taimen
- Department of Pathology, University of Turku, Institute of Biomedicine and Turku University Hospital, Turku, Finland
| | - Hannu J Aronen
- Department of Pathology, University of Turku, Institute of Biomedicine and Turku University Hospital, Turku, Finland
- Turku University Hospital, Medical Imaging Centre of Southwest Finland, Turku, Finland
| | - Peter J Boström
- Department of Urology, University of Turku and Turku University Hospital, Turku, Finland
| | - Ashutosh Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Eric Klein
- Cleveland Clinic, Glickman Urological and Kidney Institute, Cleveland, OH, USA
| | - Andrei Purysko
- Cleveland Clinic, Imaging Institute, Section of Abdominal Imaging, Cleveland, OH, USA
| | - Natalie Nc Shih
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Feldman
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, OH, USA
- Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH, USA
| | - Priti Lal
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
- Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH, USA.
| |
Collapse
|
13
|
Ayyad SM, Shehata M, Shalaby A, Abou El-Ghar M, Ghazal M, El-Melegy M, Abdel-Hamid NB, Labib LM, Ali HA, El-Baz A. Role of AI and Histopathological Images in Detecting Prostate Cancer: A Survey. SENSORS (BASEL, SWITZERLAND) 2021; 21:2586. [PMID: 33917035 PMCID: PMC8067693 DOI: 10.3390/s21082586] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/29/2021] [Accepted: 04/04/2021] [Indexed: 02/07/2023]
Abstract
Prostate cancer is one of the most identified cancers and second most prevalent among cancer-related deaths of men worldwide. Early diagnosis and treatment are substantial to stop or handle the increase and spread of cancer cells in the body. Histopathological image diagnosis is a gold standard for detecting prostate cancer as it has different visual characteristics but interpreting those type of images needs a high level of expertise and takes too much time. One of the ways to accelerate such an analysis is by employing artificial intelligence (AI) through the use of computer-aided diagnosis (CAD) systems. The recent developments in artificial intelligence along with its sub-fields of conventional machine learning and deep learning provide new insights to clinicians and researchers, and an abundance of research is presented specifically for histopathology images tailored for prostate cancer. However, there is a lack of comprehensive surveys that focus on prostate cancer using histopathology images. In this paper, we provide a very comprehensive review of most, if not all, studies that handled the prostate cancer diagnosis using histopathological images. The survey begins with an overview of histopathological image preparation and its challenges. We also briefly review the computing techniques that are commonly applied in image processing, segmentation, feature selection, and classification that can help in detecting prostate malignancies in histopathological images.
Collapse
Affiliation(s)
- Sarah M. Ayyad
- Computers and Systems Department, Faculty of Engineering, Mansoura University, Mansoura 35511, Egypt; (S.M.A.); (N.B.A.-H.); (L.M.L.); (H.A.A.)
| | - Mohamed Shehata
- BioImaging Laboratory, Bioengineering Department, University of Louisville, Louisville, KY 40292, USA; (M.S.); (A.S.)
| | - Ahmed Shalaby
- BioImaging Laboratory, Bioengineering Department, University of Louisville, Louisville, KY 40292, USA; (M.S.); (A.S.)
| | - Mohamed Abou El-Ghar
- Department of Radiology, Urology and Nephrology Center, Mansoura University, Mansoura 35516, Egypt;
| | - Mohammed Ghazal
- Department of Electrical and Computer Engineering, College of Engineering, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates;
| | - Moumen El-Melegy
- Department of Electrical Engineering, Assiut University, Assiut 71511, Egypt;
| | - Nahla B. Abdel-Hamid
- Computers and Systems Department, Faculty of Engineering, Mansoura University, Mansoura 35511, Egypt; (S.M.A.); (N.B.A.-H.); (L.M.L.); (H.A.A.)
| | - Labib M. Labib
- Computers and Systems Department, Faculty of Engineering, Mansoura University, Mansoura 35511, Egypt; (S.M.A.); (N.B.A.-H.); (L.M.L.); (H.A.A.)
| | - H. Arafat Ali
- Computers and Systems Department, Faculty of Engineering, Mansoura University, Mansoura 35511, Egypt; (S.M.A.); (N.B.A.-H.); (L.M.L.); (H.A.A.)
| | - Ayman El-Baz
- BioImaging Laboratory, Bioengineering Department, University of Louisville, Louisville, KY 40292, USA; (M.S.); (A.S.)
| |
Collapse
|
14
|
Hameed BMZ, Shah M, Naik N, Ibrahim S, Somani B, Rice P, Soomro N, Rai BP. Contemporary application of artificial intelligence in prostate cancer: an i-TRUE study. Ther Adv Urol 2021; 13:1756287220986640. [PMID: 33633799 PMCID: PMC7841858 DOI: 10.1177/1756287220986640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/16/2020] [Indexed: 01/04/2023] Open
Abstract
Artificial intelligence (AI) involves technology that is able to emulate tasks previously carried out by humans. The growing incidence, novel diagnostic strategies and newer available therapeutic options have had resource and economic impacts on the healthcare organizations providing prostate cancer care. AI has the potential to be an adjunct to and, in certain cases, a replacement for human input in prostate cancer care delivery. Automation can also address issues such as inter- and intra-observer variability and has the ability to deliver analysis of large volume datasets quickly and accurately. The continuous training and testing of AI algorithms will facilitate development of futuristic AI models that will have integral roles to play in diagnostics, enhanced training and surgical outcomes and developments of prostate cancer predictive tools. These AI related innovations will enable clinicians to provide individualized care. Despite its potential benefits, it is vital that governance with AI related care is maintained and responsible adoption is achieved.
Collapse
Affiliation(s)
- B M Zeeshan Hameed
- Department of Urology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Milap Shah
- Department of Urology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Nithesh Naik
- Department of Mechanical and Manufacturing Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sufyan Ibrahim
- i-TRUE: International Training and Research in Uro-oncology and Endourology, Manipal, Karnataka, India
| | - Bhaskar Somani
- Department of Urology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Patrick Rice
- Department of Urology, University Hospital Southampton NHS Trust, Southampton, UK
| | - Naeem Soomro
- Department of Urology, Freeman Hospital, Newcastle, UK
| | - Bhavan Prasad Rai
- i-TRUE: International Training and Research in Uro-oncology and Endourology, Manipal, Karnataka, India
| |
Collapse
|
15
|
Barisoni L, Lafata KJ, Hewitt SM, Madabhushi A, Balis UGJ. Digital pathology and computational image analysis in nephropathology. Nat Rev Nephrol 2020; 16:669-685. [PMID: 32848206 PMCID: PMC7447970 DOI: 10.1038/s41581-020-0321-6] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2020] [Indexed: 12/17/2022]
Abstract
The emergence of digital pathology - an image-based environment for the acquisition, management and interpretation of pathology information supported by computational techniques for data extraction and analysis - is changing the pathology ecosystem. In particular, by virtue of our new-found ability to generate and curate digital libraries, the field of machine vision can now be effectively applied to histopathological subject matter by individuals who do not have deep expertise in machine vision techniques. Although these novel approaches have already advanced the detection, classification, and prognostication of diseases in the fields of radiology and oncology, renal pathology is just entering the digital era, with the establishment of consortia and digital pathology repositories for the collection, analysis and integration of pathology data with other domains. The development of machine-learning approaches for the extraction of information from image data, allows for tissue interrogation in a way that was not previously possible. The application of these novel tools are placing pathology centre stage in the process of defining new, integrated, biologically and clinically homogeneous disease categories, to identify patients at risk of progression, and shifting current paradigms for the treatment and prevention of kidney diseases.
Collapse
Affiliation(s)
- Laura Barisoni
- Department of Pathology, Duke University, Durham, NC, USA.
- Department of Medicine, Division of Nephrology, Duke University, Durham, NC, USA.
| | - Kyle J Lafata
- Department of Radiology, Duke University, Durham, NC, USA
- Department of Radiation Oncology, Duke University, Durham, NC, USA
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Louis Stokes Veterans Administration Medical Center, Cleveland, OH, USA
| | | |
Collapse
|
16
|
Computer Extracted Features from Initial H&E Tissue Biopsies Predict Disease Progression for Prostate Cancer Patients on Active Surveillance. Cancers (Basel) 2020; 12:cancers12092708. [PMID: 32967377 PMCID: PMC7563653 DOI: 10.3390/cancers12092708] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 01/21/2023] Open
Abstract
In this work, we assessed the ability of computerized features of nuclear morphology from diagnostic biopsy images to predict prostate cancer (CaP) progression in active surveillance (AS) patients. Improved risk characterization of AS patients could reduce over-testing of low-risk patients while directing high-risk patients to therapy. A total of 191 (125 progressors, 66 non-progressors) AS patients from a single site were identified using The Johns Hopkins University's (JHU) AS-eligibility criteria. Progression was determined by pathologists at JHU. 30 progressors and 30 non-progressors were randomly selected to create the training cohort D1 (n = 60). The remaining patients comprised the validation cohort D2 (n = 131). Digitized Hematoxylin & Eosin (H&E) biopsies were annotated by a pathologist for CaP regions. Nuclei within the cancer regions were segmented using a watershed method and 216 nuclear features describing position, shape, orientation, and clustering were extracted. Six features associated with disease progression were identified using D1 and then used to train a machine learning classifier. The classifier was validated on D2. The classifier was further compared on a subset of D2 (n = 47) against pro-PSA, an isoform of prostate specific antigen (PSA) more linked with CaP, in predicting progression. Performance was evaluated with area under the curve (AUC). A combination of nuclear spatial arrangement, shape, and disorder features were associated with progression. The classifier using these features yielded an AUC of 0.75 in D2. On the 47 patient subset with pro-PSA measurements, the classifier yielded an AUC of 0.79 compared to an AUC of 0.42 for pro-PSA. Nuclear morphometric features from digitized H&E biopsies predicted progression in AS patients. This may be useful for identifying AS-eligible patients who could benefit from immediate curative therapy. However, additional multi-site validation is needed.
Collapse
|
17
|
Abstract
Artificial intelligence (AI) - the ability of a machine to perform cognitive tasks to achieve a particular goal based on provided data - is revolutionizing and reshaping our health-care systems. The current availability of ever-increasing computational power, highly developed pattern recognition algorithms and advanced image processing software working at very high speeds has led to the emergence of computer-based systems that are trained to perform complex tasks in bioinformatics, medical imaging and medical robotics. Accessibility to 'big data' enables the 'cognitive' computer to scan billions of bits of unstructured information, extract the relevant information and recognize complex patterns with increasing confidence. Computer-based decision-support systems based on machine learning (ML) have the potential to revolutionize medicine by performing complex tasks that are currently assigned to specialists to improve diagnostic accuracy, increase efficiency of throughputs, improve clinical workflow, decrease human resource costs and improve treatment choices. These characteristics could be especially helpful in the management of prostate cancer, with growing applications in diagnostic imaging, surgical interventions, skills training and assessment, digital pathology and genomics. Medicine must adapt to this changing world, and urologists, oncologists, radiologists and pathologists, as high-volume users of imaging and pathology, need to understand this burgeoning science and acknowledge that the development of highly accurate AI-based decision-support applications of ML will require collaboration between data scientists, computer researchers and engineers.
Collapse
|
18
|
Shen L, Thompson PM. Brain Imaging Genomics: Integrated Analysis and Machine Learning. PROCEEDINGS OF THE IEEE. INSTITUTE OF ELECTRICAL AND ELECTRONICS ENGINEERS 2020; 108:125-162. [PMID: 31902950 PMCID: PMC6941751 DOI: 10.1109/jproc.2019.2947272] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Brain imaging genomics is an emerging data science field, where integrated analysis of brain imaging and genomics data, often combined with other biomarker, clinical and environmental data, is performed to gain new insights into the phenotypic, genetic and molecular characteristics of the brain as well as their impact on normal and disordered brain function and behavior. It has enormous potential to contribute significantly to biomedical discoveries in brain science. Given the increasingly important role of statistical and machine learning in biomedicine and rapidly growing literature in brain imaging genomics, we provide an up-to-date and comprehensive review of statistical and machine learning methods for brain imaging genomics, as well as a practical discussion on method selection for various biomedical applications.
Collapse
Affiliation(s)
- Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark & Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90232, USA
| |
Collapse
|
19
|
Ren J, Singer EA, Sadimin E, Foran DJ, Qi X. Statistical Analysis of Survival Models Using Feature Quantification on Prostate Cancer Histopathological Images. J Pathol Inform 2019; 10:30. [PMID: 31620309 PMCID: PMC6788183 DOI: 10.4103/jpi.jpi_85_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 06/14/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Grading of prostatic adenocarcinoma is based on the Gleason scoring system and the more recently established prognostic grade groups. Typically, prostate cancer grading is performed by pathologists based on the morphology of the tumor on hematoxylin and eosin (H and E) slides. In this study, we investigated the histopathological image features with various survival models and attempted to study their correlations. METHODS Three texture methods (speeded-up robust features, histogram of oriented gradient, and local binary pattern) and two convolutional neural network (CNN)-based methods were applied to quantify histopathological image features. Five survival models were assessed on those image features in the context with other prostate clinical prognostic factors, including primary and secondary Gleason patterns, prostate-specific antigen levels, age, and clinical tumor stages. RESULTS Based on statistical comparisons among different image features with survival models, image features from CNN-based method with a recurrent neural network called CNN-long-short-term memory provided the highest hazard ratio of prostate cancer recurrence under Cox regression with an elastic net penalty. CONCLUSIONS This approach outperformed the other image quantification methods listed above. Using this approach, patient outcomes were highly correlated with the histopathological image features of the tissue samples. In future studies, we plan to investigate the potential use of this approach for predicting recurrence in a wider range of cancer types.
Collapse
Affiliation(s)
- Jian Ren
- Department of Electrical and Computer Engineering, Rutgers University, Piscataway, NJ, USA
| | - Eric A. Singer
- Department of Pathology and Laboratory Medicine, Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
- Center for Biomedical Imaging and Informatics, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Evita Sadimin
- Department of Pathology and Laboratory Medicine, Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - David J. Foran
- Center for Biomedical Imaging and Informatics, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Xin Qi
- Center for Biomedical Imaging and Informatics, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
20
|
Janowczyk A, Zuo R, Gilmore H, Feldman M, Madabhushi A. HistoQC: An Open-Source Quality Control Tool for Digital Pathology Slides. JCO Clin Cancer Inform 2019; 3:1-7. [PMID: 30990737 PMCID: PMC6552675 DOI: 10.1200/cci.18.00157] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2019] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Digital pathology (DP), referring to the digitization of tissue slides, is beginning to change the landscape of clinical diagnostic workflows and has engendered active research within the area of computational pathology. One of the challenges in DP is the presence of artefacts and batch effects, unintentionally introduced during both routine slide preparation (eg, staining, tissue folding) and digitization (eg, blurriness, variations in contrast and hue). Manual review of glass and digital slides is laborious, qualitative, and subject to intra- and inter-reader variability. Therefore, there is a critical need for a reproducible automated approach of precisely localizing artefacts to identify slides that need to be reproduced or regions that should be avoided during computational analysis. METHODS Here we present HistoQC, a tool for rapidly performing quality control to not only identify and delineate artefacts but also discover cohort-level outliers (eg, slides stained darker or lighter than others in the cohort). This open-source tool employs a combination of image metrics (eg, color histograms, brightness, contrast), features (eg, edge detectors), and supervised classifiers (eg, pen detection) to identify artefact-free regions on digitized slides. These regions and metrics are presented to the user via an interactive graphical user interface, facilitating artefact detection through real-time visualization and filtering. These same metrics afford users the opportunity to explicitly define acceptable tolerances for their workflows. RESULTS The output of HistoQC on 450 slides from The Cancer Genome Atlas was reviewed by two pathologists and found to be suitable for computational analysis more than 95% of the time. CONCLUSION These results suggest that HistoQC could provide an automated, quantifiable, quality control process for identifying artefacts and measuring slide quality, in turn helping to improve both the repeatability and robustness of DP workflows.
Collapse
Affiliation(s)
| | - Ren Zuo
- Case Western Reserve University, Cleveland, OH
| | - Hannah Gilmore
- University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Michael Feldman
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Anant Madabhushi
- Case Western Reserve University, Cleveland, OH
- Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH
| |
Collapse
|
21
|
Advanced Morphologic Analysis for Diagnosing Allograft Rejection: The Case of Cardiac Transplant Rejection. Transplantation 2019; 102:1230-1239. [PMID: 29570167 PMCID: PMC6059998 DOI: 10.1097/tp.0000000000002189] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Allograft rejection remains a significant concern after all solid organ transplants. Although qualitative morphologic analysis with histologic grading of biopsy samples is the main tool employed for diagnosing allograft rejection, this standard has significant limitations in precision and accuracy that affect patient care. The use of endomyocardial biopsy to diagnose cardiac allograft rejection illustrates the significant shortcomings of current approaches for diagnosing allograft rejection. Despite disappointing interobserver variability, concerns about discordance with clinical trajectories, attempts at revising the histologic criteria and efforts to establish new diagnostic tools with imaging and gene expression profiling, no method has yet supplanted endomyocardial biopsy as the diagnostic gold standard. In this context, automated approaches to complex data analysis problems—often referred to as “machine learning”—represent promising strategies to improve overall diagnostic accuracy. By focusing on cardiac allograft rejection, where tissue sampling is relatively frequent, this review highlights the limitations of the current approach to diagnosing allograft rejection, introduces the basic methodology behind machine learning and automated image feature detection, and highlights the initial successes of these approaches within cardiovascular medicine. The authors review “machine learning“ and “automated image feature detection“ to improve the diagnostic accuracy of endomyocardial biopsies to diagnose cardiac allograft rejection.
Collapse
|
22
|
Real-time feature selection technique with concept drift detection using adaptive micro-clusters for data stream mining. Knowl Based Syst 2018. [DOI: 10.1016/j.knosys.2018.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
23
|
Leo P, Elliott R, Shih NNC, Gupta S, Feldman M, Madabhushi A. Stable and discriminating features are predictive of cancer presence and Gleason grade in radical prostatectomy specimens: a multi-site study. Sci Rep 2018; 8:14918. [PMID: 30297720 PMCID: PMC6175913 DOI: 10.1038/s41598-018-33026-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 09/20/2018] [Indexed: 01/17/2023] Open
Abstract
Site variation in fixation, staining, and scanning can confound automated tissue based image classifiers for disease characterization. In this study we incorporated stability into four feature selection methods for identifying the most robust and discriminating features for two prostate histopathology classification tasks. We evaluated 242 morphology features from N = 212 prostatectomy specimens from four sites for automated cancer detection and grading. We quantified instability as the rate of significant cross-site feature differences. We mapped feature stability and discriminability using 188 non-cancerous and 210 cancerous regions via 3-fold cross validation, then held one site out, creating independent training and testing sets. In training, one feature set was selected only for discriminability, another for discriminability and stability. We trained a classifier with each feature set, testing on the hold out site. Experiments were repeated with 117 Gleason grade 3 and 112 grade 4 regions. Stability was calculated across non-cancerous regions. Gland shape features yielded the best stability and area under the receiver operating curve (AUC) trade-off while co-occurrence texture features were generally unstable. Our stability-informed method produced a cancer detection AUC of 0.98 ± 0.05 and increased average Gleason grading AUC by 4.38%. Color normalization of the images tended to exacerbate feature instability.
Collapse
Affiliation(s)
- Patrick Leo
- Case Western Reserve University, Dept. of Biomedical Engineering, Cleveland, OH, 44106, United States
| | - Robin Elliott
- Case Western Reserve University, Dept. of Pathology, Cleveland, OH, 44106, United States
| | - Natalie N C Shih
- University of Pennsylvania, Dept. of Pathology, Philadelphia, PA, 19104, United States
| | - Sanjay Gupta
- Case Western Reserve University, Dept. of Urology, Cleveland, OH, 44106, United States
| | - Michael Feldman
- University of Pennsylvania, Dept. of Pathology, Philadelphia, PA, 19104, United States
| | - Anant Madabhushi
- Case Western Reserve University, Dept. of Biomedical Engineering, Cleveland, OH, 44106, United States.
| |
Collapse
|
24
|
Ren J, Karagoz K, Gatza ML, Singer EA, Sadimin E, Foran DJ, Qi X. Recurrence analysis on prostate cancer patients with Gleason score 7 using integrated histopathology whole-slide images and genomic data through deep neural networks. J Med Imaging (Bellingham) 2018; 5:047501. [PMID: 30840742 PMCID: PMC6237203 DOI: 10.1117/1.jmi.5.4.047501] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 10/23/2018] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer is the most common nonskin-related cancer, affecting one in seven men in the United States. Gleason score, a sum of the primary and secondary Gleason patterns, is one of the best predictors of prostate cancer outcomes. Recently, significant progress has been made in molecular subtyping prostate cancer through the use of genomic sequencing. It has been established that prostate cancer patients presented with a Gleason score 7 show heterogeneity in both disease recurrence and survival. We built a unified system using publicly available whole-slide images and genomic data of histopathology specimens through deep neural networks to identify a set of computational biomarkers. Using a survival model, the experimental results on the public prostate dataset showed that the computational biomarkers extracted by our approach had hazard ratio as 5.73 and C -index as 0.74, which were higher than standard clinical prognostic factors and other engineered image texture features. Collectively, the results of this study highlight the important role of neural network analysis of prostate cancer and the potential of such approaches in other precision medicine applications.
Collapse
Affiliation(s)
- Jian Ren
- Rutgers, the State University of New Jersey, Department of Electrical and Computer Engineering, Piscataway, New Jersey, United States
| | - Kubra Karagoz
- Rutgers Cancer Institute of New Jersey, Department of Radiation Oncology, New Brunswick, New Jersey, United States
| | - Michael L. Gatza
- Rutgers Cancer Institute of New Jersey, Department of Radiation Oncology, New Brunswick, New Jersey, United States
| | - Eric A. Singer
- Rutgers Cancer Institute of New Jersey, Section of Urologic Oncology, New Brunswick, New Jersey, United States
| | - Evita Sadimin
- Rutgers Cancer Institute of New Jersey, Department of Pathology and Laboratory Medicine, New Brunswick, New Jersey, United States
| | - David J. Foran
- Rutgers Cancer Institute of New Jersey, Department of Pathology and Laboratory Medicine, New Brunswick, New Jersey, United States
| | - Xin Qi
- Rutgers Cancer Institute of New Jersey, Department of Pathology and Laboratory Medicine, New Brunswick, New Jersey, United States
| |
Collapse
|
25
|
Penzias G, Singanamalli A, Elliott R, Gollamudi J, Shih N, Feldman M, Stricker PD, Delprado W, Tiwari S, Böhm M, Haynes AM, Ponsky L, Fu P, Tiwari P, Viswanath S, Madabhushi A. Identifying the morphologic basis for radiomic features in distinguishing different Gleason grades of prostate cancer on MRI: Preliminary findings. PLoS One 2018; 13:e0200730. [PMID: 30169514 PMCID: PMC6118356 DOI: 10.1371/journal.pone.0200730] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 07/02/2018] [Indexed: 12/29/2022] Open
Abstract
Translation of radiomics into the clinic may require a more comprehensive understanding of the underlying morphologic tissue characteristics they reflect. In the context of prostate cancer (PCa), some studies have correlated gross histological measurements of gland lumen, epithelium, and nuclei with disease appearance on MRI. Quantitative histomorphometry (QH), like radiomics for radiologic images, is the computer based extraction of features for describing tumor morphology on digitized tissue images. In this work, we attempt to establish the histomorphometric basis for radiomic features for prostate cancer by (1) identifying the radiomic features from T2w MRI most discriminating of low vs. intermediate/high Gleason score, (2) identifying QH features correlated with the most discriminating radiomic features previously identified, and (3) evaluating the discriminative ability of QH features found to be correlated with spatially co-localized radiomic features. On a cohort of 36 patients (23 for training, 13 for validation), Gabor texture features were identified as being most predictive of Gleason grade on MRI (AUC of 0.69) and gland lumen shape features were identified as the most predictive QH features (AUC = 0.75). Our results suggest that the PCa grade discriminability of Gabor features is a consequence of variations in gland shape and morphology at the tissue level.
Collapse
Affiliation(s)
- Gregory Penzias
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Asha Singanamalli
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Robin Elliott
- University Hospitals, Cleveland, OH, United States of America
| | - Jay Gollamudi
- University Hospitals, Cleveland, OH, United States of America
| | - Natalie Shih
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Michael Feldman
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, United States of America
| | | | - Warick Delprado
- Douglass Hanly Moir Pathology, Macquarie Park, NSW, Australia
| | - Sarita Tiwari
- Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
| | - Maret Böhm
- Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
| | - Anne-Maree Haynes
- Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
| | - Lee Ponsky
- University Hospitals, Cleveland, OH, United States of America
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States of America
| | - Pallavi Tiwari
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Satish Viswanath
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| |
Collapse
|
26
|
Charmsaz S, Prencipe M, Kiely M, Pidgeon GP, Collins DM. Innovative Technologies Changing Cancer Treatment. Cancers (Basel) 2018; 10:cancers10060208. [PMID: 29921753 PMCID: PMC6025540 DOI: 10.3390/cancers10060208] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 12/28/2022] Open
Abstract
Conventional therapies for cancer such as chemotherapy and radiotherapy remain a mainstay in treatment, but in many cases a targeted approach is lacking, and patients can be vulnerable to drug resistance. In recent years, novel concepts have been emerging to improve the traditional therapeutic options in cancers with poor survival outcomes. New therapeutic strategies involving areas like energy metabolism and extracellular vesicles along with advances in immunotherapy and nanotechnology are driving the next generation of cancer treatments. The development of fields such as theranostics in nanomedicine is also opening new doors for targeted drug delivery and nano-imaging. Here we discuss the use of innovative technologies presented at the Irish Association for Cancer Research (IACR) Annual Meeting, highlighting examples of where new approaches may lead to promising new treatment options for a range of cancer types.
Collapse
Affiliation(s)
- Sara Charmsaz
- RCSI Surgery, Royal College of Surgeons in Ireland, 31A York Street, Dublin 2, Ireland.
| | - Maria Prencipe
- School of Biomolecular and Biomedical Research, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Maeve Kiely
- Graduate Entry Medical School, University of Limerick, Limerick, Ireland.
| | - Graham P Pidgeon
- Trinity Translational Medicine Institute (TTMI), St. James's Hospital and Trinity College Dublin, Dublin 2, Ireland.
| | - Denis M Collins
- Cancer Biotherapeutics, National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
27
|
Digital pathology in nephrology clinical trials, research, and pathology practice. Curr Opin Nephrol Hypertens 2018; 26:450-459. [PMID: 28858910 DOI: 10.1097/mnh.0000000000000360] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW In this review, we will discuss (i) how the recent advancements in digital technology and computational engineering are currently applied to nephropathology in the setting of clinical research, trials, and practice; (ii) the benefits of the new digital environment; (iii) how recognizing its challenges provides opportunities for transformation; and (iv) nephropathology in the upcoming era of kidney precision and predictive medicine. RECENT FINDINGS Recent studies highlighted how new standardized protocols facilitate the harmonization of digital pathology database infrastructure and morphologic, morphometric, and computer-aided quantitative analyses. Digital pathology enables robust protocols for clinical trials and research, with the potential to identify previously underused or unrecognized clinically useful parameters. The integration of digital pathology with molecular signatures is leading the way to establishing clinically relevant morpho-omic taxonomies of renal diseases. SUMMARY The introduction of digital pathology in clinical research and trials, and the progressive implementation of the modern software ecosystem, opens opportunities for the development of new predictive diagnostic paradigms and computer-aided algorithms, transforming the practice of renal disease into a modern computational science.
Collapse
|
28
|
Mandal A, Maji P. FaRoC: Fast and Robust Supervised Canonical Correlation Analysis for Multimodal Omics Data. IEEE TRANSACTIONS ON CYBERNETICS 2018; 48:1229-1241. [PMID: 28391216 DOI: 10.1109/tcyb.2017.2685625] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
One of the main problems associated with high dimensional multimodal real life data sets is how to extract relevant and significant features. In this regard, a fast and robust feature extraction algorithm, termed as FaRoC, is proposed, integrating judiciously the merits of canonical correlation analysis (CCA) and rough sets. The proposed method extracts new features sequentially from two multidimensional data sets by maximizing their relevance with respect to class label and significance with respect to already-extracted features. To generate canonical variables sequentially, an analytical formulation is introduced to establish the relation between regularization parameters and CCA. The formulation enables the proposed method to extract required number of correlated features sequentially with lesser computational cost as compared to existing methods. To compute both significance and relevance measures of a feature, the concept of hypercuboid equivalence partition matrix of rough hypercuboid approach is used. It also provides an efficient way to find optimum regularization parameters employed in CCA. The efficacy of the proposed FaRoC algorithm, along with a comparison with other existing methods, is extensively established on several real life data sets.
Collapse
|
29
|
Martel AL, Hosseinzadeh D, Senaras C, Zhou Y, Yazdanpanah A, Shojaii R, Patterson ES, Madabhushi A, Gurcan MN. An Image Analysis Resource for Cancer Research: PIIP-Pathology Image Informatics Platform for Visualization, Analysis, and Management. Cancer Res 2017; 77:e83-e86. [PMID: 29092947 DOI: 10.1158/0008-5472.can-17-0323] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/28/2017] [Accepted: 08/09/2017] [Indexed: 01/18/2023]
Abstract
Pathology Image Informatics Platform (PIIP) is an NCI/NIH sponsored project intended for managing, annotating, sharing, and quantitatively analyzing digital pathology imaging data. It expands on an existing, freely available pathology image viewer, Sedeen. The goal of this project is to develop and embed some commonly used image analysis applications into the Sedeen viewer to create a freely available resource for the digital pathology and cancer research communities. Thus far, new plugins have been developed and incorporated into the platform for out of focus detection, region of interest transformation, and IHC slide analysis. Our biomarker quantification and nuclear segmentation algorithms, written in MATLAB, have also been integrated into the viewer. This article describes the viewing software and the mechanism to extend functionality by plugins, brief descriptions of which are provided as examples, to guide users who want to use this platform. PIIP project materials, including a video describing its usage and applications, and links for the Sedeen Viewer, plug-ins, and user manuals are freely available through the project web page: http://pathiip.org Cancer Res; 77(21); e83-86. ©2017 AACR.
Collapse
Affiliation(s)
- Anne L Martel
- Sunnybrook Research Institute, Toronto, Canada. .,Medical Biophysics, University of Toronto, Toronto, Canada
| | | | | | - Yu Zhou
- Case Western Reserve University, Cleveland, Ohio
| | | | | | | | | | | |
Collapse
|
30
|
Bhargava R, Madabhushi A. Emerging Themes in Image Informatics and Molecular Analysis for Digital Pathology. Annu Rev Biomed Eng 2017; 18:387-412. [PMID: 27420575 DOI: 10.1146/annurev-bioeng-112415-114722] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pathology is essential for research in disease and development, as well as for clinical decision making. For more than 100 years, pathology practice has involved analyzing images of stained, thin tissue sections by a trained human using an optical microscope. Technological advances are now driving major changes in this paradigm toward digital pathology (DP). The digital transformation of pathology goes beyond recording, archiving, and retrieving images, providing new computational tools to inform better decision making for precision medicine. First, we discuss some emerging innovations in both computational image analytics and imaging instrumentation in DP. Second, we discuss molecular contrast in pathology. Molecular DP has traditionally been an extension of pathology with molecularly specific dyes. Label-free, spectroscopic images are rapidly emerging as another important information source, and we describe the benefits and potential of this evolution. Third, we describe multimodal DP, which is enabled by computational algorithms and combines the best characteristics of structural and molecular pathology. Finally, we provide examples of application areas in telepathology, education, and precision medicine. We conclude by discussing challenges and emerging opportunities in this area.
Collapse
Affiliation(s)
- Rohit Bhargava
- Departments of Bioengineering, Chemical and Biomolecular Engineering, Electrical and Computer Engineering, Mechanical Science and Engineering, and Chemistry, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801;
| | - Anant Madabhushi
- Center for Computational Imaging and Personalized Diagnostics; Departments of Biomedical Engineering, Urology, Pathology, Radiology, Radiation Oncology, General Medical Sciences, Electrical Engineering, and Computer Science; and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106;
| |
Collapse
|
31
|
Luo G, Dong S, Wang K, Zuo W, Cao S, Zhang H. Multi-Views Fusion CNN for Left Ventricular Volumes Estimation on Cardiac MR Images. IEEE Trans Biomed Eng 2017; 65:1924-1934. [PMID: 29035205 DOI: 10.1109/tbme.2017.2762762] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Left ventricular (LV) volume estimation is a critical procedure for cardiac disease diagnosis. The objective of this paper is to address a direct LV volume prediction task. METHODS In this paper, we propose a direct volume prediction method based on the end-to-end deep convolutional neural networks. We study the end-to-end LV volume prediction method in items of the data preprocessing, network structure, and multiview fusion strategy. The main contributions of this paper are the following aspects. First, we propose a new data preprocessing method on cardiac magnetic resonance (CMR). Second, we propose a new network structure for end-to-end LV volume estimation. Third, we explore the representational capacity of different slices and propose a fusion strategy to improve the prediction accuracy. RESULTS The evaluation results show that the proposed method outperforms other state-of-the-art LV volume estimation methods on the open accessible benchmark datasets. The clinical indexes derived from the predicted volumes agree well with the ground truth ( ${\rm{EDV:R}}^{{\rm 2}}={\text{0.974}}$, ${\rm{RMSE\,}}= {\text{9.6}}{\rm{\,ml}}$; ${\rm{ESV:R}}^{{\rm 2}}={\text{0.976}}$, ${\rm{RMSE}}= {\text{7.1}}\,{\text{ml}}$; ${\rm{EF:R}}^{{\rm 2}} ={\text{0.828}}$, ${\rm{RMSE}}= {\text{4.71}}\% $). CONCLUSION Experimental results prove that the proposed method may be useful for the LV volume prediction task. SIGNIFICANCE The proposed method not only has application potential for cardiac diseases screening for large-scale CMR data, but also can be extended to other medical image research fields.
Collapse
|
32
|
Discriminative Scale Learning (DiScrn): Applications to Prostate Cancer Detection from MRI and Needle Biopsies. Sci Rep 2017; 7:12375. [PMID: 28959011 PMCID: PMC5620056 DOI: 10.1038/s41598-017-12569-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 08/22/2017] [Indexed: 01/03/2023] Open
Abstract
There has been recent substantial interest in extracting sub-visual features from medical images for improved disease characterization compared to what might be achievable via visual inspection alone. Features such as Haralick and Gabor can provide a multi-scale representation of the original image by extracting measurements across differently sized neighborhoods. While these multi-scale features are effective, on large-scale digital pathological images, the process of extracting these features is computationally expensive. Moreover for different problems, different scales and neighborhood sizes may be more or less important and thus a large number of features extracted might end up being redundant. In this paper, we present a Discriminative Scale learning (DiScrn) approach that attempts to automatically identify the distinctive scales at which features are able to best separate cancerous from non-cancerous regions on both radiologic and digital pathology tissue images. To evaluate the efficacy of our approach, our approach was employed to detect presence and extent of prostate cancer on a total of 60 MRI and digitized histopathology images. Compared to a multi-scale feature analysis approach invoking features across all scales, DiScrn achieved 66% computational efficiency while also achieving comparable or even better classifier performance.
Collapse
|
33
|
Singanamalli A, Wang H, Madabhushi A. Cascaded Multi-view Canonical Correlation (CaMCCo) for Early Diagnosis of Alzheimer's Disease via Fusion of Clinical, Imaging and Omic Features. Sci Rep 2017; 7:8137. [PMID: 28811553 PMCID: PMC5558022 DOI: 10.1038/s41598-017-03925-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 04/24/2017] [Indexed: 12/14/2022] Open
Abstract
The introduction of mild cognitive impairment (MCI) as a diagnostic category adds to the challenges of diagnosing Alzheimer’s Disease (AD). No single marker has been proven to accurately categorize patients into their respective diagnostic groups. Thus, previous studies have attempted to develop fused predictors of AD and MCI. These studies have two main limitations. Most do not simultaneously consider all diagnostic categories and provide suboptimal fused representations using the same set of modalities for prediction of all classes. In this work, we present a combined framework, cascaded multiview canonical correlation (CaMCCo), for fusion and cascaded classification that incorporates all diagnostic categories and optimizes classification by selectively combining a subset of modalities at each level of the cascade. CaMCCo is evaluated on a data cohort comprising 149 patients for whom neurophysiological, neuroimaging, proteomic and genomic data were available. Results suggest that fusion of select modalities for each classification task outperforms (mean AUC = 0.92) fusion of all modalities (mean AUC = 0.54) and individual modalities (mean AUC = 0.90, 0.53, 0.71, 0.73, 0.62, 0.68). In addition, CaMCCo outperforms all other multi-class classification methods for MCI prediction (PPV: 0.80 vs. 0.67, 0.63).
Collapse
Affiliation(s)
- Asha Singanamalli
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Haibo Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | | |
Collapse
|
34
|
Li W, Bai X, Hu E, Huang H, Li Y, He Y, Lv J, Chen L, He W. Identification of breast cancer prognostic modules based on weighted protein-protein interaction networks. Oncol Lett 2017; 13:3935-3941. [PMID: 28529601 DOI: 10.3892/ol.2017.5917] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 01/31/2017] [Indexed: 01/30/2023] Open
Abstract
Breast cancer is one of the leading causes of mortality in females. A number of prognostic markers have been identified, including single genes, multi-gene signatures and network modules; however, the robustness of these prognostic markers is insufficient. Thus, the present study proposed a more robust method to identify breast cancer prognostic modules based on weighted protein-protein interaction networks, by integrating four sets of disease-associated expression profiles. Three identified prognostic modules were closely associated with prognosis-associated functions and survival time, as determined by Cox regression and Kaplan-Meier survival analyses. The robustness of these modules was verified with an independent profile from another platform. Genes from these modules may be useful as breast cancer prognostic markers. The prognostic modules could be used to determine the prognoses of patients with breast cancer and characterize patient recovery.
Collapse
Affiliation(s)
- Wan Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Xue Bai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Erqiang Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Hao Huang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yiran Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yuehan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Junjie Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Lina Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Weiming He
- Institute of Opto-electronics, Harbin Institute of Technology, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
35
|
Yang G, Hu Z. Gene Feature Extraction Based on Nonnegative Dual Graph Regularized Latent Low-Rank Representation. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1096028. [PMID: 28466003 PMCID: PMC5390636 DOI: 10.1155/2017/1096028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 03/13/2017] [Indexed: 01/16/2023]
Abstract
Aiming at the problem of gene expression profile's high redundancy and heavy noise, a new feature extraction model based on nonnegative dual graph regularized latent low-rank representation (NNDGLLRR) is presented on the basis of latent low-rank representation (Lat-LRR). By introducing dual graph manifold regularized constraint, the NNDGLLRR can keep the internal spatial structure of the original data effectively and improve the final clustering accuracy while segmenting the subspace. The introduction of nonnegative constraints makes the computation with some sparsity, which enhances the robustness of the algorithm. Different from Lat-LRR, a new solution model is adopted to simplify the computational complexity. The experimental results show that the proposed algorithm has good feature extraction performance for the heavy redundancy and noise gene expression profile, which, compared with LRR and Lat-LRR, can achieve better clustering accuracy.
Collapse
Affiliation(s)
- Guoliang Yang
- School of Electrical Engineering and Automation, Jiangxi University of Science and Technology, Ganzhou 341000, China
| | - Zhengwei Hu
- School of Electrical Engineering and Automation, Jiangxi University of Science and Technology, Ganzhou 341000, China
| |
Collapse
|
36
|
|
37
|
Viswanath SE, Tiwari P, Lee G, Madabhushi A. Dimensionality reduction-based fusion approaches for imaging and non-imaging biomedical data: concepts, workflow, and use-cases. BMC Med Imaging 2017; 17:2. [PMID: 28056889 PMCID: PMC5217665 DOI: 10.1186/s12880-016-0172-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 12/09/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND With a wide array of multi-modal, multi-protocol, and multi-scale biomedical data being routinely acquired for disease characterization, there is a pressing need for quantitative tools to combine these varied channels of information. The goal of these integrated predictors is to combine these varied sources of information, while improving on the predictive ability of any individual modality. A number of application-specific data fusion methods have been previously proposed in the literature which have attempted to reconcile the differences in dimensionalities and length scales across different modalities. Our objective in this paper was to help identify metholodological choices that need to be made in order to build a data fusion technique, as it is not always clear which strategy is optimal for a particular problem. As a comprehensive review of all possible data fusion methods was outside the scope of this paper, we have focused on fusion approaches that employ dimensionality reduction (DR). METHODS In this work, we quantitatively evaluate 4 non-overlapping existing instantiations of DR-based data fusion, within 3 different biomedical applications comprising over 100 studies. These instantiations utilized different knowledge representation and knowledge fusion methods, allowing us to examine the interplay of these modules in the context of data fusion. The use cases considered in this work involve the integration of (a) radiomics features from T2w MRI with peak area features from MR spectroscopy for identification of prostate cancer in vivo, (b) histomorphometric features (quantitative features extracted from histopathology) with protein mass spectrometry features for predicting 5 year biochemical recurrence in prostate cancer patients, and (c) volumetric measurements on T1w MRI with protein expression features to discriminate between patients with and without Alzheimers' Disease. RESULTS AND CONCLUSIONS Our preliminary results in these specific use cases indicated that the use of kernel representations in conjunction with DR-based fusion may be most effective, as a weighted multi-kernel-based DR approach resulted in the highest area under the ROC curve of over 0.8. By contrast non-optimized DR-based representation and fusion methods yielded the worst predictive performance across all 3 applications. Our results suggest that when the individual modalities demonstrate relatively poor discriminability, many of the data fusion methods may not yield accurate, discriminatory representations either. In summary, to outperform the predictive ability of individual modalities, methodological choices for data fusion must explicitly account for the sparsity of and noise in the feature space.
Collapse
Affiliation(s)
- Satish E Viswanath
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Wickenden 523, Cleveland, OH, USA.
| | - Pallavi Tiwari
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Wickenden 523, Cleveland, OH, USA
| | - George Lee
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Wickenden 523, Cleveland, OH, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Wickenden 523, Cleveland, OH, USA
| | | |
Collapse
|
38
|
Maji P, Mandal A. Multimodal Omics Data Integration Using Max Relevance--Max Significance Criterion. IEEE Trans Biomed Eng 2016; 64:1841-1851. [PMID: 27834637 DOI: 10.1109/tbme.2016.2624823] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE This paper presents a novel supervised regularized canonical correlation analysis, termed as CuRSaR, to extract relevant and significant features from multimodal high dimensional omics datasets. METHODS The proposed method extracts a new set of features from two multidimensional datasets by maximizing the relevance of extracted features with respect to sample categories and significance among them. It integrates judiciously the merits of regularized canonical correlation analysis (RCCA) and rough hypercuboid approach. An analytical formulation, based on spectral decomposition, is introduced to establish the relation between canonical correlation analysis (CCA) and RCCA. The concept of hypercuboid equivalence partition matrix of rough hypercuboid is used to compute both relevance and significance of a feature. SIGNIFICANCE The analytical formulation makes the computational complexity of the proposed algorithm significantly lower than existing methods. The equivalence partition matrix offers an efficient way to find optimum regularization parameters employed in CCA. RESULTS The superiority of the proposed algorithm over other existing methods, in terms of computational complexity and classification accuracy, is established extensively on real life data.
Collapse
|
39
|
Leo P, Lee G, Shih NNC, Elliott R, Feldman MD, Madabhushi A. Evaluating stability of histomorphometric features across scanner and staining variations: prostate cancer diagnosis from whole slide images. J Med Imaging (Bellingham) 2016; 3:047502. [PMID: 27803941 DOI: 10.1117/1.jmi.3.4.047502] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 09/16/2016] [Indexed: 01/04/2023] Open
Abstract
Quantitative histomorphometry (QH) is the process of computerized feature extraction from digitized tissue slide images to predict disease presence, behavior, and outcome. Feature stability between sites may be compromised by laboratory-specific variables including dye batch, slice thickness, and the whole slide scanner used. We present two new measures, preparation-induced instability score and latent instability score, to quantify feature instability across and within datasets. In a use case involving prostate cancer, we examined QH features which may detect cancer on whole slide images. Using our method, we found that five feature families (graph, shape, co-occurring gland tensor, sub-graph, and texture) were different between datasets in 19.7% to 48.6% of comparisons while the values expected without site variation were 4.2% to 4.6%. Color normalizing all images to a template did not reduce instability. Scanning the same 34 slides on three scanners demonstrated that Haralick features were most substantively affected by scanner variation, being unstable in 62% of comparisons. We found that unstable feature families performed significantly worse in inter- than intrasite classification. Our results appear to suggest QH features should be evaluated across sites to assess robustness, and class discriminability alone should not represent the benchmark for digital pathology feature selection.
Collapse
Affiliation(s)
- Patrick Leo
- Case Western Reserve University , Department of Biomedical Engineering, 2071 Martin Luther King Jr. Drive, Cleveland, Ohio 44106, United States
| | - George Lee
- Case Western Reserve University , Department of Biomedical Engineering, 2071 Martin Luther King Jr. Drive, Cleveland, Ohio 44106, United States
| | - Natalie N C Shih
- University of Pennsylvania , Department of Pathology, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, United States
| | - Robin Elliott
- Case Western Reserve University , Department of Pathology, 11100 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Michael D Feldman
- University of Pennsylvania , Department of Pathology, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, United States
| | - Anant Madabhushi
- Case Western Reserve University , Department of Biomedical Engineering, 2071 Martin Luther King Jr. Drive, Cleveland, Ohio 44106, United States
| |
Collapse
|
40
|
Lu C, Xu H, Xu J, Gilmore H, Mandal M, Madabhushi A. Multi-Pass Adaptive Voting for Nuclei Detection in Histopathological Images. Sci Rep 2016; 6:33985. [PMID: 27694950 PMCID: PMC5046183 DOI: 10.1038/srep33985] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 09/02/2016] [Indexed: 12/15/2022] Open
Abstract
Nuclei detection is often a critical initial step in the development of computer aided diagnosis and prognosis schemes in the context of digital pathology images. While over the last few years, a number of nuclei detection methods have been proposed, most of these approaches make idealistic assumptions about the staining quality of the tissue. In this paper, we present a new Multi-Pass Adaptive Voting (MPAV) for nuclei detection which is specifically geared towards images with poor quality staining and noise on account of tissue preparation artifacts. The MPAV utilizes the symmetric property of nuclear boundary and adaptively selects gradient from edge fragments to perform voting for a potential nucleus location. The MPAV was evaluated in three cohorts with different staining methods: Hematoxylin &Eosin, CD31 &Hematoxylin, and Ki-67 and where most of the nuclei were unevenly and imprecisely stained. Across a total of 47 images and nearly 17,700 manually labeled nuclei serving as the ground truth, MPAV was able to achieve a superior performance, with an area under the precision-recall curve (AUC) of 0.73. Additionally, MPAV also outperformed three state-of-the-art nuclei detection methods, a single pass voting method, a multi-pass voting method, and a deep learning based method.
Collapse
Affiliation(s)
- Cheng Lu
- College of Computer Science, Shaanxi Normal University, Xi’an, Shaanxi Province, 710119, China
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106-7207, USA
| | - Hongming Xu
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Alberta, T6G 2V4, Canada
| | - Jun Xu
- Jiangsu Key Laboratory of Big Data Analysis Technique, Nanjing University of Information Science and Technology, Nanjing, 210044, China
| | - Hannah Gilmore
- Department of Pathology-Anatomic, University Hospitals Case Medial Center, Case Western Reserve University, Cleveland, OH, 44106-7207, USA
| | - Mrinal Mandal
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Alberta, T6G 2V4, Canada
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106-7207, USA
| |
Collapse
|
41
|
Adeli E, Lalush DS. Multi-Level Canonical Correlation Analysis for Standard-Dose PET Image Estimation. IEEE TRANSACTIONS ON IMAGE PROCESSING : A PUBLICATION OF THE IEEE SIGNAL PROCESSING SOCIETY 2016; 25:3303-3315. [PMID: 27187957 PMCID: PMC5106345 DOI: 10.1109/tip.2016.2567072] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Positron emission tomography (PET) images are widely used in many clinical applications, such as tumor detection and brain disorder diagnosis. To obtain PET images of diagnostic quality, a sufficient amount of radioactive tracer has to be injected into a living body, which will inevitably increase the risk of radiation exposure. On the other hand, if the tracer dose is considerably reduced, the quality of the resulting images would be significantly degraded. It is of great interest to estimate a standard-dose PET (S-PET) image from a low-dose one in order to reduce the risk of radiation exposure and preserve image quality. This may be achieved through mapping both S-PET and low-dose PET data into a common space and then performing patch-based sparse representation. However, a one-size-fits-all common space built from all training patches is unlikely to be optimal for each target S-PET patch, which limits the estimation accuracy. In this paper, we propose a data-driven multi-level canonical correlation analysis scheme to solve this problem. In particular, a subset of training data that is most useful in estimating a target S-PET patch is identified in each level, and then used in the next level to update common space and improve estimation. In addition, we also use multi-modal magnetic resonance images to help improve the estimation with complementary information. Validations on phantom and real human brain data sets show that our method effectively estimates S-PET images and well preserves critical clinical quantification measures, such as standard uptake value.
Collapse
|
42
|
Lee G, Veltri RW, Zhu G, Ali S, Epstein JI, Madabhushi A. Nuclear Shape and Architecture in Benign Fields Predict Biochemical Recurrence in Prostate Cancer Patients Following Radical Prostatectomy: Preliminary Findings. Eur Urol Focus 2016; 3:457-466. [PMID: 28753763 DOI: 10.1016/j.euf.2016.05.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/21/2016] [Accepted: 05/26/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Gleason scoring represents the standard for diagnosis of prostate cancer (PCa) and assessment of prognosis following radical prostatectomy (RP), but it does not account for patterns in neighboring normal-appearing benign fields that may be predictive of disease recurrence. OBJECTIVE To investigate (1) whether computer-extracted image features within tumor-adjacent benign regions on digital pathology images could predict recurrence in PCa patients after surgery and (2) whether a tumor plus adjacent benign signature (TABS) could better predict recurrence compared with Gleason score or features from benign or cancerous regions alone. DESIGN, SETTING, AND PARTICIPANTS We studied 140 tissue microarray cores (0.6mm each) from 70 PCa patients following surgery between 2000 and 2004 with up to 14 yr of follow-up. Overall, 22 patients experienced recurrence (biochemical [prostate-specific antigen], local, or distant recurrence and cancer death) and 48 did not. INTERVENTION RP was performed in all patients. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The top 10 features identified as most predictive of recurrence within both the benign and cancerous regions were combined into a 10-feature signature (TABS). Computer-extracted nuclear shape and architectural features from cancerous regions, adjacent benign fields, and TABS were evaluated via random forest classification accuracy and Kaplan-Meier survival analysis. RESULTS AND LIMITATIONS Tumor-adjacent benign field features were predictive of recurrence (area under the receiver operating characteristic curve [AUC]: 0.72). Tumor-field nuclear shape descriptors and benign-field local nuclear arrangement were the predominant features found for TABS (AUC: 0.77). Combining TABS with Gleason sum further improved identification of recurrence (AUC: 0.81). All experiments were performed using threefold cross-validation without independent test set validation. CONCLUSIONS Computer-extracted nuclear features within cancerous and benign regions predict recurrence following RP. Furthermore, TABS was shown to provide added value to common predictors including Gleason sum and Kattan and Stephenson nomograms. PATIENT SUMMARY Future studies may benefit from evaluation of benign regions proximal to the tumor on surgically excised prostate cancer tissue for assessing risk of disease recurrence.
Collapse
Affiliation(s)
- George Lee
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| | - Robert W Veltri
- Department of Urology, James Buchanan Brady Urological Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guangjing Zhu
- Department of Urology, James Buchanan Brady Urological Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sahirzeeshan Ali
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Jonathan I Epstein
- Department of Urology, James Buchanan Brady Urological Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
43
|
Thomas S, Hao L, Ricke WA, Li L. Biomarker discovery in mass spectrometry-based urinary proteomics. Proteomics Clin Appl 2016; 10:358-70. [PMID: 26703953 DOI: 10.1002/prca.201500102] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 12/05/2015] [Accepted: 12/21/2015] [Indexed: 01/03/2023]
Abstract
Urinary proteomics has become one of the most attractive topics in disease biomarker discovery. MS-based proteomic analysis has advanced continuously and emerged as a prominent tool in the field of clinical bioanalysis. However, only few protein biomarkers have made their way to validation and clinical practice. Biomarker discovery is challenged by many clinical and analytical factors including, but not limited to, the complexity of urine and the wide dynamic range of endogenous proteins in the sample. This article highlights promising technologies and strategies in the MS-based biomarker discovery process, including study design, sample preparation, protein quantification, instrumental platforms, and bioinformatics. Different proteomics approaches are discussed, and progresses in maximizing urinary proteome coverage and standardization are emphasized in this review. MS-based urinary proteomics has great potential in the development of noninvasive diagnostic assays in the future, which will require collaborative efforts between analytical scientists, systems biologists, and clinicians.
Collapse
Affiliation(s)
- Samuel Thomas
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Ling Hao
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - William A Ricke
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA.,Department of Urology, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA.,Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
44
|
Ginsburg SB, Lee G, Ali S, Madabhushi A. Feature Importance in Nonlinear Embeddings (FINE): Applications in Digital Pathology. IEEE TRANSACTIONS ON MEDICAL IMAGING 2016; 35:76-88. [PMID: 26186772 DOI: 10.1109/tmi.2015.2456188] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Quantitative histomorphometry (QH) refers to the process of computationally modeling disease appearance on digital pathology images by extracting hundreds of image features and using them to predict disease presence or outcome. Since constructing a robust and interpretable classifier is challenging in a high dimensional feature space, dimensionality reduction (DR) is often implemented prior to classifier construction. However, when DR is performed it can be challenging to quantify the contribution of each of the original features to the final classification result. We have previously presented a method for scoring features based on their importance for classification on an embedding derived via principal components analysis (PCA). However, nonlinear DR involves the eigen-decomposition of a kernel matrix rather than the data itself, compounding the issue of classifier interpretability. In this paper we present feature importance in nonlinear embeddings (FINE), an extension of our PCA-based feature scoring method to kernel PCA (KPCA), as well as several NLDR algorithms that can be cast as variants of KPCA. FINE is applied to four digital pathology datasets to identify key QH features for predicting the risk of breast and prostate cancer recurrence. Measures of nuclear and glandular architecture and clusteredness were found to play an important role in predicting the likelihood of recurrence of both breast and prostate cancers. Compared to the t-test, Fisher score, and Gini index, FINE was able to identify a stable set of features that provide good classification accuracy on four publicly available datasets from the NIPS 2003 Feature Selection Challenge.
Collapse
|
45
|
Trifiletti DM, Showalter TN. Big Data and Comparative Effectiveness Research in Radiation Oncology: Synergy and Accelerated Discovery. Front Oncol 2015; 5:274. [PMID: 26697409 PMCID: PMC4672039 DOI: 10.3389/fonc.2015.00274] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/23/2015] [Indexed: 12/18/2022] Open
Abstract
Several advances in large data set collection and processing have the potential to provide a wave of new insights and improvements in the use of radiation therapy for cancer treatment. The era of electronic health records, genomics, and improving information technology resources creates the opportunity to leverage these developments to create a learning healthcare system that can rapidly deliver informative clinical evidence. By merging concepts from comparative effectiveness research with the tools and analytic approaches of “big data,” it is hoped that this union will accelerate discovery, improve evidence for decision making, and increase the availability of highly relevant, personalized information. This combination offers the potential to provide data and analysis that can be leveraged for ultra-personalized medicine and high-quality, cutting-edge radiation therapy.
Collapse
Affiliation(s)
- Daniel M Trifiletti
- Department of Radiation Oncology, University of Virginia School of Medicine , Charlottesville, VA , USA
| | - Timothy N Showalter
- Department of Radiation Oncology, University of Virginia School of Medicine , Charlottesville, VA , USA
| |
Collapse
|
46
|
Guo Y, Wu G, Yap PT, Jewells V, Lin W, Shen D. Segmentation of Infant Hippocampus Using Common Feature Representations Learned for Multimodal Longitudinal Data. MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION : MICCAI ... INTERNATIONAL CONFERENCE ON MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION 2015; 9351:63-71. [PMID: 27019875 DOI: 10.1007/978-3-319-24574-4_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Aberrant development of the human brain during the first year after birth is known to cause critical implications in later stages of life. In particular, neuropsychiatric disorders, such as attention deficit hyperactivity disorder (ADHD), have been linked with abnormal early development of the hippocampus. Despite its known importance, studying the hippocampus in infant subjects is very challenging due to the significantly smaller brain size, dynamically varying image contrast, and large across-subject variation. In this paper, we present a novel method for effective hippocampus segmentation by using a multi-atlas approach that integrates the complementary multimodal information from longitudinal T1 and T2 MR images. In particular, considering the highly heterogeneous nature of the longitudinal data, we propose to learn their common feature representations by using hierarchical multi-set kernel canonical correlation analysis (CCA). Specifically, we will learn (1) within-time-point common features by projecting different modality features of each time point to its own modality-free common space, and (2) across-time-point common features by mapping all time-point-specific common features to a global common space for all time points. These final features are then employed in patch matching across different modalities and time points for hippocampus segmentation, via label propagation and fusion. Experimental results demonstrate the improved performance of our method over the state-of-the-art methods.
Collapse
Affiliation(s)
- Yanrong Guo
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, NC, USA
| | - Guorong Wu
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, NC, USA
| | - Pew-Thian Yap
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, NC, USA
| | - Valerie Jewells
- Department of Radiology, University of North Carolina at Chapel Hill, NC, USA
| | - Weili Lin
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, NC, USA
| | - Dinggang Shen
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, NC, USA
| |
Collapse
|