Artificial intelligence in digital pathology - new tools for diagnosis and precision oncology

Kaustav Bera, Kurt A Schalper, David L Rimm, Vamsidhar Velcheti, Anant Madabhushi, Kaustav Bera, Kurt A Schalper, David L Rimm, Vamsidhar Velcheti, Anant Madabhushi

Abstract

In the past decade, advances in precision oncology have resulted in an increased demand for predictive assays that enable the selection and stratification of patients for treatment. The enormous divergence of signalling and transcriptional networks mediating the crosstalk between cancer, stromal and immune cells complicates the development of functionally relevant biomarkers based on a single gene or protein. However, the result of these complex processes can be uniquely captured in the morphometric features of stained tissue specimens. The possibility of digitizing whole-slide images of tissue has led to the advent of artificial intelligence (AI) and machine learning tools in digital pathology, which enable mining of subvisual morphometric phenotypes and might, ultimately, improve patient management. In this Perspective, we critically evaluate various AI-based computational approaches for digital pathology, focusing on deep neural networks and 'hand-crafted' feature-based methodologies. We aim to provide a broad framework for incorporating AI and machine learning tools into clinical oncology, with an emphasis on biomarker development. We discuss some of the challenges relating to the use of AI, including the need for well-curated validation datasets, regulatory approval and fair reimbursement strategies. Finally, we present potential future opportunities for precision oncology.

Figures

Fig. 1 |. Milestones in computational pathology.
Fig. 1 |. Milestones in computational pathology.
Over the past two decades, technological advances have enabled efficient digitization of whole-slide images, subsequently helping to streamline pathology workflows across pathology labs worldwide. Slide digitization has enabled the creation of large-scale digital-slide libraries, the most popular of which is probably The Cancer Genome Atlas, which has enabled researchers around the world to freely access a richly curated and annotated dataset of pathology images linked with clinical, outcome and genomic information, in turn spurring substantial research activity into artificial intelligence for digital pathology and oncology.
Fig. 2 |. Workflow and general framework…
Fig. 2 |. Workflow and general framework for artificial intelligence (Ai) approaches in digital pathology.
Typical steps involved in the use of two popular categories of AI approaches: deep learning and hand-crafted feature engineering.
Fig. 3 |. Visual representations of hand-crafted…
Fig. 3 |. Visual representations of hand-crafted features across cancer types.
a | Spatial arrangement of clusters of tissue-infiltrating lymphocytes in a non-small-cell lung carcinoma (NSCLC) whole-slide image. b | Features developed using quantitative immunofluorescence of tissue-infiltrating lymphocyte subpopulations (including detection of CD4+ and CD8+ T cells and CD20+ B cells) in NSCLC samples. c | Features reflecting the distribution and entropy of global cell cluster graphs constructed using NSCLC specimens. d | Features computing the relative orientation of the glands present in prostate cancer tissue. e | Diversity of texture of cancer cell nuclei in an oral cavity squamous cell carcinoma. f | Nuclear shape feature computed on cancer cell nuclei in a human papillomavirus-positive oropharyngeal carcinoma. g | Graph feature showing the spatial relationships of different cancer cell nuclei in an oral cavity carcinoma. h | Hand-crafted feature capturing cellular heterogeneity in an oestrogen receptor-positive breast cancer.
Fig. 4 |. Artificial intelligence (Al) and…
Fig. 4 |. Artificial intelligence (Al) and machine learning approaches complement the expertise and support the pathologist and oncologist.
Some of the existing AI approaches currently used by pathologists to analyse images from tumours are depicted. For the practicing oncologist, AI approaches can be used to aid decision making for different aspects of the management of patients with cancer.

References

    1. Prewitt JMS Intelligent microscopes: recent and near-future advances. Proc. SPIE 10.1117/12.958214 (1979).
    1. Prewitt JMS Parametric and nonparametric recognition by computer: an application to leukocyte image processing. Adv. Comput. 12, 285–414 (1972).
    1. Prewitt JMS & Mendelsohn ML The analysis of cell images. Ann. NY Acad. Sci. 128, 1035–1053 (1966).
    1. McCarthy J, Minsky ML, Rochester N & Shannon CE A proposal for the Dartmouth summer research project on artificial intelligence, August 31, 1955 AI Mag. 27, 12 (2006).
    1. McCarthy JJ, Minsky ML & Rochester N Artificial intelligence. Research Laboratory of Electronics (RLE) at the Massachusetts Institute of Technology (MIT) (1959).
    1. Yao X Evolving artificial neural networks. Proc. IEEE 87 1423–1447 (1999).
    1. Haykin S Neural Networks (Prentice Hall, 1994).
    1. Deng L Deep learning: methods and applications. Found. Trends® Signal Process. 7, 197–387 (2014).
    1. LeCun Y, Bengio Y & Hinton G Deep learning. Nature 521, 436–444 (2015).
    1. Hinton GE & Salakhutdinov RR Reducing the dimensionality of data with neural networks. Science 313, 504–507 (2006).
    1. Krizhevsky A, Sutskever I & Hinton GE ImageNet classification with deep convolutional neural networks. (2012).
    1. LeCun Y, Huang FJ & Bottou L in Proc. 2004 IEEE Comput. Soc; Conf. Comput. Vis. Pattern Recognit. II-104 (IEEE, 2004).
    1. LeCun Y & Bengio Y in The handbook of brain theory and neural networks (ed. Arbib MA) 255–258 (MIT Press, 1998).
    1. LeCun Y, Bottou L, Bengio Y & Haffner P Gradient-based learning applied to document recognition. Proc. IEEE 86, 2278–2324 (1998).
    1. Deng J, et al. in 2009 IEEE Conf. Comput. Vis. Pattern Recognit. 248–255 (IEEE, 2009).
    1. Mukhopadhyay S et al. Whole slide imaging versus microscopy for primary diagnosis in surgical pathology: a multicenter blinded randomized noninferiority study of 1992 cases (Pivotal Study). Am. J. Surg. Pathol. 42, 39–52 (2018).
    1. Kujan O et al. Why oral histopathology suffers inter-observer variability on grading oral epithelial dysplasia: an attempt to understand the sources of variation. Oral Oncol. 43, 224–231 (2007).
    1. Chi AC, Katabi N, Chen H-S & Cheng Y-SL Interobserver variation among pathologists in evaluating perineural invasion for oral squamous cell carcinoma. Head Neck Pathol. 10, 451–464 (2016).
    1. Evans AJ et al. Interobserver variability between expert urologic pathologists for extraprostatic extension and surgical margin status in radical prostatectomy specimens. Am. J. Surg. Pathol. 32, 1503–1512 (2008).
    1. Shanes JG et al. Interobserver variability in the pathologic interpretation of endomyocardial biopsy results. Circulation 75, 401–405 (1987).
    1. Elmore JG et al. Diagnostic concordance among pathologists interpreting breast biopsy specimens. JAMA 313, 1122–1132 (2015).
    1. Brimo F, Schultz L & Epstein JI The value of mandatory second opinion pathology review of prostate needle biopsy interpretation before radical prostatectomy. J. Urol. 184, 126–130 (2010).
    1. Kilgore ML & Goldman DP Drug costs and out-of-pocket spending in cancer clinical trials. Contemp. Clin. Trials 29, 1–8 (2008).
    1. Agarwal A, Ressler D & Snyder G The current and future state of companion diagnostics. Pharmacogenomics Pers. Med. 8, 99–110 (2015).
    1. Michor F & Polyak K The origins and implications of intratumor heterogeneity. Cancer Prev. Res. 3, 1361–1364 (2010).
    1. Cyll K et al. Tumour heterogeneity poses a significant challenge to cancer biomarker research. Br. J. Cancer 117, 367–375 (2017).
    1. Bejnordi BE et al. Diagnostic assessment of deep learning algorithms for detection of lymph node metastases in women with breast cancer. JAMA 318, 2199–2210 (2017).
    1. Nagpal K et al. Development and validation of a deep learning algorithm for improving Gleason scoring of prostate cancer. npj Digital Med. 2, 48 (2019).
    1. Bychkov D et al. Deep learning based tissue analysis predicts outcome in colorectal cancer. Sci. Rep. 8, 3395 (2018).
    1. Wang D, Khosla A, Gargeya R, Irshad H & Beck AH Deep learning for identifying metastatic breast cancer. (2016).
    1. Steiner D et al. Impact of deep learning assistance on the histopathologic review of lymph nodes for metastatic breast cancer. Am. J. Surg. Pathol. 42, 1636–1646 (2018).
    1. Chen J & Srinivas C Automatic lymphocyte detection in H&E images with deep neural networks. (2016).
    1. Garcia E, et al. in 2017 IEEE 30th Int. Symp. Comput.-Based Med. Sys. (CBMS). 200–204 (IEEE, 2017).
    1. Lu C et al. Multi-pass adaptive voting for nuclei detection in histopathological images. Sci. Rep. 6, 33985 (2016).
    1. Sornapudi S et al. Deep learning nuclei detection in digitized histology images by superpixels. J. Pathol. Inform. 10.4103/jpi.jpi_74_17 (2018).
    1. Wang H et al. Mitosis detection in breast cancer pathology images by combining handcrafted and convolutional neural network features. J. Med. Imaging (Bellingham) 1, 034003 (2014).
    1. Al-Kofahi Y, Lassoued W, Lee W & Roysam B Improved automatic detection and segmentation of cell nuclei in histopathology images. IEEE Trans. Biomed. Eng. 57, 841–852 (2010).
    1. Naik S, et al. in 2008 5th IEEE International Symposium on Biomedical Imaging: From Nano to Macro. 284–287 (IEEE, 2008).
    1. Nguyen K, Jain AK & Allen RL in 2010 20th Int. Conf. Pattern Recognit. 1497–1500 (IEEE, 2010).
    1. Kothari S, et al. in 2011 IEEE International Symposium on Biomedical Imaging: From Nano to Macro 657–660 (2011, IEEE; ).
    1. Sirinukunwattana K et al. Gland segmentation in colon histology images: the glas challenge contest. Med. Image Anal. 35, 489–502 (2017).
    1. Lee G et al. Co-occurring gland angularity in localized subgraphs: predicting biochemical recurrence in intermediate-risk prostate cancer patients. PLOS ONE 9, e97954 (2014).
    1. Lee G et al. Nuclear shape and architecture in benign fields predict biochemical recurrence in prostate cancer patients following radical prostatectomy: preliminary findings. Eur. Urol. Focus 3, 457–466 (2017).
    1. Lu C et al. Nuclear shape and orientation features from H&E images predict survival in early-stage estrogen receptor-positive breast cancers. Lab. Investig. J. Tech. Methods Pathol. 98, 1438–1448 2018
    1. Corredor G et al. Spatial architecture and arrangement of tumor-infiltrating lymphocytes for predicting likelihood of recurrence in early-stage non-small cell lung cancer. Clin. Cancer Res. 25, 1526–1534 (2018).
    1. Mungle T et al. MRF-ANN: a machine learning approach for automated ER scoring of breast cancer immunohistochemical images. J. Microsc. 267, 117–129 (2017).
    1. Wang X et al. Prediction of recurrence in early stage non-small cell lung cancer using computer extracted nuclear features from digital H&E images. Sci. Rep. 7, 13543 (2017).
    1. Rosado B et al. Accuracy of computer diagnosis of melanoma: a quantitative meta-analysis. Arch. Dermatol. 139, 361–367 (2003).
    1. Rosenbaum BE et al. Computer-assisted measurement of primary tumor area is prognostic of recurrence-free survival in stage IB melanoma patients. Mod. Pathol. 30, 1402–1410 (2017).
    1. Teramoto A, Tsukamoto T, Kiriyama Y & Fujita H Automated classification of lung cancer types from cytological images using deep convolutional neural networks. Biomed. Res. Int. 10.1155/2017/4067832 (2017).
    1. Wu M, Yan C, Liu H & Liu Q Automatic classification of ovarian cancer types from cytological images using deep convolutional neural networks. Biosci. Rep. 10.1042/BSR20180289 (2018).
    1. Ali S, Basavanhally A, Ganesan S & Madabhushi A Histogram of Hosoya indices for assessing similarity across subgraph populations: breast cancer prognosis prediction from digital pathology [abstract 118]. Lab. Invest. (supplement) 95, 32A (2015).
    1. Yu K-H et al. Predicting non-small cell lung cancer prognosis by fully automated microscopic pathology image features. Nat. Commun. 7, 12474 (2016).
    1. Cruz-Roa A et al. Accurate and reproducible invasive breast cancer detection in whole-slide images: a deep learning approach for quantifying tumor extent. Sci. Rep. 7, 46450 (2017).
    1. Liu Y et al. Artificial intelligence-based breast cancer nodal metastasis detection. Arch. Pathol. Lab. Med. 143, 859–868 (2018).
    1. Litjens G et al. 1399 H&E-stained sentinel lymph node sections of breast cancer patients: the CAMELYON dataset. GigaScience 7, giy065 (2018).
    1. Liu Y et al. Detecting cancer metastases on gigapixel pathology images. (2017).
    1. Madabhushi A & Lee G Image analysis and machine learning in digital pathology: challenges and opportunities. Med. Image Anal. 33, 170–175 (2016).
    1. Jansen I et al. Histopathology: ditch the slides, because digital and 3D are on show. World J. Urol. 36, 549–555 (2018).
    1. Ali S, Lewis J & Madabhushi A Spatially aware cell cluster(spACC1) graphs: predicting outcome in oropharyngeal pl6+ tumors. Med. Image Comput. Comput. Assist. Interv. 16, 412–419 (2013).
    1. Mobadersany P et al. Predicting cancer outcomes from histology and genomics using convolutional networks. Proc. Natl Acad. Sci. USA 115, E2970–E2979 (2018).
    1. Wang X et al. Computer extracted features of cancer nuclei from H&E stained tissues of tumor predicts response to nivolumab in non-small cell lung cancer. J. Clin. Oncol. 36(15_suppl), 12061–12061 (2018).
    1. Barrera C et al. Computer-extracted features relating to spatial arrangement of tumor infiltrating lymphocytes to predict response to nivolumab in non-small cell lung cancer (NSCLC). J. Clin. Oncol. 36, 12115–12115 (2018).
    1. Wang X, et al. Computerized nuclear morphometric features from H&E slide images are prognostic of recurrence and predictive of added benefit of adjuvant chemotherapy in early stage non-small cell lung cancer. Presented at the United States and Canadian Academy of Pathology’s 108th Annual Meeting. (2019).
    1. Gisselsson D et al. Abnormal nuclear shape in solid tumors reflects mitotic instability. Am. J. Pathol. 158, 199–206 (2001).
    1. Malon CD & Cosatto E Classification of mitotic figures with convolutional neural networks and seeded blob features. J. Pathol. Inform. 4, 9 (2013).
    1. Whitney J et al. Quantitative nuclear histomorphometry predicts oncotype DX risk categories for early stage ER+ breast cancer. BMC Cancer 18, 610 (2018).
    1. Simon I, Pound CR, Partin AW, Clemens JQ & Christens-Barry WA Automated image analysis system for detecting boundaries of live prostate cancer cells. Cytom. J. Int. Soc. Anal. Cytol. 31, 287–294 (1998).
    1. Basavanhally A et al. Multi-field-of-view framework for distinguishing tumor grade in ER+ breast cancer from entire histopathology slides. IEEE Trans. Biomed. Eng. 60, 2089–2099 (2013).
    1. Lewis JS, Ali S, Luo J, Thorstad WL & Madabhushi A A quantitative histomorphometric classifier (QuHbIC) identifies aggressive versus indolent p16-positive oropharyngeal squamous cell carcinoma. Am. J. Surg. Pathol. 38, 128–137 (2014).
    1. Barker J, Hoogi A, Depeursinge A & Rubin DL Automated classification of brain tumor type in whole-slide digital pathology images using local representative tiles. Med. Image Anal. 30, 60–71 (2016).
    1. Kong J et al. Machine-based morphologic analysis of glioblastoma using whole-slide pathology images uncovers clinically relevant molecular correlates. PLOS ONE 8, e81049 (2013).
    1. Osareh A & Shadgar B in 2010 5th Int. Symp. Health Informat. Bioinformat. 114–120 (2010, IEEE; ).
    1. Veta M et al. Prognostic value of automatically extracted nuclear morphometric features in whole slide images of male breast cancer. Mod. Pathol. 25, 1559 (2012).
    1. Lu C et al. An oral cavity squamous cell carcinoma quantitative histomorphometric-based image classifier of nuclear morphology can risk stratify patients for disease-specific survival. Mod. Pathol. 30, 1655–1665 (2017).
    1. Saltz J et al. Spatial organization and molecular correlation of tumor-infiltrating lymphocytes using deep learning on pathology images. Cell Rep. 23, 181–193.e7 (2018).
    1. Yuan Y Modelling the spatial heterogeneity and molecular correlates of lymphocytic infiltration in triple-negative breast cancer. J. R. Soc. Interface 10.1098/rsif.2014.1153 (2015).
    1. Heindl A, et al. Relevance of spatial heterogeneity of immune infiltration for predicting risk of recurrence after endocrine therapy of ER+ breast cancer. J. Natl Cancer. Inst. 10.1093/jnci/djx137 (2018).
    1. Beck AH et al. Systematic analysis of breast cancer morphology uncovers stromal features associated with survival. Sci. Transl Med. 3, 108ra113 (2011).
    1. Bhargava HK et al. Computer-extracted stromal features of African-Americans versus Caucasians from H&E slides and impact on prognosis of biochemical recurrence. J. Clin. Oncol. 36(15_suppl), 12075–12075 (2018).
    1. Vamathevan J et al. Applications of machine learning in drug discovery and development. Nat. Rev. Drug Discov 18, 463–477 (2019).
    1. Janowczyk A & Madabhushi A Deep learning for digital pathology image analysis: A comprehensive tutorial with selected use cases. J. Pathol. Inform. 7, 29 (2016).
    1. Araujo T et al. Classification of breast cancer histology images using Convolutional Neural Networks. PLOS ONE 12, e0177544 (2017).
    1. Ehteshami Bejnordi B et al. Using deep convolutional neural networks to identify and classify tumor-associated stroma in diagnostic breast biopsies. Mod. Pathol. 31, 1502–1512 (2018).
    1. Haenssle HA et al. Man against machine: diagnostic performance of a deep learning convolutional neural network for dermoscopic melanoma recognition in comparison to 58 dermatologists. Ann. Oncol. 29, 1836–1842 (2018).
    1. Sheikhzadeh F, Guillaud M & Ward RK Automatic labeling of molecular biomarkers of whole slide immunohistochemistry images using fully convolutional networks. PLOS ONE 13, e0190783 (2018).
    1. Basavanhally AN et al. Computerized image-based detection and grading of lymphocytic infiltration in HER2+ breast cancer histopathology. IEEE Trans. Biomed. Eng. 57, 642–653 (2010).
    1. Alzubaidi L et al. A robust deep learning approach to detect nuclei in histopathological images. Int. J. Innov. Res. Comp. Commun. Eng. 5, 4461–4467 (2007).
    1. Hofener H et al. Deep learning nuclei detection: a simple approach can deliver state-of-the-art results. Comput. Med. Imaging Graph. 70, 43–52 (2018).
    1. Xu J, Luo X, Wang G, Gilmore H & Madabhushi A A deep convolutional neural network for segmenting and classifying epithelial and stromal regions in histopathological images. Neurocomputing 191, 214–223 (2016).
    1. Senaras C, Niazi MKK, Lozanski G & Gurcan MN DeepFocus: detection of out-of-focus regions in whole slide digital images using deep learning. PLOS ONE 13, e0205387 (2018).
    1. Esteva A et al. Dermatologist-level classification of skin cancer with deep neural networks. Nature 542, 115–118 (2017).
    1. Tschandl P et al. Comparison of the accuracy of human readers versus machine-learning algorithms for pigmented skin lesion classification: an open, web-based, international, diagnostic study. Lancet Oncol. 20, 938–947 (2019).
    1. Couture HD et al. Image analysis with deep learning to predict breast cancer grade, ER status, histologic subtype, and intrinsic subtype. npj Breast Cancer 4, 30 (2018).
    1. Romo-Bucheli D, Janowczyk A, Gilmore H, Romero E & Madabhushi A A deep learning based strategy for identifying and associating mitotic activity with gene expression derived risk categories in estrogen receptor positive breast cancers. Cytomtery A 91, 566–573 (2017).
    1. Geessink OGF, et al. Computer aided quantification of intratumoral stroma yields an independent prognosticator in rectal cancer. Cell Oncol. 42, 331–341 (2019).
    1. Kather JN et al. Predicting survival from colorectal cancer histology slides using deep learning: A retrospective multicenter study. PLOS Med. 16, e1002730 (2019).
    1. Rodner E et al. Fully convolutional networks in multimodal nonlinear microscopy images for automated detection of head and neck carcinoma: pilot study. Head Neck 41, 116–121 (2019).
    1. Cruz-Roa A et al. High-throughput adaptive sampling for whole-slide histopathology image analysis (HASHI) via convolutional neural networks: application to invasive breast cancer detection. PLOS ONE 13, e0196828 (2018).
    1. Rumelhart DE, Hinton GE & Williams RJ Learning representations by back-propagating errors. Nature 323, 533–536 (1986).
    1. Hochreiter S & Schmidhuber J Long short-term memory. Neural Comput. 9, 1735–1780 (1997).
    1. Li W, Gauci M & Gross R in Proc. 15th Ann. Conf. Genet. Evolut. Computat. 223–230. (ACM, 2013).
    1. Hou L, et al. Unsupervised histopathology image synthesis. (2017).
    1. Shaban MT, Baur C, Navab N & Albarqouni S StainGAN: stain style transfer for digital histological images. IEEE 10.1109/ISBI.2019. 8759152 (2018).
    1. Xu Z, Moro CF, Bozoky B & Zhang Q GAN-based virtual re-staining: a promising solution for whole slide image analysis. (2019).
    1. Gadermayr M, Gupta L, Klinkhammer BM, Boor P & Merhof D Unsupervisedly training GANs for segmenting digital pathology with automatically generated annotations. Proc. Machine Learn. 102, 175–184 (2018).
    1. Kapil A, et al. Deep semi supervised generative learning for automated PD-L1 tumor cell scoring on NSCLC tissue needle biopsies. Sci. Rep. 8,17343 (2018).
    1. Boutros PC The path to routine use of genomic biomarkers in the cancer clinic. Genome Res. 25, 1508–1513 (2015).
    1. Chatterjee SK & Zetter BR Cancer biomarkers: knowing the present and predicting the future. Future Oncol. 1, 37–50 (2005).
    1. Grigg C & Rizvi NA PD-L1 biomarker testing for non-small cell lung cancer: truth or fiction? J. Immunother. Cancer 4, 48 (2016).
    1. Mroz EA & Rocco JW Intra-tumor heterogeneity in head and neck cancer and its clinical implications. World J. Otorhinolaryngol. Head Neck Surg. 2, 60–67 (2016).
    1. Morris LGT et al. Pan-cancer analysis of intratumor heterogeneity as a prognostic determinant of survival. Oncotarget 7, 10051–10063 (2016).
    1. Sottoriva A et al. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc. Natl Acad. Sci. USA 110, 4009–4014 (2013).
    1. Verma N et al. Image-based risk score to predict recurrence of ER+ breast cancer in ECOG-ACRIN Cancer Research Group E2197. J. Clin. Oncol. 36 (15_suppl), 540 (2018).
    1. Ginsburg SB, Lee G, Ali S & Madabhushi A Feature importance in nonlinear embeddings (FINE): applications in digital pathology. IEEE Trans. Med. Imaging. 35, 76–88 (2016).
    1. Cooper LAD et al. Novel genotype-phenotype associations in human cancers enabled by advanced molecular platforms and computational analysis of whole slide images. Lab. Investig. J. Tech. Methods Pathol. 95, 366–376 (2015).
    1. Lloyd MC, Johnson JO, Kasprzak A & Bui MM Image analysis of the tumor microenvironment. Adv. Exp. Med. Biol. 936, 1–10 (2016).
    1. Hamilton PW et al. Digital pathology and image analysis in tissue biomarker research. Methods 70, 59–73 (2014).
    1. Barsoum I, Tawedrous E, Faragalla H & Yousef GM Histo-genomics: digital pathology at the forefront of precision medicine. Diagnosis 10.1515/dx-2018-0064 (2018).
    1. FDA. Developing a software precertification program: a working model. https:// (2019).
    1. Daniel G, Silcox C, Sharma I & Wright M Current state and near-term priorities for AI-enabled diagnostic support software in health care. (2019).
    1. Pesapane F, Volonté C, Codari M & Sardanelli F Artificial intelligence as a medical device in radiology: ethical and regulatory issues in Europe and the United States. Insights Imaging 9, 745–753 (2018).
    1. Rudin C Stop explaining black box machine learning models for high stakes decisions and use interpretable models instead. Nat. Mach. Intell. 1, 206–215 (2019).
    1. Food and Drug Administration. FDA in brief: FDA proposes improvements to the de novo pathway for novel medical devices to advance safe, effective, and innovative treatments for patients. (2018).
    1. Commission European. MDCG 2018–2 Future EU medical device nomenclature - description of requirements. (2018).
    1. European Economic Community. Council Directive 93/42/EEC of 14 June 1993 concerning medical devices. : 20071011:EN:PDF (1993).
    1. Parliament European. Regulation (EU) 2017/745 of the European Parliament and of the Council of 5 April 2017 on medical devices, amending directive 2001/83/EC, regulation (EC) no 178/2002 and regulation (EC) no 1223/2009 and repealing council directives 90/385/EEC and 93/42/EEC (text with EEA relevance). (2017).
    1. Parliament European. Regulation (EU) 2017/746 of the European Parliament and of the council of 5 April 2017 on in vitro diagnostic medical devices and repealing directive 98/79/EC and commission decision 2010/227/EU (Text with EEA relevance) (2017).
    1. García-Rojo M, et al. New European Union regulations related to whole slide image scanners and image analysis software. J. Pathol. Inform. 10, 10.4103/jpi.jpi_33_18 (2019).
    1. Arterys. Medical imaging cloud AI. (2019).
    1. Arterys Inc. Arterys receives first FDA clearance for broad oncology imaging suite with deep learning. (2018).
    1. Food and Drug Administration. IntelliSite Pathology Solution (PIPS, Philips Medical Systems). (2017).
    1. PAIGE. PAIGE. (2019).
    1. Wire Business. FDA grants breakthrough designation to . (2019).
    1. Bueno G, Fernández-Carrobles MM, Deniz O & García-Rojo M New trends of emerging technologies in digital pathology. Pathobiology 83, 61–69 (2016).
    1. Food and Drug Administration. Radiological computer-assisted diagnostic (CADx) software for lesions suspicious for cancer. . (2017).
    1. Agendia. MammaPrint Test. (2019).
    1. Food and Drug Administration. MammaPrint 510(k) premarket notification. (2019).
    1. OncotypeIQ. Smarter Cancer Care - Genomic Testing. (2019).
    1. Food and Drug Administration. FDA issues warning letter to genomics lab for illegally marketing genetic test that claims to predict patients’ responses to specific medications. (2019).
    1. Food and Drug Administration. Discussion paper on laboratory developed tests (LDTs) January 13, 2017 (2019).
    1. Paxton A Laboratory-developed tests CAP suggests added oversight of homebrews. (2009).
    1. Higgins C Applications and challenges of digital pathology and whole slide imaging. Biotech. Histochem. 90, 341–347 (2015).
    1. Tizhoosh HR & Pantanowitz L Artificial intelligence and digital pathology: challenges and opportunities. J. Pathol. Inform. 9, 38 (2018).
    1. Doyle S, Feldman M, Tomaszewski J & Madabhushi A A boosted Bayesian multiresolution classifier for prostate cancer detection from digitized needle biopsies. IEEE Trans. Biomed. Eng. 59, 1205–1218 (2012).
    1. Huang B, Bates M & Zhuang X Super resolution fluorescence microscopy. Annu. Rev. Biochem. 78, 993–1016 (2009).
    1. Kleppe A et al. Chromatin organisation and cancer prognosis: a pan-cancer study. Lancet Oncol. 19, 356–369 (2018).
    1. Gallas BD NCIP Hub - Group: eeDAP studies ~ Wiki: High-throughput Truthing - Year 2. (2018).
    1. Challenge Grand. All Challenges. (2019).
    1. Ching P et al. Opportunities and obstacles for deep learning in biology and medicine. J. R. Soc. Interface 15, 20170387 (2018).
    1. Coudray N et al. Classification and mutation prediction from non-small cell lung cancer histopathology images using deep learning. Nat. Med. 24, 1559–1567 (2018).
    1. Zech JR et al. Variable generalization performance of a deep learning model to detect pneumonia in chest radiographs: a cross-sectional study. PLOS Med. 15, e1002683 (2018).
    1. Yagi Y & Gilbertson JR Digital Imaging in Pathology: The Case for Standardization. (SAGE Publications, 2005).
    1. Janowczyk A, Basavanhally A & Madabhushi A Stain Normalization using Sparse AutoEncoders (StaNoSA): application to digital pathology. Comput. Med. Imaging Graph. 57, 50–61 (2017).
    1. Khan AM, Rajpoot N, Treanor D & Magee D A nonlinear mapping approach to stain normalization in digital histopathology images using image-specific color deconvolution. IEEE Trans. Biomed. Eng. 61, 1729–1738 (2014).
    1. Vahadane A et al. Structure-preserving color normalization and sparse stain separation for histological images. IEEE Trans. Med. Imaging. 35, 1962–1971 (2016).
    1. 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. 3, 1–7 (2019).
    1. choosehappy. HistoQC is an Open-source Quality Control Tool for Digital Pathology Slides: choosehappy/ HistoQC. (2019).
    1. Leo P et al. Stable and discriminating features are predictive of cancer presence and Gleason grade in radical prostatectomy specimens: a multi-site study. Sci. Rep. 8, 14918 (2018).
    1. Leo P et al. Evaluating stability of histomorphometric features across scanner and staining variations: prostate cancer diagnosis from whole slide images. J. Med. Imaging 3, 047502 (2016).
    1. Lowe A et al. Validation of digital pathology in a healthcare environment. Digitalpathologyassociation. org (2011).
    1. Montalto MC An industry perspective: an update on the adoption of whole slide imaging. J. Pathol. Inform. 7, 18 (2016).
    1. Digital Pathology Association. Healthcare FAQs. (2019).
    1. Simon RM, Paik S & Hayes DF Use of archived specimens in evaluation of prognostic and predictive biomarkers. J. Natl Cancer Inst. 101, 1446–1452 (2009).
    1. Hayes DF et al. Tumor marker utility grading system: a framework to evaluate clinical utility of tumor markers. J. Natl Cancer Inst. 88, 1456–1466 (1996).
    1. Goetz MP et al. NCCN Guidelines insights: breast cancer, version 3.2018. J. Natl Compr. Canc. Netw. 17, 118–126 (2019).
    1. Sparano JA et al. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N. Engl. J. Med. 379, 111–121 (2018).
    1. Cardoso F et al. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N. Engl. J. Med. 375, 717–729 (2016).
    1. Paik S et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N. Engl. J. Med. 351, 2817–2826 (2004).
    1. Paik S et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J. Clin. Oncol. 24, 3726–3734 (2006).
    1. DecipherBio. GenomeDx Biosciences: Where to get decipher prostate cancer classifier. (2019).
    1. Erho N et al. Discovery and validation of a prostate cancer genomic classifier that predicts early metastasis following radical prostatectomy. PLOS ONE. 8, e66855 (2013).
    1. Carroll PH & Mohler JL NCCN guidelines updates: prostate cancer and prostate cancer early detection. J. Natl Compr. Canc. Netw. 16(5S), 620–623 (2018).
    1. Bhargava R & Madabhushi A Emerging themes in image informatics and molecular analysis for digital pathology. Annu. Rev. Biomed. Eng. 18, 387–412 (2016).
    1. Madabhushi A Digital pathology image analysis: opportunities and challenges. Imaging Med. 1, 7–10 (2009).
    1. Madabhushi A, Agner S, Basavanhally A, Doyle S &Lee G Computer-aided prognosis: predicting patient and disease outcome via quantitative fusion of multi-scale, multi-modal data. Comput. Med. Imaging Graph. 35, 506–514 (2011).
    1. Pantanowitz L et al. Validating whole slide imaging for diagnostic purposes in pathology: guideline from the College of American Pathologists Pathology and Laboratory Quality Center. Arch. Pathol. Lab. Med. 137, 1710–1722 (2013).
    1. Fuchs TJ, Wild PJ, Moch H & Buhmann JM Computational pathology analysis of tissue microarrays predicts survival of renal clear cell carcinoma patients. Med. Image Comput. Comput. Assist. Interv. 11, 1–8 (2008).
    1. Hayback J Mechanisms of Molecular Carcinogenesis - Volume 2. (Springer, 2017).
    1. Proscia. Proscia digital pathology. (2019).
    1. Lens Deep. Digital pathology cloud platform. (2019).
    1. PathAI. PathAI. (2019).
    1. Aifora. WebMicroscope. Big pictures. Deep Diagnosis. https:// (2019).
    1. Glaser AK, et al. Light-sheet microscopy for slide-free non-destructive pathology of large clinical specimens. Nat. Biomed. Eng. 1, 0084 (2017).
    1. Fereidouni F et al. Microscopy with ultraviolet surface excitation for rapid slide-free histology. Nat. Biomed. Eng. 1, 957 (2017).
    1. Fraggetta F, Garozzo S, Zannoni GF, Pantanowitz L & Rossi ED Routine digital pathology workflow: the Catania experience. J. Pathol. Inform. 8, 51 (2017).
    1. Cheng CL et al. Enabling digital pathology in the diagnostic setting: navigating through the implementation journey in an academic medical centre. J. Clin. Pathol. 69, 784–792 (2016).
    1. Tomczak K, Czerwinska P & Wiznerowicz M The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge. Contemp. Oncol. 19, A68–77 (2015).
    1. Gutman DA et al. Cancer Digital Slide Archive: an informatics resource to support integrated in silico analysis of TCGA pathology data. J. Am. Med. Inform. Assoc. 20, 1091–1098 (2013).

Source: PubMed

3
Tilaa