S. Eric, L. M. Lander, B. Linton, C. Birren, . Nusbaum et al.,

D. Houle, R. Diddahally, S. Govindaraju, and . Omholt, Phenomics: the next challenge, Nature reviews genetics, vol.11, issue.12, pp.855-866, 2010.

M. Mahner and M. Kary, What exactly are genomes, genotypes and phenotypes? and what about phenomes?, Journal of theoretical biology, vol.186, issue.1, pp.55-63, 1997.

G. Myers, Why bioimage informatics matters, Nature methods, vol.9, issue.7, p.659, 2012.

A. Steven and . Haney, High content screening: science, techniques and applications, 2008.

B. Neumann, T. Walter, J. Hériché, J. Bulkescher, H. Erfle et al., Phenotypic profiling of the human genome by timelapse microscopy reveals cell division genes, Nature, vol.464, issue.7289, p.721, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01144034

C. Jeremy, B. Simpson, V. Joggerst, F. Laketa, C. Verissimo et al., Genome-wide rnai screening identifies human proteins with a regulatory function in the early secretory pathway, Nature cell biology, vol.14, issue.7, pp.764-774, 2012.

C. Collinet, M. Stöter, R. Charles, N. Bradshaw, . Samusik et al., Systems survey of endocytosis by multiparametric image analysis, Nature, vol.464, issue.7286, pp.243-249, 2010.

V. Michael, . Boland, K. Mia, R. Markey, and . Murphy, Automated recognition of patterns characteristic of subcellular structures in fluorescence microscopy images, Cytometry: The Journal of the International Society for Analytical Cytology, vol.33, issue.3, pp.366-375, 1998.

A. Steven, P. Haney, J. Lapan, J. Pan, and . Zhang, High-content screening moves to the front of the line, Drug discovery today, vol.11, issue.19, pp.889-894, 2006.

R. Pepperkok and J. Ellenberg, High-throughput fluorescence microscopy for systems biology, Nature reviews Molecular cell biology, vol.7, issue.9, pp.690-696, 2006.

C. David, J. Swinney, and . Anthony, How were new medicines discovered?, Nature reviews Drug discovery, vol.10, issue.7, pp.507-519, 2011.

F. William, J. T. Scherer, G. O. Syverton, and . Gey, Studies on the propagation in vitro of poliomyelitis viruses: Iv. viral multiplication in a stable strain of human malignant epithelial cells (strain hela) derived from an epidermoid carcinoma of the cervix, The Journal of experimental medicine, vol.97, issue.5, pp.695-710, 1953.

V. Marx, Cell-line authentication demystified, Nature methods, vol.11, issue.5, p.483, 2014.

S. Terjung, T. Walter, A. Seitz, B. Neumann, R. Pepperkok et al., High-throughput microscopy using live mammalian cells, Cold Spring Harbor Protocols, issue.8, p.84, 2010.

. Zachary-e-perlman, D. Michael, Y. Slack, T. J. Feng, L. F. Mitchison et al., Multidimensional drug profiling by automated microscopy, Science, vol.306, issue.5699, pp.1194-1198, 2004.

L. Cynthia, V. Adams, . Kutsyy, A. Daniel, G. Coleman et al., Compound classification using image-based cellular phenotypes, In Methods in enzymology, vol.414, pp.440-468, 2006.

M. Bray, S. Singh, H. Han, T. Chadwick, B. Davis et al., Cell painting, a highcontent image-based assay for morphological profiling using multiplexed fluorescent dyes, Nature protocols, vol.11, issue.9, p.1757, 2016.

N. Orlov, L. Shamir, T. Macura, J. Johnston, . Mark-eckley et al., Wnd-charm: Multi-purpose image classification using compound image transforms, Pattern recognition letters, vol.29, issue.11, pp.1684-1693, 2008.

V. Uhlmann, S. Singh, and A. E. Carpenter, Cp-charm: segmentation-free image classification made accessible, BMC bioinformatics, vol.17, issue.1, p.51, 2016.

Z. Oren, J. L. Kraus, B. J. Ba, and . Frey, Classifying and segmenting microscopy images with deep multiple instance learning, Bioinformatics, vol.32, issue.12, pp.52-59, 2016.

D. Michael, E. D. Slack, L. F. Martinez, S. J. Wu, and . Altschuler, Characterizing heterogeneous cellular responses to perturbations. Proceedings of the National Academy of Sciences, p.807038105, 2008.

J. William, I. Godinez, . Hossain, E. Stanley, J. W. Lazic et al., A multi-scale convolutional neural network for phenotyping high-content cellular images, Bioinformatics, vol.33, issue.13, pp.2010-2019, 2017.

C. Juan, S. Caicedo, F. Cooper, S. Heigwer, P. Warchal et al., Data-analysis strategies for image-based cell profiling, Nature methods, vol.14, issue.9, p.849, 2017.

A. Euan and . Ashley, Towards precision medicine, Nature Reviews Genetics, vol.17, issue.9, p.507, 2016.

C. Krittanawong, H. Zhang, Z. Wang, M. Aydar, and T. Kitai, Artificial intelligence in precision cardiovascular medicine, Journal of the American College of Cardiology, vol.69, issue.21, pp.2657-2664, 2017.

T. Hulsen, A. Saumya-shekhar-jamuar, J. Moody, V. Hansen-karnes, S. Orsolya et al., From big data to precision medicine, Frontiers in medicine, vol.6, p.34, 2019.

C. James, L. M. Costello, E. Heiser, M. Georgii, . Gönen et al., A community effort to assess and improve drug sensitivity prediction algorithms, Nature biotechnology, vol.32, issue.12, p.1202, 2014.

J. Steven, L. F. Altschuler, and . Wu, Cellular heterogeneity: do differences make a difference?, Cell, vol.141, issue.4, pp.559-563, 2010.

F. Rose, S. Basu, E. Rexhepaj, A. Chauchereau, E. D. Nery et al., Compound functional prediction using multiple unrelated morphological profiling assays, SLAS TECHNOLOGY: Translating Life Sciences Innovation, vol.23, issue.3, pp.243-251, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02425375

J. Scott, . Warchal, C. John, N. O. Dawson, and . Carragher, Development of the theta comparative cell scoring method to quantify diverse phenotypic responses between distinct cell types. Assay and drug development technologies, vol.14, pp.395-406, 2016.

C. Sommer, R. Hoefler, M. Samwer, and D. W. Gerlich, A deep learning and novelty detection framework for rapid phenotyping in high-content screening, Molecular biology of the cell, vol.28, issue.23, pp.3428-3436, 2017.

O. Russakovsky, J. Deng, H. Su, J. Krause, S. Satheesh et al., Imagenet large scale visual recognition challenge, International Journal of Computer Vision, vol.115, issue.3, pp.211-252, 2015.

T. Lin, M. Maire, S. Belongie, J. Hays, P. Perona et al., Microsoft coco: Common objects in context, European conference on computer vision, pp.740-755, 2014.

V. Ljosa, K. L. Sokolnicki, and A. E. Carpenter, Annotated high-throughput microscopy image sets for validation, Nature methods, vol.9, issue.7, pp.637-637, 2012.

R. E. Peter-d-caie, A. Walls, S. Ingleston-orme, T. Daya, R. Houslay et al., Highcontent phenotypic profiling of drug response signatures across distinct cancer cells, Molecular cancer therapeutics, vol.9, issue.6, pp.1913-1926, 2010.

C. Kandaswamy, M. Luís, . Silva, J. M. Luís-a-alexandre, and . Santos, High-content analysis of breast cancer using single-cell deep transfer learning, Journal of biomolecular screening, vol.21, issue.3, pp.252-259, 2016.

D. Mahajan, R. Girshick, V. Ramanathan, K. He, M. Paluri et al., Exploring the limits of weakly supervised pretraining, Proceedings of the European Conference on Computer Vision (ECCV), pp.181-196, 2018.

T. Chen, S. Kornblith, M. Norouzi, and G. Hinton, A simple framework for contrastive learning of visual representations, 2020.

M. Eric, . Christiansen, J. Samuel, . Yang, A. Michael-ando et al., In silico labeling: Predicting fluorescent labels in unlabeled images, Cell, vol.173, issue.3, pp.792-803, 2018.

C. Ounkomol, S. Seshamani, M. M. Maleckar, F. Collman, and G. , Label-free prediction of threedimensional fluorescence images from transmitted-light microscopy, Nature methods, vol.15, issue.11, p.917, 2018.

P. Sajith-kecheril-sadanandan, S. L. Ranefall, C. Guyader, and . Wählby, Automated training of deep convolutional neural networks for cell segmentation, Scientific reports, vol.7, issue.1, p.7860, 2017.

P. Diederik, M. Kingma, and . Welling, Auto-encoding variational bayes, 2013.

J. Ian and . Goodfellow, Jonathon Shlens, and Christian Szegedy. Explaining and harnessing adversarial examples, 2014.

A. Van-den-oord, N. Kalchbrenner, and K. Kavukcuoglu, Pixel recurrent neural networks, 2016.

A. Osokin, A. Chessel, R. Salas, and F. Vaggi, Gans for biological image synthesis, Proceedings of the IEEE International Conference on Computer Vision, pp.2233-2242, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01611692

P. Isola, J. Zhu, T. Zhou, and A. A. Efros, Image-toimage translation with conditional adversarial networks, Proceedings of the IEEE conference on computer vision and pattern recognition, pp.1125-1134, 2017.

J. Zhu, T. Park, P. Isola, and A. A. Efros, Unpaired image-to-image translation using cycle-consistent adversarial networks, Proceedings of the IEEE international conference on computer vision, pp.2223-2232, 2017.

A. M. Stephan-j-ihle, S. Reichmuth, H. Girardin, F. Han, A. Stauffer et al., Unsupervised data to content transformation with histogram-matching cycle-consistent generative adversarial networks, Nature Machine Intelligence, vol.1, issue.10, pp.461-470, 2019.

J. Boyd, A. Pinhiero, E. D. Nery, F. Reyal, and T. Walter, Analysing double-strand breaks in cultured cells for drug screening applications by causal inference, IEEE 15th International Symposium on, pp.445-448, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01984323

H. Ajakan, P. Germain, H. Larochelle, F. Laviolette, and M. Marchand, Domain-adversarial neural networks, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01624607

C. Joseph, A. Boyd, E. D. Pinheiro, F. Nery, T. Reyal et al., Domain-invariant features for mechanism of action prediction in a multi-cell-line drug screen, Bioinformatics, vol.36, issue.5, pp.1607-1613, 2020.

J. Boyd, F. Reyal, A. Pinheiro, E. D. Nery, and T. Walter, Domain-invariant features for mechanism of action prediction in a multi-cell-line drug screen, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02440603

J. Boyd, Z. Gouveia, F. Perez, and T. Walter, Experimentally-generated ground truth for detecting cell types in an image-based immunotherapy screen, 2020 IEEE 17th International Symposium on Biomedical Imaging, 2020.

J. Boyd, Z. Gouveia, F. Perez, and T. Walter, Lymphocytes dataset, 2019.

P. Naylor, J. Boyd, M. Laé, F. Reyal, and T. Walter, Predicting residual cancer burden in a triple negative breast cancer cohort, 2019 IEEE 16th International Symposium on Biomedical Imaging (ISBI 2019), pp.933-937, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02440647

B. Khalfaoui, J. Boyd, and J. Vert, Adaptive structured noise injection for shallow and deep neural networks, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02025929

T. Tieleman and G. Hinton, Lecture 6.5-rmsprop: Divide the gradient by a running average of its recent magnitude. COURSERA: Neural networks for machine learning, vol.4, pp.26-31, 2012.

P. Diederik, J. Kingma, and . Ba, Adam: A method for stochastic optimization, 2014.

G. E. David-e-rumelhart, R. Hinton, and . Williams, Learning internal representations by error propagation, 1985.

Y. Lecun, L. Bottou, Y. Bengio, and P. Haffner, Gradientbased learning applied to document recognition, Proceedings of the IEEE, vol.86, issue.11, pp.2278-2324, 1998.

D. Matthew, R. Zeiler, and . Fergus, Visualizing and understanding convolutional networks, European conference on computer vision, pp.818-833, 2014.

A. Krizhevsky, I. Sutskever, and G. E. Hinton, Imagenet classification with deep convolutional neural networks, Advances in neural information processing systems, pp.1097-1105, 2012.

K. Simonyan and A. Zisserman, Very deep convolutional networks for large-scale image recognition, 2014.

C. Szegedy, W. Liu, Y. Jia, P. Sermanet, S. Reed et al., Going deeper with convolutions, Proceedings of the IEEE conference on computer vision and pattern recognition, pp.1-9, 2015.

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.

M. Tan, V. Quoc, . Le, and . Efficientnet, Rethinking model scaling for convolutional neural networks, 2019.

A. Karpathy, What i learned from competing against a convnet on imagenet, Andrej Karpathy Blog, vol.5, pp.1-15, 2014.

A. Paszke, S. Gross, S. Chintala, G. Chanan, E. Yang et al., Automatic differentiation in pytorch, 2017.

M. Everingham, L. Van-gool, K. I. Christopher, J. Williams, A. Winn et al., The pascal visual object classes (voc) challenge, International journal of computer vision, vol.88, issue.2, pp.303-338, 2010.

P. Sermanet, D. Eigen, X. Zhang, and M. Mathieu, Rob Fergus, and Yann LeCun. Overfeat: Integrated recognition, localization and detection using convolutional networks, 2013.

R. Girshick, J. Donahue, T. Darrell, and J. Malik, Rich feature hierarchies for accurate object detection and semantic segmentation, Proceedings of the IEEE conference on computer vision and pattern recognition, pp.580-587, 2014.

R. Girshick, Fast r-cnn, Proceedings of the IEEE international conference on computer vision, pp.1440-1448, 2015.

K. Shaoqing-ren, R. He, J. Girshick, and . Sun, Faster r-cnn: Towards real-time object detection with region proposal networks, Advances in neural information processing systems, pp.91-99, 2015.

K. He, G. Gkioxari, P. Dollár, and R. Girshick, Mask r-cnn, Proceedings of the IEEE international conference on computer vision, pp.2961-2969, 2017.

K. Hornik, M. Stinchcombe, and H. White, Multilayer feedforward networks are universal approximators, Neural networks, vol.2, issue.5, pp.359-366, 1989.

N. Srivastava, G. Hinton, A. Krizhevsky, I. Sutskever, and R. Salakhutdinov, Dropout: a simple way to prevent neural networks from overfitting, The Journal of Machine Learning Research, vol.15, issue.1, pp.1929-1958, 2014.

S. Ioffe and C. Szegedy, Batch normalization: Accelerating deep network training by reducing internal covariate shift, 2015.

C. Shorten, M. Taghi, and . Khoshgoftaar, A survey on image data augmentation for deep learning, Journal of Big Data, vol.6, issue.1, p.60, 2019.

V. Nitesh, K. W. Chawla, L. O. Bowyer, W. Hall, and . Kegelmeyer, Smote: synthetic minority over-sampling technique, Journal of artificial intelligence research, vol.16, pp.321-357, 2002.

Y. Patrice, D. Simard, J. C. Steinkraus, and . Platt, Best practices for convolutional neural networks applied to visual document analysis, Icdar, vol.3, 2003.

I. Goodfellow, J. Pouget-abadie, M. Mirza, B. Xu, D. Warde-farley et al., Generative adversarial nets, Advances in neural information processing systems, pp.2672-2680, 2014.

A. Leon, A. S. Gatys, M. Ecker, and . Bethge, Image style transfer using convolutional neural networks, Proceedings of the IEEE conference on computer vision and pattern recognition, pp.2414-2423, 2016.

Q. Sinno-jialin-pan and . Yang, A survey on transfer learning, IEEE Transactions on knowledge and data engineering, vol.22, issue.10, pp.1345-1359, 2009.

A. Sharif-razavian, H. Azizpour, J. Sullivan, and S. Carlsson, Cnn features off-the-shelf: an astounding baseline for recognition, Proceedings of the IEEE conference on computer vision and pattern recognition workshops, pp.806-813, 2014.

S. Ben-david, J. Blitzer, K. Crammer, A. Kulesza, F. Pereira et al., A theory of learning from different domains, Machine learning, vol.79, issue.1-2, pp.151-175, 2010.

Y. Ganin and V. Lempitsky, Unsupervised domain adaptation by backpropagation, 2014.

L. Metz, B. Poole, D. Pfau, and J. Sohl-dickstein, Unrolled generative adversarial networks, 2016.

A. Radford, L. Metz, and S. Chintala, Unsupervised representation learning with deep convolutional generative adversarial networks, 2015.

M. Mirza and S. Osindero, Conditional generative adversarial nets, 2014.

A. Bansal, S. Ma, D. Ramanan, and Y. Sheikh, Recycle-gan: Unsupervised video retargeting, Proceedings of the European conference on computer vision (ECCV), pp.119-135, 2018.

T. Karras, T. Aila, S. Laine, and J. Lehtinen, Progressive growing of gans for improved quality, stability, and variation, 2017.

A. Clifford, L. Hudis, and . Gianni, Triple-negative breast cancer: an unmet medical need, Oncologist, vol.16, 2011.

J. Kathryn, . Chavez, V. Sireesha, S. Garimella, and . Lipkowitz, Triple negative breast cancer cell lines: one tool in the search for better treatment of triple negative breast cancer, Breast disease, vol.32, issue.1-2, p.35, 2010.

M. Homero-gonçalves, J. Guerra, and . Cintra, Vívian Assis Fayer, Igor Vilela Brum, and Maria Teresa Bustamante Teixeira. Survival study of triple-negative and non-triple-negative breast cancer in a brazilian cohort, Clinical Medicine Insights: Oncology, vol.12, p.1179554918790563, 2018.

R. Hatem, R. E. Botty, S. Chateau-joubert, J. Servely, D. Labiod et al., Céline Callens, Sophie Vacher, et al. Targeting mtor pathway inhibits tumor growth in different molecular subtypes of triple-negative breast cancers, Oncotarget, vol.7, issue.30, p.48206, 2016.

J. Filmus, N. Michael, R. Pollak, R. N. Cailleau, and . Buick, Mda-468, a human breast cancer cell line with a high number of epidermal growth factor (egf) receptors, has an amplified egf receptor gene and is growth inhibited by egf, Biochemical and biophysical research communications, vol.128, issue.2, pp.898-905, 1985.

X. Dai, H. Cheng, Z. Bai, and J. Li, Breast cancer cell line classification and its relevance with breast tumor subtyping, Journal of Cancer, vol.8, issue.16, p.3131, 2017.

Y. Etienne, L. Lasfargues, and . Ozzello, Cultivation of human breast carcinomas, Journal of the National Cancer Institute, vol.21, issue.6, pp.1131-1147, 1958.

F. Adi, V. Gazdar, A. Kurvari, L. Virmani, M. Gollahon et al., Characterization of paired tumor and non-tumor cell lines established from patients with breast cancer, International journal of cancer, vol.78, issue.6, pp.766-774, 1998.

J. Adeline, H. S. Hackett, L. Smith, R. B. Springer, . Owens et al., Two syngeneic cell lines from human breast tissue: the aneuploid mammary epithelial (hs578t) and the diploid myoepithelial (hs578bst) cell lines. Journal of the National Cancer Institute, vol.58, pp.1795-1806, 1977.

D. Herbert, . Soule, M. Terry, S. R. Maloney, . Wolman et al., Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, mcf-10, Cancer research, vol.50, issue.18, pp.6075-6086, 1990.

. Br-brinkley, . Beall, . Lj-wible, . Mace, S. Donna et al., Variations in cell form and cytoskeleton in human breast carcinoma cells in vitro, Cancer research, vol.40, issue.9, pp.3118-3129, 1980.

T. Huang, G. Yang, and . Tang, A fast two-dimensional median filtering algorithm, IEEE Transactions on Acoustics, Speech, and Signal Processing, vol.27, issue.1, pp.13-18, 1979.

S. Beucher and C. Lantuéjoul, Use of watersheds in contour detection, 1979.

M. Grimaud, New measure of contrast: the dynamics, vol.1769, pp.292-305, 1992.

A. Thouis-r-jones, P. Carpenter, and . Golland, Voronoi-based segmentation of cells on image manifolds, International Workshop on Computer Vision for Biomedical Image Applications, pp.535-543, 2005.

N. Otsu, A threshold selection method from gray-level histograms, IEEE transactions on systems, man, and cybernetics, vol.9, pp.62-66, 1979.

M. Held, H. A. Michael, B. Schmitz, T. Fischer, B. Walter et al., Cellcognition: time-resolved phenotype annotation in highthroughput live cell imaging, Nature methods, vol.7, issue.9, pp.747-754, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01431427

T. Walter, M. Held, B. Neumann, J. Hériché, C. Conrad et al., Automatic identification and clustering of chromosome phenotypes in a genome wide rnai screen by time-lapse imaging, Journal of structural biology, vol.170, issue.1, pp.1-9, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01427996

I. Caraus, A. A. Alsuwailem, R. Nadon, and V. Makarenkov, Detecting and overcoming systematic bias in highthroughput screening technologies: A comprehensive review of practical issues and methodological solutions, vol.16, pp.974-986

V. Ljosa and A. E. Carpenter, Introduction to the Quantitative Analysis of Two-Dimensional Fluorescence Microscopy Images for Cell-Based Screening, vol.5, p.1000603

. De-pegg, Viability assays for preserved cells, tissues, and organs, Cryobiology, vol.26, issue.3, pp.212-231, 1989.

L. Mcinnes and J. Healy, Umap: Uniform manifold approximation and projection for dimension reduction, 2018.

R. Pepperkok and J. Ellenberg, High-throughput fluorescence microscopy for systems biology, Nature reviews Molecular cell biology, vol.7, issue.9, p.690, 2006.

V. Ljosa, D. Peter, R. T. Caie, K. L. Horst, E. L. Sokolnicki et al., Comparison of methods for imagebased profiling of cellular morphological responses to small-molecule treatment, Journal of biomolecular screening, vol.18, issue.10, pp.1321-1329, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02902061

P. Serge, W. Horbach, and . Halffman, The ghosts of hela: How cell line misidentification contaminates the scientific literature, PloS one, vol.12, issue.10, p.186281, 2017.

Y. Liu, Y. Mi, T. Mueller, S. Kreibich, E. G. Williams et al., Multi-omic measurements of heterogeneity in hela cells across laboratories, Nature biotechnology, vol.37, issue.3, p.314, 2019.

S. Singh, M. Bray, A. E. Jones, and . Carpenter, Pipeline for illumination correction of images for high-throughput microscopy, Journal of microscopy, vol.256, issue.3, pp.231-236, 2014.

T. Walter, M. Held, B. Neumann, J. Hériché, C. Conrad et al., Automatic identification and clustering of chromosome phenotypes in a genome wide RNAi screen by time-lapse imaging, Journal of structural biology, vol.170, issue.1, pp.1-9, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01427996

D. Müllner, Fast hierarchical, agglomerative clustering routines for r and python, Journal of Statistical Software, vol.53, issue.9, pp.1-18, 2013.

. Lit-hsin, L. F. Loo, S. J. Wu, and . Altschuler, Image-based multivariate profiling of drug responses from single cells, Nature methods, vol.4, issue.5, p.445, 2007.

R. Caruana-;-yaroslav-ganin, E. Ustinova, H. Ajakan, P. Germain, H. Larochelle et al., Domain-adversarial training of neural networks, The Journal of Machine Learning Research, vol.28, issue.1, pp.2096-2030, 1997.

F. Pedregosa, G. Varoquaux, A. Gramfort, V. Michel, B. Thirion et al., Scikitlearn: Machine learning in Python, Journal of Machine Learning Research, vol.12, pp.2825-2830, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00650905

F. Chollet, , 2015.

J. L. Stéfan-van-der-walt, J. Schönberger, F. Nunez-iglesias, J. D. Boulogne, N. Warner et al., scikit-image: image processing in Python. PeerJ, 2:e453, 2014.

L. Van-der-maaten and G. Hinton, Visualizing data using t-sne, Journal of machine learning research, vol.9, pp.2579-2605, 2008.

X. Chen, Y. Duan, R. Houthooft, J. Schulman, I. Sutskever et al., Infogan: Interpretable representation learning by information maximizing generative adversarial nets, Advances in neural information processing systems, pp.2172-2180, 2016.

M. Benmebarek, C. H. Karches, B. L. Cadilha, S. Lesch, S. Endres et al., Killing mechanisms of chimeric antigen receptor (car) t cells. International journal of molecular sciences, vol.20, p.1283, 2019.

. Shannon-l-maude, T. David, . Teachey, L. David, S. A. Porter et al., Cd19-targeted chimeric antigen receptor t-cell therapy for acute lymphoblastic leukemia, Blood, vol.125, issue.26, pp.4017-4023, 2015.

M. Sadelain, I. Brentjens, and . Riviere, The basic principles of chimeric antigen receptor (car) design, vol.3, 2013.

P. Stephen, . Hunger, and . Charles-g-mullighan, Acute lymphoblastic leukemia in children, New England Journal of Medicine, vol.373, issue.16, pp.1541-1552, 2015.

N. Shannon-l-maude, P. A. Frey, R. Shaw, . Aplenc, M. David et al., Chimeric antigen receptor t cells for sustained remissions in leukemia, New England Journal of Medicine, vol.371, issue.16, pp.1507-1517, 2014.

Z. Wang, Y. Guo, and W. Han, Current status and perspectives of chimeric antigen receptor modified t cells for cancer treatment, Protein & cell, vol.8, issue.12, pp.896-925, 2017.

. Ma-epstein, Y. M. Bg-achong, . Barr, G. Zajac, W. Henle et al., Morphological and virological investigations on cultured burkitt tumor lymphoblasts (strain raji), Journal of the National Cancer Institute, vol.37, issue.4, pp.547-559, 1966.

G. Lee, J. Oh, M. Kang, N. Her, M. Kim et al., Deephcs: Bright-field to fluorescence microscopy image conversion using deep learning for label-free high-content screening, International Conference on Medical Image Computing and Computer-Assisted Intervention, pp.335-343, 2018.

O. Ronneberger, P. Fischer, and T. Brox, U-net: Convolutional networks for biomedical image segmentation, International Conference on Medical image computing and computer-assisted intervention, pp.234-241, 2015.

J. Redmon, S. Divvala, R. Girshick, and A. Farhadi, You only look once: Unified, real-time object detection, Proceedings of the IEEE conference on computer vision and pattern recognition, pp.779-788, 2016.

M. Raghu, C. Zhang, J. Kleinberg, and S. Bengio, Transfusion: Understanding transfer learning with applications to medical imaging, 2019.

D. Matthew-d-zeiler, . Krishnan, W. Graham, R. Taylor, and . Fergus, Deconvolutional networks, 2010 IEEE Computer Society Conference on computer vision and pattern recognition, pp.2528-2535, 2010.

K. Simonyan, A. Vedaldi, and A. Zisserman, Deep inside convolutional networks: Visualising image classification models and saliency maps, 2013.

A. Mahendran and A. Vedaldi, Understanding deep image representations by inverting them, Proceedings of the IEEE conference on computer vision and pattern recognition, pp.5188-5196, 2015.

L. Gatys, A. S. Ecker, and M. Bethge, Texture synthesis using convolutional neural networks, Advances in neural information processing systems, pp.262-270, 2015.

X. Zheng, T. Chalasani, K. Ghosal, S. Lutz, A. Smolic et al., Style transfer as data augmentation, 2019.

P. Virtanen, R. Gommers, T. E. Oliphant, M. Haberland, T. Reddy et al., Paul van Mulbregt, and SciPy 1. 0 Contributors. SciPy 1.0: Fundamental Algorithms for Scientific Computing in Python, Nature Methods, vol.17, pp.261-272, 2020.

M. Bock, A. Kumar-tyagi, J. Kreft, and W. Alt, Generalized voronoi tessellation as a model of two-dimensional cell tissue dynamics, Bulletin of mathematical biology, vol.72, issue.7, pp.1696-1731, 2010.

J. Johnson, A. Alahi, and L. Fei-fei, Perceptual losses for realtime style transfer and super-resolution, European conference on computer vision, pp.694-711, 2016.

R. Hollandi, A. Szkalisity, T. Toth, E. Tasnadi, C. Molnar et al., A deep learning framework for nucleus segmentation using image style transfer. bioRxiv, p.580605, 2019.

C. Fu, S. Lee, D. J. Ho, S. Han, P. Salama et al., Three dimensional fluorescence microscopy image synthesis and segmentation, Proceedings of the IEEE Conference on Computer Vision and Pattern Recognition Workshops, pp.2221-2229, 2018.

J. Wang and Y. Yang, A context-sensitive deep learning approach for microcalcification detection in mammograms, Pattern recognition, vol.78, pp.12-22, 2018.

P. Devin, C. F. Sullivan, L. Winsnes, M. Åkesson, M. Hjelmare et al., Deep learning is combined with massivescale citizen science to improve large-scale image classification, Nature biotechnology, vol.36, issue.9, pp.820-828, 2018.

J. Whitehill and A. Ramakrishnan, Automatic classifiers as scientific instruments: One step further away from ground-truth, 2018.

F. Eduati, M. Lara, T. Mangravite, H. Wang, C. Tang et al.,

. Yang, Prediction of human population responses to toxic compounds by a collaborative competition, Nature biotechnology, vol.33, issue.9, pp.933-940, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01428019

C. Benoit-playe, V. Azencott, and . Stoven, Efficient multi-task chemogenomics for drug specificity prediction, PloS one, vol.13, issue.10, 2018.

D. Avondoglio, T. Scott, W. J. Kil, M. Sproull, J. Philip et al., High throughput evaluation of gamma-h2ax, Radiation Oncology, vol.4, issue.1, p.31, 2009.

C. Garcia-canton, A. Anadon, and C. Meredith, Assessment of the in vitro ?h2ax assay by high content screening as a novel genotoxicity test, Mutation Research/Genetic Toxicology and Environmental Mutagenesis, vol.757, issue.2, pp.158-166, 2013.

C. Chailleux, F. Aymard, P. Caron, V. Daburon, C. Courilleau et al., Quantifying dna double-strand breaks induced by site-specific endonucleases in living cells by ligation-mediated purification, Nature protocols, vol.9, issue.3, pp.517-528, 2014.

P. Naylor, M. Lae, F. Reyal, and T. Walter, Nuclei segmentation in histopathology images using deep neural networks, Proceedings of the 12th IEEE International Symposium on Biomedical Imaging (ISBI): From nano to macro, pp.933-936, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01683969

T. Walter, P. Massin, A. Erginay, R. Ordonez, C. Jeulin et al., Automatic detection of microaneurysms in color fundus images, Medical Image Analysis, vol.11, issue.6, pp.555-66, 2007.

J. Pearl, Causal inference in statistics: An overview, Statistics Surveys, vol.3, pp.96-146, 2009.

M. Ester, H. Kriegel, J. Sander, and X. Xu, A densitybased algorithm for discovering clusters in large spatial databases with noise, Kdd, vol.96, pp.226-231, 1996.

U. Von and L. , A tutorial on spectral clustering. Statistics and computing, vol.17, issue.4, pp.395-416, 2007.

H. Joe and . Ward, Hierarchical grouping to optimize an objective function, Journal of the American statistical association, vol.58, issue.301, pp.236-244, 1963.