M. D. Zarella, A practical guide to whole slide imaging: a white paper from the digital pathology association, Arch. Pathol. Lab. Med, vol.143, pp.222-234, 2019.

S. Mukhopadhyay, 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, vol.42, pp.39-52, 2018.

H. Wang, Mitosis detection in breast cancer pathology images by combining handcrafted and convolutional neural network features, J. Med. Imaging Bellingham Wash, vol.1, p.34003, 2014.

R. Turkki, N. Linder, P. E. Kovanen, T. Pellinen, and J. Lundin, Antibodysupervised deep learning for quantification of tumor-infiltrating immune cells in hematoxylin and eosin stained breast cancer samples, J. Pathol. Inform, vol.7, p.38, 2016.

L. Hou, Patch-based convolutional neural network for whole slide tissue image classification, Proc. IEEE Comput. Soc. Conf. Comput. Vis. Pattern Recognit, pp.2424-2433, 2016.

W. Bulten, Automated deep-learning system for Gleason grading of prostate cancer using biopsies: a diagnostic study, Lancet Oncol, vol.21, pp.233-241, 2020.

M. C. Montalto and R. Edwards, And they said it couldn't be done: predicting known driver mutations from H&E slides, J. Pathol. Inform, vol.10, p.17, 2019.

N. Coudray, Classification and mutation prediction from non-small cell lung cancer histopathology images using deep learning, Nat. Med, vol.24, pp.1559-1567, 2018.

P. Mobadersany, Predicting cancer outcomes from histology and genomics using convolutional networks, Proc. Natl Acad. Sci. USA, vol.115, pp.2970-2979, 2018.

A. J. Schaumberg, M. A. Rubin, and T. J. Fuchs, H&E-stained whole slide image deep learning predicts SPOP mutation state in prostate cancer, 2018.

P. Chang, Deep-learning convolutional neural networks accurately classify genetic mutations in gliomas, Am. J. Neuroradiol, vol.39, pp.1201-1207, 2018.

J. Noorbakhsh, Pan-cancer classifications of tumor histological images using deep learning, 2020.

H. Xu, S. Park, S. H. Lee, and T. H. Hwang, Using transfer learning on whole slide images to predict tumor mutational burden in bladder cancer patients, Bioinformatics, 2019.

E. Segal, N. Friedman, N. Kaminski, A. Regev, and D. Koller, From signatures to models: understanding cancer using microarrays, Nat. Genet, vol.37, pp.38-45, 2005.

E. S. Lander, Array of hope, Nat. Genet, vol.21, pp.3-4, 1999.

A. Conesa, A survey of best practices for RNA-seq data analysis, Genome Biol, vol.17, p.13, 2016.

S. Anders and W. Huber, Differential expression analysis for sequence count data, Genome Biol, vol.11, p.106, 2010.

S. Serratì, Next-generation sequencing: advances and applications in cancer diagnosis, OncoTargets Ther, vol.9, pp.7355-7365, 2016.

M. R. Stratton, P. J. Campbell, and P. Futreal, The cancer genome, Nature, vol.458, pp.719-724, 2009.

R. Kamps, Next-generation sequencing in oncology: genetic diagnosis, risk prediction and cancer classification, Int. J. Mol. Sci, vol.18, p.308, 2017.

N. Merienne, Cell-type-specific gene expression profiling in adult mouse brain reveals normal and disease-state signatures, Cell Rep, vol.26, pp.2477-2493, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02087719

I. Nassiri and M. N. Mccall, Systematic exploration of cell morphological phenotypes associated with a transcriptomic query, Nucleic Acids Res, vol.46, pp.116-116, 2018.

U. Mcdermott, J. R. Downing, and M. R. Stratton, Genomics and the continuum of cancer care, N. Engl. J. Med, vol.364, pp.340-350, 2011.

P. Courtiol, E. W. Tramel, M. Sanselme, and G. Wainrib, Classification and disease localization in histopathology using only global labels: a weaklysupervised approach, 2018.

E. K. Kleczko, J. W. Kwak, E. L. Schenk, and R. A. Nemenoff, Targeting the complement pathway as a therapeutic strategy in lung cancer, Front. Immunol, vol.10, p.954, 2019.

I. Todros-dawda, L. Kveberg, J. T. Vaage, and M. Inngjerdingen, The tetraspanin CD53 modulates responses from activating NK cell receptors, promoting LFA-1 activation and dampening NK cell effector functions, PLoS ONE, vol.9, p.97844, 2014.

Q. G. Medley, Characterization of GMP-17, a granule membrane protein that moves to the plasma membrane of natural killer cells following target cell recognition, Proc. Natl Acad. Sci. USA, vol.93, pp.685-689, 1996.

T. Sakurai and M. Kudo, Molecular link between liver fibrosis and hepatocellular carcinoma, Liver Cancer, vol.2, pp.365-366, 2013.

P. Apostolou and I. Papasotiriou, Current perspectives on CHEK2 mutations in breast cancer, Breast Cancer Dove Med. Press, vol.9, pp.331-335, 2017.

S. Zrihan-licht, Association of csk-homologous kinase (CHK) (formerly MATK) with HER-2/ErbB-2 in breast cancer cells, J. Biol. Chem, vol.272, pp.1856-1863, 1997.

R. L. Sutherland and E. A. Musgrove, Cyclins and breast cancer, J. Mammary Gland Biol. Neoplasia, vol.9, pp.95-104, 2004.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, pp.646-674, 2011.

Y. Rivenson, Virtual histological staining of unlabelled tissueautofluorescence images via deep learning, Nat. Biomed. Eng, vol.3, pp.466-477, 2019.

J. Saltz, Spatial organization and molecular correlation of tumorinfiltrating lymphocytes using deep learning on pathology images, Cell Rep, vol.23, pp.181-193, 2018.

J. N. Kather, Predicting survival from colorectal cancer histology slides using deep learning: A retrospective multicenter study, PLoS Med, vol.16, p.1002730, 2019.

J. N. Kather, 000 histological images of human colorectal cancer and healthy tissue (Version v0.1), vol.100, 2018.

A. J. Gentles, The prognostic landscape of genes and infiltrating immune cells across human cancers, Nat. Med, vol.21, pp.938-945, 2015.

E. Y. Loh, Identification and sequence of a fourth human T cell antigen receptor chain, Nature, vol.330, pp.569-572, 1987.

S. Kneissl, CD19 and CD20 targeted vectors induce minimal activation of resting B lymphocytes, PLoS ONE, vol.8, p.79047, 2013.

P. Bankhead, QuPath: open source software for digital pathology image analysis, Sci. Rep, vol.7, pp.1-7, 2017.

W. Bulten, Epithelium segmentation using deep learning in H&E-stained prostate specimens with immunohistochemistry as reference standard, Sci. Rep, vol.9, p.864, 2019.

W. Bulten, PESO: prostate epithelium segmentation on H&E-stained prostatectomy whole slide images (Version 1, 2018.

C. Saillard, Predicting survival after hepatocellular carcinoma resection using deep-learning on histological slides, Hepatology, 2020.

T. Maeda, Small hepatocellular carcinoma of single nodular type: a specific reference to its surrounding cancerous area undetected radiologically and macroscopically, J. Surg. Oncol, vol.60, pp.75-79, 1995.

I. O. Ng, J. Na, E. C. Lai, S. T. Fan, and M. Ng, Ki-67 antigen expression in hepatocellular carcinoma using monoclonal antibody MIB1. A comparison with proliferating cell nuclear antigen, Am. J. Clin. Pathol, vol.104, p.313, 1995.

K. Shirabe, A long-term survivor of ruptured hepatocellular carcinoma after hepatic resection, J. Gastroenterol. Hepatol, vol.10, p.351, 1995.

Y. Luo, Clinicopathological and prognostic significance of high Ki-67 labeling index in hepatocellular carcinoma patients: a meta-analysis, Int. J. Clin. Exp. Med, vol.8, pp.10235-10247, 2015.

J. M. Llovet and C. Brú, Bruix Prognosis of hepatocellular carcinoma: the BCLC staging classification, J. Semin Liver Dis, vol.19, pp.329-338, 1999.

I. Cortes-ciriano, S. Lee, W. Park, T. Kim, and P. J. Park, A molecular portrait of microsatellite instability across multiple cancers, Nat. Commun, vol.8, p.15180, 2017.

D. I. Strokotov, Is there a difference between T-and B-lymphocyte morphology, J. Biomed. Optics, vol.14, p.64036, 2009.

A. K. Win, Colorectal and other cancer risks for carriers and noncarriers from families with a DNA mismatch repair gene mutation: a prospective cohort study, J. Clin. Oncol, vol.30, pp.958-964, 2012.

C. R. Boland and A. Goel, Microsatellite instability in colorectal cancer, Gastroenterology, vol.138, 2010.

D. T. Le, Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade, Science, vol.357, pp.409-413, 2017.

S. Lemery, P. Keegan, and R. Pazdur, First FDA approval agnostic of cancer sitewhen a biomarker defines the indication, N. Engl. J. Med, vol.377, pp.1409-1412, 2017.

J. N. Kather, N. Halama, and D. Jaeger, Genomics and emerging biomarkers for immunotherapy of colorectal cancer, Semin. Cancer Biol, vol.52, pp.189-197, 2018.

J. N. Kather, Deep learning can predict microsatellite instability directly from histology in gastrointestinal cancer, Nat. Med, vol.25, pp.1054-1056, 2019.

A. H. Beck, Systematic analysis of breast cancer morphology uncovers stromal features associated with survival, Sci. Transl. Med, vol.3, pp.108-113, 2011.

J. Whitney, Quantitative nuclear histomorphometry predicts oncotype DX risk categories for early stage ER+ breast cancer, BMC Cancer, vol.18, p.610, 2018.

R. R. Rawat, D. Ruderman, P. Macklin, D. L. Rimm, and D. B. Agus, Correlating nuclear morphometric patterns with estrogen receptor status in breast cancer pathologic specimens, Breast Cancer, vol.4, p.32, 2018.

S. Haider and R. Pal, Integrated analysis of transcriptomic and proteomic data, Curr. Genomics, vol.2, pp.91-110, 2013.

M. I. Love, W. Huber, and S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, vol.15, p.550, 2014.

N. Otsu, A threshold selection method from gray-level histograms, IEEE Trans. Syst., man, Cybern, vol.9, pp.62-66, 1979.

K. He, X. Zhang, S. Ren, and J. Sun, Deep residual learning for image recognition, Proceedings of the IEEE Conference on Computer Vision and Pattern Recognition, pp.770-778, 2016.

J. Deng, Imagenet: a large-scale hierarchical image database, 2009 IEEE Conference on Computer Vision and Pattern Recognition, pp.248-255, 2009.

R. Achanta, SLIC superpixels compared to state-of-the-art superpixel methods, IEEE Trans. Pattern Anal. Mach. Intell, vol.34, pp.2274-2282, 2022.

A. Colaprico, TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data, Nucleic Acids Res, vol.44, pp.71-71, 2016.

C. R. Boland, A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer, Cancer Res, vol.58, pp.5248-5257, 1998.

, Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer, Nature, vol.487, p.330, 2012.

R. Arora, A. Basu, P. Mianjy, and A. Mukherjee, Understanding deep neural networks with rectified linear units, 2016.