scholarly journals Artificial intelligence in digital pathology — new tools for diagnosis and precision oncology

2019 ◽  
Vol 16 (11) ◽  
pp. 703-715 ◽  
Author(s):  
Kaustav Bera ◽  
Kurt A. Schalper ◽  
David L. Rimm ◽  
Vamsidhar Velcheti ◽  
Anant Madabhushi
2021 ◽  
Author(s):  
Changjiang Zhou ◽  
Xiaobing Feng ◽  
Yi Jin ◽  
Harvest F. Gu ◽  
Youcai Zhao ◽  
...  

Abstract BackgroundThe possibility of digitizing whole-slide images (WSI) of tissue has led to the advent of artificial intelligence (AI) in digital pathology. Advances in precision oncology have resulted in an increasing demand for predictive assays that enable mining of subvisual morphometric phenotypes and might improve patient care ultimately. Hence, a pathologist-annotated and artificial intelligence-empowered platform for integration and analysis of WSI data and molecular detection data in tumors was established, called PAI-WSIT (http://www.paiwsit.com).MethodsThe standardized data collection process was used for data collection in PAI-WSIT, while a multifunctional annotation tool was developed and a user-friendly search engine and web interface were integrated for the database access. Furthermore, deep learning frameworks were applied in two tasks to detect malignant regions and classify phenotypic subtypes in colorectal cancers (CRCs), respectively.ResultsPAI-WSIT recorded 8633 WSIs of 1772 tumor cases, of which CRC from four regional hospitals in China and The Cancer Genome Atlas (TCGA) were the main ones, as well as cancers in breast, lung, prostate, bladder, and kidneys from two Chinese hospitals. A total of 1298 WSIs with high-quality annotations were evaluated by a panel of 8 pathologists. Gene detection reports of 582 tumor cases were collected. Clinical information of all tumor cases was documented. Besides, we reached overall accuracy of 0.933 in WSI classification for malignant region detection of CRC, and aera under the curves (AUC) of 0.719 on colorectal subtype dataset.ConclusionsCollectively, the annotation function, data integration and AI function analysis of PAI-WSIT provide support for AI-assisted tumor diagnosis, all of which have provided a comprehensive curation of carcinomas pathology.


Healthcare ◽  
2020 ◽  
Vol 9 (1) ◽  
pp. 30
Author(s):  
Daniele Giansanti

Thanks to the incredible changes promoted by Information and Communication Technology (ICT) conveyed today by electronic-health (eHealth) and mobile-health (mHealth), many new applications of both organ and cellular diagnostics are now possible [...]


2021 ◽  
Author(s):  
Changjiang Zhou ◽  
Xiaobing Feng ◽  
Hongbin Cai ◽  
Yi Jin ◽  
Harvest F. Gu ◽  
...  

2021 ◽  
pp. jclinpath-2020-207351
Author(s):  
Jenny Fitzgerald ◽  
Debra Higgins ◽  
Claudia Mazo Vargas ◽  
William Watson ◽  
Catherine Mooney ◽  
...  

Clinical workflows in oncology depend on predictive and prognostic biomarkers. However, the growing number of complex biomarkers contributes to costly and delayed decision-making in routine oncology care and treatment. As cancer is expected to rank as the leading cause of death and the single most important barrier to increasing life expectancy in the 21st century, there is a major emphasis on precision medicine, particularly individualisation of treatment through better prediction of patient outcome. Over the past few years, both surgical and pathology specialties have suffered cutbacks and a low uptake of pathology specialists means a solution is required to enable high-throughput screening and personalised treatment in this area to alleviate bottlenecks. Digital imaging in pathology has undergone an exponential period of growth. Deep-learning (DL) platforms for hematoxylin and eosin (H&E) image analysis, with preliminary artificial intelligence (AI)-based grading capabilities of specimens, can evaluate image characteristics which may not be visually apparent to a pathologist and offer new possibilities for better modelling of disease appearance and possibly improve the prediction of disease stage and patient outcome. Although digital pathology and AI are still emerging areas, they are the critical components for advancing personalised medicine. Integration of transcriptomic analysis, clinical information and AI-based image analysis is yet an uncultivated field by which healthcare professionals can make improved treatment decisions in cancer. This short review describes the potential application of integrative AI in offering better detection, quantification, classification, prognosis and prediction of breast and prostate cancer and also highlights the utilisation of machine learning systems in biomarker evaluation.


2020 ◽  
pp. 367-382 ◽  
Author(s):  
Stephanie A. Harmon ◽  
Thomas H. Sanford ◽  
G. Thomas Brown ◽  
Chris Yang ◽  
Sherif Mehralivand ◽  
...  

PURPOSE To develop an artificial intelligence (AI)–based model for identifying patients with lymph node (LN) metastasis based on digital evaluation of primary tumors and train the model using cystectomy specimens available from The Cancer Genome Atlas (TCGA) Project; patients from our institution were included for validation of the leave-out test cohort. METHODS In all, 307 patients were identified for inclusion in the study (TCGA, n = 294; in-house, n = 13). Deep learning models were trained from image patches at 2.5×, 5×, 10×, and 20× magnifications, and spatially resolved prediction maps were combined with microenvironment (lymphocyte infiltration) features to derive a final patient-level AI score (probability of LN metastasis). Training and validation included 219 patients (training, n = 146; validation, n = 73); 89 patients (TCGA, n = 75; in-house, n = 13) were reserved as an independent testing set. Multivariable logistic regression models for predicting LN status based on clinicopathologic features alone and a combined model with AI score were fit to training and validation sets. RESULTS Several patients were determined to have positive LN metastasis in TCGA (n = 105; 35.7%) and in-house (n = 3; 23.1%) cohorts. A clinicopathologic model that considered using factors such as age, T stage, and lymphovascular invasion demonstrated an area under the curve (AUC) of 0.755 (95% CI, 0.680 to 0.831) in the training and validation cohorts compared with the cross validation of the AI score (likelihood of positive LNs), which achieved an AUC of 0.866 (95% CI, 0.812 to 0.920; P = .021). Performance in the test cohort was similar, with a clinicopathologic model AUC of 0.678 (95% CI, 0.554 to 0.802) and an AI score of 0.784 (95% CI, 0.702 to 0.896; P = .21). In addition, the AI score remained significant after adjusting for clinicopathologic variables ( P = 1.08 × 10−9), and the combined model significantly outperformed clinicopathologic features alone in the test cohort with an AUC of 0.807 (95% CI, 0.702 to 0.912; P = .047). CONCLUSION Patients who are at higher risk of having positive LNs during cystectomy can be identified on primary tumor samples using novel AI-based methodologies applied to digital hematoxylin and eosin–stained slides.


2020 ◽  
pp. jclinpath-2020-206715
Author(s):  
Nikolaos Stathonikos ◽  
Tri Q Nguyen ◽  
Paul J van Diest

Since 2007, we have gradually been building up infrastructure for digital pathology, starting with a whole slide scanner park to build up a digital archive to streamline doing multidisciplinary meetings, student teaching and research, culminating in a full digital diagnostic workflow where we are currently integrating artificial intelligence algorithms. In this paper, we highlight the different steps in this process towards digital diagnostics, which was at times a rocky road with definitely issues in implementation, but eventually an exciting new way to practice pathology in a more modern and efficient way where patient safety has clearly gone up.


2019 ◽  
Vol 20 (5) ◽  
pp. e253-e261 ◽  
Author(s):  
Muhammad Khalid Khan Niazi ◽  
Anil V Parwani ◽  
Metin N Gurcan

2019 ◽  
Vol 37 (4_suppl) ◽  
pp. TPS717-TPS717
Author(s):  
Selin Kurnaz ◽  
Arturo Loaiza-Bonilla ◽  
Jason Lawrence Freedman ◽  
Belisario Augusto Arango ◽  
Kristin Johnston ◽  
...  

TPS717 Background: Precision oncology encompasses the implementation of high level of evidence disease-specific and biomarker-driven diagnostic and treatment recommendations for optimized cancer care. Artificial Intelligence (AI), telemedicine and value-based care may optimize clinical trial enrollment (CTE) and overall cost-benefit. This ongoing, international registry for cancer pts evaluates the feasibility and clinical utility of an AI-based precision oncology clinical trial matching tool, powered by a virtual tumor boards (VTB) program, and its clinical impact on pts with advanced cancer to facilitate CTE, as well as the financial impact, and potential outcomes of the intervention. Methods: The SYNERGY-AI Registry is an international prospective, observational cohort study of eligible adult and pediatric pts with advanced solid and hematological malignancies, for whom the decision to consider CTE has already been made by their primary providers (PP). Using a proprietary application programming interface (API) linked to existing electronic health records (EHR) platforms, individual clinical data is extracted, analyzed and matched to a parametric database of existing institutional and non-institutional CTs. Machine learning algorithms allow for dynamic matching based on CT allocation and availability for optimized matching. Patients voluntarily enroll into registry, which is non-interventional with no protocol-mandated tests/procedures—all treatment decisions are made at the discretion of PP in consultation with their pts, based on the AI CT matching report, and VTB support. CTE will be assessed on variables including biomarkers, barriers to enrollment. Study duration anticipated as ~36 mo (~24-mo enrollment followed by 12 mo of data collection, to occur every 3 mo). The primary analysis will be performed 12 mo after last pt enrolled. The impact time to initiation of CTE on PFS and OS will be estimated by Kaplan-Meier and Cox multivariable survival analysis. Enrollment is ongoing, with a target of ≥ 1500 patients. Key inclusion criteria: Pts with solid and hematological malignancies; cancer-related biomarkers. Key exclusion criteria: ECOG PS > 2; abnormal organ function; hospice enrollment Clinical trial information: NCT03452774.


Sign in / Sign up

Export Citation Format

Share Document