E. Grillot, Etude de marché sur les Biomarqueurs-Définition des critères des industriels pour le développement d'un biomarqueur

A. Perez, Les révolutions de la recherche sur le cancer. Fondation ARC pour la recherche sur le cancer

N. Pottier, Pharmacogénomique des anticancéreux. Exemples de la Bléomycine et du

. Methotrexate, Sciences du Vivant, 2010.

. Hanahan, Hallmarks of Cancer: The Next Generation, vol.144, pp.646-674, 2011.

G. Maxime-ulysse, Découverte de biomarqueurs prédictifs en cancer du sein par Intégration Transcriptome-Interactome. Aix-Marseille Université, 2013.

W. Zhu, Detection of single nucleotide polymorphisms by PCR conformation-difference gel electrophoresis, J. of Biotech. Let, vol.35, pp.515-522, 2012.

A. Tsimberidou, Initiative for Molecular Profiling and Advanced Cancer Therapy and Challenges in Implementation of Precision Medicine. Personalized Medicine in a Phase 1 Clinical Trials Program, Current Problems in Cancer

,

, Guide ESMO à l'usage des patients. ESMO Médecine personnalisée

L. Cassagnes, La médecine personnalisée appliquée à l'efficacité et à la sécurité des médicaments, vol.17

D. Williams, Médecine de précision : Concrétiser la vision des résultats personnalisés en santé. Un modèle de médecine de précision

, Plan cancer, 2014.

, Quatrième rapport au président de la république, 2018.

O. Geraldine and . Coyne, Defining precision : The precision medicine initiative TRIALS NCI-MPACT and NCI-MATCH. Current Problems in Cancer

,

B. Sadick, Basket Cases: Changing the Way Cancer Is Treated. Cure. 15 avril, 2016.

F. S. Collins, M. D. Ph, H. Varmus, and M. D. , A New Initiative on Precision Medicine, N Engl J Med, vol.372, issue.9, pp.793-798, 2015.
DOI : 10.1056/nejmp1500523

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5101938/pdf

I. Curie, La médecine personnalisée, espoir de lutte contre le cancer. Note de presse, 2012.

. Dr, M. D. Mamzer, and P. , Les nouveaux enjeux éthiques de la médecine prédictive et personnalisée, 2012.

X. Guchet, « Le patient « actionnable » de la médecine personnalisée, vol.29, pp.37-51, 2014.

W. Boder, Bienvenue dans l'ère de la médecine personnalisée, 1921.

P. Marquet, Recherche translationnelle : médecine personnalisée, médecine de précision, thérapies ciblées : marketing ou science ? Thérapie, Janvier-Février, vol.70, issue.1, pp.1-10, 2015.

V. Von-manstein, Tumor cell resistance against targeted therapeutics: the density of cultured glioma tumor cells enhances Stat3 activity and offers protection against the tyrosine kinase inhibitor canertinib, Med. Chem. Commun, vol.8, pp.96-102, 2017.

P. C. Nowell, The clonal evolution of tumor cell populations, Science, vol.194, pp.23-31, 1976.

L. G. Ahronian, Strategies for monitoring and combating resistance to combination kinase inhibitors for cancer therapy, Genome Med, vol.9, p.37, 2017.

R. A. Burrell, Tumour heterogeneity and the evolution of polyclonal drug resistance

, Mol Oncol, vol.8, issue.6, pp.1095-111, 2014.

M. Tellez-gabriel, Tumour Heterogeneity: The Key Advantages of Single-Cell Analysis, International Journal of Molecular Sciences, vol.17, pp.2142-2142, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01466086

,

N. Cherdyntseva, Circulating tumor cells in breast cancer: functional heterogeneity, pathogenetic and clinical aspects, Experimental Oncology, vol.39, issue.1

A. Brouwer, Evaluation and consequences of heterogeneity in the circulating tumor cell compartment, Oncotarget, vol.7, pp.48625-48643, 2016.

N. Mcgranahan, Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future, Cell, vol.168, pp.613-628, 2017.

M. Jamal-hanjani, Translational Implications of Tumor Heterogeneity, Clin Cancer Res, vol.21, issue.6, 2015.

J. De-lartigue, Tumor heterogeneity: a central foe in the war on cancer The JCSO, vol.16, pp.167-174, 2018.

N. Amirouchene-angelozzi, Tumor Evolution as a Therapeutic Target, 2017.

V. D. Jonsson, Novel computational method for predicting polytherapy switching strategies to overcome tumor heterogeneity and evolution, Scientific Reports, vol.7, p.44206, 2017.

L. Lupini, High-sensitivity assay for monitoring ESR1 mutations in circulating cellfree DNA of breast cancer patients receiving endocrine therapy, Sci Rep, vol.8, p.4371, 2018.

M. Gerlinger, How Darwinian models inform therapeutic failure initiated by clonal heterogeneity in cancer medicine, Br J Cancer, vol.103, issue.8, pp.1139-1143, 2010.

P. Ojamies, Clonal evolution and heterogeneity of cancer in the context of individualized medicine, 2018.

D. H. Roukos, Spatiotemporal diversification of intrapatient genomic clones and early drug development concepts realize the roadmap of precision cancer medicine, Drug Discovery Today, vol.22, issue.8, pp.1148-1164, 2017.

S. H. Youssef and . Abdelaziz, Somatic Mutation Detection in Leukemia-Derived Circulating DNA: Utility in

, Monitoring Clonal Dynamics and Disease Response in Pediatric Acute Lymphoblastic Leukemia, Theses and Dissertations, 2018.

D. L. Ellsworth, Single-cell sequencing and tumorigenesis: improved understanding of tumor evolution and metastasis, Clin Transl Med, vol.6, issue.1, p.15, 2017.

V. Almendro, A. Marusyk, and K. Polyak, Cellular Heterogeneity and Molecular Evolution in Cancer, Annual Review of Pathology: Mechanisms of Disease, vol.8, issue.1, pp.277-302, 2013.

L. William and . Hwang, The promise of circulating tumor cells for precision cancer therapy Biomark Med, vol.10, pp.1269-1285, 2016.

D. S. Micalizzi, A conduit to metastasis: circulating tumor cell biology, Genes Dev

, Sep, vol.15, issue.18, pp.1827-1840

A. Markiewicz, A. ?aczek, and J. , The Landscape of Circulating Tumor Cell Research in the Context of Epithelial-Mesenchymal Transition, Pathobiology, vol.84, pp.264-283, 2017.

Z. Piotrowska, Heterogeneity and Coexistence of T790M and T790 Wild-Type Resistant Subclones Drive Mixed Response to Third-Generation Epidermal Growth Factor Receptor Inhibitors in Lung Cancer, JCO Precis Oncol, 2018.

R. Wang, Cell-free circulating tumor DNA analysis for breast cancer and its clinical utilization as a biomarker, Oncotarget, vol.8, issue.43, pp.75742-75755, 2017.

B. Liu, Y. Yang, Z. Yi, X. Guan, and F. Ma, The application of estrogen receptor-1 mutations' detection through circulating tumor dna in breast cancer, Cancer Transl Med, vol.3, pp.46-52, 2017.

H. E. Sabaawy, Genetic Heterogeneity and Clonal Evolution of Tumor Cells and their Impact on Precision Cancer Medicine, J Leuk (Los Angel), vol.1, p.124, 2013.

G. Siravegna, Integrating liquid biopsies into the management of cancer, Nat Rev Clin Oncol, vol.14, issue.9, pp.531-548, 2017.

J. A. Gallaher, Spatial Heterogeneity and Evolutionary Dynamics Modulate Time to Recurrence in Continuous and Adaptive Cancer Therapies, Cancer Research, vol.78, issue.8, pp.2127-2139, 2018.

K. Asi?, , 2016.

/. Oncology and . Hematology, , vol.97, pp.178-196

C. M. Stewart, The value of cell-free DNA for molecular pathology, J Pathol, 2018.

, , vol.244, pp.616-627, 2018.

T. Venesio, Liquid Biopsies for Monitoring Temporal Genomic Heterogeneity in Breast and Colon Cancers, Pathobiology, vol.85, issue.1-2, pp.146-154, 2017.

F. Massari, Circulating tumor cells in genitourinary tumors, Ther Adv Urol, 2018.

J. Lack, Circulating tumor cells capture disease evolution in advanced prostate cancer, Journal of Translational Medicine201715, p.44

V. Velcheti, Non-invasive diagnostic platforms in management of nonsmall cell lung cancer: opportunities and challenges, vol.5, 2017.

J. L. Van-etten, Clonal origin and spread of metastatic prostate cancer, Endocr Relat Cancer, vol.23, issue.4, pp.207-217, 2016.

P. Mandel and P. Metais, Les acides nucléiques du plasma sanguin chez l'homme

C. R. , Seances Soc. Biol. Fil, vol.142, pp.241-243, 1948.

S. A. Leon, B. Shapiro, D. M. Sklaroff, and M. J. Yaros, Free DNA in the serum of cancer patients and the effect of therapy, Cancer Res, vol.37, pp.646-650, 1977.

M. Stroun, Neoplastic characteristics of the DNA found in the plasma of cancer patients, Oncology, vol.46, pp.318-322, 1989.

G. D. Sorenson, D. M. Pribish, and F. H. Valone, Soluble normal and mutated DNA sequences from single-copy genes in human blood, Cancer Epidemiol Biomarkers Prev, 1994.

D. A. Landau, Clonal evolution in hematological malignancies and therapeutic implications, Leukemia, vol.28, issue.1, pp.34-43, 2014.

M. Fleischhacker and B. Schmidt, Circulating nucleic acids (CNAs) and cancer-a survey, Biochim Biophys Acta, vol.1775, issue.1, pp.181-232, 2007.

M. Murtaza, Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA, Nature, vol.497, pp.108-112, 2013.

S. Misale, Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer, Nature, vol.486, pp.532-536, 2012.

L. A. Diaz, The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers, Nature, vol.486, pp.537-540, 2012.

R. Palmirotta, Liquid biopsy of cancer: a multimodal diagnostic tool in clinical oncology, Ther Adv Med Oncol, vol.10, p.1758835918794630, 2018.

S. Pakkala, Personalized therapy for lung cancer: Striking a moving target, JCI Insight

, Aug, vol.9, issue.15

M. Murtaza, Multifocal clonal evolution characterized using circulating tumour DNA in a case of metastatic breast cancer, Nat Commun, vol.6, p.8760, 2015.

J. D. Cohen, Detection and localization of surgically resectable cancers with a multianalyte blood test, Science, vol.359, issue.6378, pp.926-930, 2018.

I. Kinde, Evaluation of DNA from the Papanicolaou Test to Detect Ovarian and Endometrial Cancers, Sci Transl Med, vol.5, issue.167, pp.167-171, 2009.

S. J. Dawson, Analysis of circulating tumor DNA to monitor metastatic breast cancer

, N Engl J Med, vol.368, issue.13, pp.1199-209, 2013.

A. R. Thierry, Origin and quantification of circulating DNA in mice with human colorectal cancer xenografts, Nucleic Acids Res, vol.38, issue.18, pp.6159-75, 2010.

C. Bettegowda, Detection of circulating tumor DNA in early-and late-stage human malignancies, Sci Transl Med, vol.6, issue.224, pp.224-248, 2014.

A. A. Kamat, Circulating cell-free DNA: a novel biomarker for response to therapy in ovarian carcinoma, Cancer Biol Ther, vol.5, issue.10, pp.1369-74, 2006.

T. Lecomte, Detection of free-circulating tumor-associated DNA in plasma of colorectal cancer patients and its association with prognosis, Int J Cancer, vol.100, issue.5, pp.542-550, 2002.

G. Sozzi, Quantification of free circulating DNA as a diagnostic marker in lung cancer, J Clin Oncol, vol.21, issue.21, pp.3902-3910, 2003.

E. Crowley, D. Nicolantonio, F. Loupakis, and F. , Liquid biopsy: monitoring cancergenetics in the blood, Nat Rev Clin Oncol, vol.10, pp.472-84, 2013.

M. J. Higgins, Detection of tumor PIK3CA status in metastatic breast cancer using peripheral blood, Clin Cancer Res, vol.18, issue.12, pp.3462-3471, 2012.

S. Mithraprabhu, Circulating tumour DNA analysis in multiple myeloma

, Oncotarget, vol.8, issue.53, pp.90610-90611, 2017.

. Lebofsky, Circulating tumor DNA as a non-invasive substitute to metastasis biopsy for tumor genotyping and personalized medicine in a prospective trial across all tumor types

, Mol. Oncol, vol.9, pp.783-790, 2015.

T. M. Butler, Exome sequencing of cell-free DNA from metastatic cancer patients identifies clinically actionable mutations distinct from primary disease, PLoS One, vol.10, p.136407, 2015.

A. Esposito, Liquid biopsies for solid tumors: Understanding tumor heterogeneity and real time monitoring of early resistance to targeted therapies, Pharmacol Ther, vol.157, pp.120-124, 2016.

M. Jamal-hanjani, Detection of ubiquitous and heterogeneous mutations in cell-free DNA from patients with early-stage non-small-cell lung cancer, Ann Oncol, vol.27, pp.862-869, 2016.

C. Abbosh, Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution, Nature, vol.545, pp.446-51, 2017.

M. Jamal-hanjani, Tracking the evolution of non-small-cell lung cancer, N Engl J Med, vol.376, pp.2109-2130, 2017.

D. Zheng, X. Ye, and M. Z. Zhang, Plasma EGFR T790M ctDNA status is associated with clinical outcome in advanced NSCLC patients with acquired EGFR-TKI resistance, Sci Rep, vol.6, issue.1, p.20913, 2016.

A. R. Thierry, E. Messaoudi, S. Mollevi, and C. , Clinical utility of circulating DNA analysis for rapid detection of actionable mutations to select metastatic colorectal patients for antiEGFR treatment, Ann Oncol, vol.28, issue.9, pp.2149-2159, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01682168

D. Bernicker, E. Li, and T. , Genomic profiling of circulating tumor DNA (ctDNA) from patients (pts) with advanced non-small cell lung cancer (NSCLC), J Clin Oncol, vol.35, p.9025, 2017.

M. C. Schwaederlé, S. P. Patel, and H. Husain, Utility of genomic assessment of bloodderived circulating tumor DNA (ctDNA) in patients with advanced lung adenocarcinoma

, Clin Cancer Res, vol.23, pp.5101-5112, 2017.

M. Qiu, J. Wang, and Y. Xu, Circulating tumor DNA is effective for the detection of EGFR mutation in non-small cell lung cancer: a meta-analysis, Cancer Epidemiol Biomarkers Prev, vol.24, pp.206-218, 2015.

E. Takai, Clinical utility of circulating tumor DNA for molecular assessment in pancreatic cancer, Sci. Rep, vol.5, p.18425, 2015.

T. Takeshita, Clinical significance of monitoring ESR1 mutations in circulating cell-free DNA in estrogen receptor positive breast cancer patients, Oncotarget, vol.7, issue.22, pp.32504-32522, 2016.

D. H. Liang, Cell-free DNA as a molecular tool for monitoring disease progression and response to therapy in breast cancer patients, Breast Cancer Res Treat, vol.155, issue.1, pp.139-188, 2016.

N. Normanno, M. G. Denis, and K. S. Thress, Guide to detecting epidermal growth factor receptor (EGFR) mutations in ctDNA of patients with advanced non-small-cell lung cancer, Oncotarget, vol.8, pp.12501-12516, 2017.

A. Bardelli, K. L. Pantel, . Biopsies, J. M. Garcia, V. Garcia et al., Extracellular tumor DNA in plasma and overall survival in breast cancer patients, Genes Chromosomes Cancer, vol.31, pp.692-701, 2006.

M. S. Kopreski, F. A. Benko, and D. J. Borys, Somatic mutation screening: identification of individuals harboring K-ras mutations with the use of plasma DNA, J Natl Cancer Inst, vol.92, pp.918-923, 2000.

F. Dianxu, Z. Shengdao, and H. Tianquan, A prospective study of detection of pancreatic carcinoma by combined plasma K-ras mutations and serum CA19-9 analysis, Pancreas, vol.25, pp.336-341, 2002.

A. Castells, P. Puig, and J. Mora, K-ras mutations in DNA extracted from the plasma of patients with pancreatic carcinoma: diagnostic utility and prognostic significance, J Clin Oncol, vol.17, pp.578-584, 1999.

S. Maheswaran, Detection of mutations in EGFR in circulating lung-cancer cells

, N Engl J Med, vol.359, pp.366-377, 2008.

M. V. Seiden, Detection of circulating tumor cells in men with localized prostate cancer, J Clin Oncol, vol.12, pp.2634-2639, 1994.

F. Bigot, N. Pecuchet, and B. Blanchet, Early TKI-pharmokinetics and circulating tumor DNA (ctDNA) to predict outcome in patients with EGFR-mutated non-small cell lung cancer (NSCLC), J Clin Oncol, vol.35, p.11544, 2017.

D. J. Merriott, Circulating Tumor DNA Quantitation for Early Response Assessment of Immune Checkpoint Inhibitors for Lung Cancer, International Journal of Radiation Oncology*Biology*Physics, vol.99, issue.2, pp.20-21, 2017.

. Frattini, Reproducibility of a semiquantitative measurement of circulating DNA in plasma from neoplastic patients, J Clin Oncol, vol.23, pp.3163-3164, 2005.

F. Diehl, Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients, Gastroenterology, vol.135, pp.489-498, 2008.

M. Yanagita, A. J. Redig, C. P. Paweletz, S. E. Dahlberg, A. O'connell et al., A prospective evaluation of circulating tumor cells and cell-free DNA in EGFR mutant nonsmall cell lung cancer patients treated with erlotinib on a phase II trial, Clin Cancer Res, vol.22, issue.24, pp.6010-6030, 2016.

M. Sausen, Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients, Nat. Commun, vol.6, p.7686, 2015.

E. Heitzer, Tumor-associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencing, Genome Med, vol.5, p.90, 2013.

L. A. Diaz and . Jr, Insights into therapeutic resistance from whole-genome analyses of circulating tumor DNA, Oncotarget, vol.4, pp.1856-1857, 2013.

J. Tie, Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer, Ann. Oncol, vol.26, pp.1715-1722, 2015.

J. S. Frenel, Serial next-generation sequencing of circulating cell-free DNA evaluating tumor clone response to molecularly targeted drug administration, Clin. Cancer Res, vol.21, pp.4586-4596, 2015.

G. R. Oxnard, C. P. Paweletz, Y. Kuang, S. L. Mach, A. O'connell et al., Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA, Clin Cancer Res, vol.20, issue.6, pp.1698-705, 2014.

I. Garcia-murillas, Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer, Sci Transl Med, vol.7, pp.302-133, 2015.

I. Garcia-murillas, G. Schiavon, and B. Weigelt, Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer, Sci Transl Med, vol.7, pp.302-133, 2015.

M. G. Krebs, Molecular analysis of circulating tumour cells-biology and biomarkers, Nat. Rev. Clin. Oncol, vol.11, pp.129-144, 2014.

T. Reinert, Analysis of circulating tumour DNA to monitor disease burden following colorectal cancer surgery, Gut, vol.65, pp.625-634, 2015.

K. Kim, Circulating cell-free DNA as a promising biomarker in patients with gastric cancer: diagnostic validity and significant reduction of cfDNA after surgical resection

, Ann. Surg. Treat. Res, vol.86, pp.136-142, 2014.

A. Chaudhuri, Analysis of circulating tumor DNA in localized lung cancer for detection of molecular residual disease and personalization of adjuvant strategies, Journal of Clinical Oncology, vol.35, pp.15-8519, 2017.

J. Tie, Y. Wang, C. Tomasetti, L. Li, S. Springer et al., Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer, Sci Transl Med, vol.8, pp.346-92, 2016.

S. Mithraprabhu, Circulating tumour DNA analysis demonstrates spatial mutational heterogeneity that coincides with disease relapse in myeloma, Leukemia, vol.31, issue.8, pp.1695-1705, 2017.

G. Siravegna, B. Mussolin, M. Buscarino, G. Corti, A. Cassingena et al., Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients

, Nat Med, vol.21, issue.7, pp.795-801, 2015.

P. A. Jänne, J. C. Yang, and D. W. Kim, AZD9291 in EGFR inhibitor-resistant non-smallcell lung cancer, N Engl J Med, vol.372, pp.1689-99, 2015.

J. J. Chabon, Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients, Nat Commun, vol.7, p.11815, 2016.

N. Karachaliou, Association of EGFR L858R mutation in circulating free DNA with survival in the EURTAC trial, JAMA Oncol, vol.1, pp.149-157, 2015.

X. Qian, J. Liu, and Y. Sun, Circulating cell-free DNA has a high degree of specificity to detect exon 19 deletions and the singlepoint substitution mutation L858R in non-small cell lung cancer, Oncotarget, vol.7, pp.29154-29165, 2016.

K. S. Thress, C. P. Paweletz, and E. Felip, Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M, Nat Med, vol.21, pp.560-562, 2015.

M. G. Kris, B. E. Johnson, and L. D. Berry, Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs, JAMA, vol.311, pp.1998-2006, 2014.

G. R. Oxnard, Association between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non-small-cell lung cancer, J. Clin. Oncol, vol.34, pp.3375-3382, 2016.

J. Y. Douillard, G. Ostoros, and M. Cobo, First-line gefitinib in Caucasian EGFR mutation-positive NSCLC patients: a phase-IV, open-label, single-arm study, Br J Cancer, vol.110, pp.55-62, 2014.

, PMA P150047: FDA summary of safety and effectiveness data page 1 summary of safety and effectiveness data (SSED), US Food and Drug Administration, 2016.

Y. L. Wu, C. Zhou, and C. K. Liam, First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: analyses from the phase III, randomized, open-label, ENSURE study, Ann Oncol, vol.26, pp.1883-189, 2015.

, US Food and Drug Administration. cobas EGFR Mutation Test v2, 2016.

T. R. Ashworth, A case of cancer in which cells similar to those in the tumours were seen in the blood after death, Aust. Med. J, vol.14, pp.146-147, 1869.

E. Heitzer, Complex tumor genomes inferred from single circulating tumor cells by array-CGH and next-generation sequencing, Cancer Res, vol.73, pp.2965-2975, 2013.

M. Yu, RNA sequencing of pancreatic circulating tumour cells implicates WNT signalling in metastasis, Nature, vol.487, pp.510-513, 2012.

M. Heidary, The dynamic range of circulating tumor DNA in metastatic breast cancer, Breast Cancer Res, vol.16, p.421, 2014.

X. Ni, Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients, Proc Natl Acad Sci U S A, vol.110, pp.21083-21088, 2013.

J. G. Lohr, Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer, Nat Biotechnol, vol.32, pp.479-484, 2014.

S. Meng, D. Tripathy, E. P. Frenkel, S. Shete, E. Z. Naftalis et al., Circulating tumor cells in patients with breast cancer dormancy, Clin Cancer Res, vol.10, pp.8152-8162, 2004.

S. Nagrath, Isolation of rare circulating tumour cells in cancer patients by microchip technology, Nature, vol.450, pp.1235-1239, 2007.

W. J. Allard, Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases, Clin Cancer Res, vol.10, pp.6897-6904, 2004.

I. Baccelli, Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay, Nat Biotechnol, vol.31, pp.539-544, 2013.

N. Aceto, Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis, Cell, vol.158, pp.1110-1122, 2014.

Y. Kienast, Real-time imaging reveals the single steps of brain metastasis formation, Nat Med, vol.16, pp.116-122, 2010.

S. L. Stott, Isolation and characterization of circulating tumor cells from patients with localized and metastatic prostate cancer, Sci Transl Med, vol.2, pp.25-48, 2010.

S. Gkountela, B. Szczerba, and C. Donato, Recent advances in the biology of human circulating tumour cells and metastasis, ESMO Open, vol.1, p.78, 2016.

E. Sinkala, Profiling protein expression in circulating tumour cells using microfluidic western blotting, Nature Communications, vol.8, p.14622, 2017.

T. Wu, Clinical Applications of Circulating Tumor Cells in Pharmacotherapy: Challenges and Perspectives, Molecular Pharmacology, vol.92, issue.3, pp.232-239, 2017.

L. Zhang and ;. B. , Circulating tumor cells predict survival in early average-to-high risk breast cancer patients, J. Natl Cancer Inst, vol.18, p.66, 2012.

M. Cristofanilli, A. Stopeck, and J. Matera, Circulating tumor cells, disease progression, and survival in metastatic breast cancer, N. Engl. J. Med, vol.351, issue.8, pp.781-791, 2004.

M. Giuliano, Circulating tumor cells as prognostic and predictive markers in metastatic breast cancer patients receiving first-line systemic treatment, Breast Cancer Res, vol.13, 2011.

B. Rack, Circulating tumor cells predict survival in early average-to-high risk breast cancer patients, J Natl Cancer Inst, vol.106, 2014.

Y. Lu, P. Wang, X. Wang, J. Peng, Y. Zhu et al., The significant prognostic value of circulating tumor cells in triple-negative breast cancer: a meta-analysis, Oncotarget, 2016.

Y. Shiomi-mouri, J. Kousaka, and T. Ando, Clinical significance of circulating tumor cells (CTCs) with respect to optimal cut-off value and tumor markers in advanced/metastatic breast cancer, Breast Cancer, vol.23, issue.1, pp.120-127, 2016.

Q. Lv, L. Gong, and T. Zhang, Prognostic value of circulating tumor cells in metastatic breast cancer: a systemic review and meta-analysis, Clin. Transl. Oncol, vol.18, issue.3, pp.322-330, 2016.

A. Goldkorn, B. Ely, and D. I. Quinn, Circulating tumor cell counts are prognostic of overall survival in SWOG S0421: a phase III trial of docetaxel with or without atrasentan for metastatic castration-resistant prostate cancer, J Clin Oncol, vol.32, pp.1136-1142, 2014.

D. Bono, J. S. Scher, H. I. Montgomery, and R. B. , Circulating tumor cells predict survival benefi t from treatment in metastatic castration-resistant prostate cancer, Clin Cancer Res, vol.14, pp.6302-6311, 2008.

H. I. Scher, X. Jia, D. Bono, and J. S. , Circulating tumour cells as prognostic markers in progressive, castration-resistant prostate cancer: a reanalysis of IMMC38 trial data, Lancet Oncol, vol.10, pp.233-239, 2009.

F. Nolé, E. Munzone, and L. Zorzino, Variation of circulating tumor cell levels during treatment of metastatic breast cancer: prognostic and therapeutic implications, Ann. Oncol, vol.19, issue.5, pp.891-897, 2008.

D. C. Danila, G. Heller, and G. A. Gignac, Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer, Clin Cancer Res, vol.13, pp.7053-7058, 2007.

J. G. Moreno, M. C. Miller, S. Gross, W. J. Allard, L. G. Gomella et al., Circulating tumor cells predict survival in patients with metastatic prostate cancer, Urology, vol.65, issue.4, pp.713-718, 2005.

M. Fleisher, D. C. Danila, and K. Fizazi, Circulating tumor cell (CTC) enumeration in men with metastatic castration-resistant prostate cancer (mCRPC) treated with enzalutamide post-chemotherapy (phase 3 AFFIRM study), J Clin Oncol, p.5035, 2015.

M. G. Krebs and L. Lancashire, Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer, J Clin Oncol, vol.30, pp.525-557, 2012.

Y. Li, J. Gong, and Q. Zhang, Dynamic monitoring of circulating tumour cells to evaluate therapeutic efficacy in advanced gastric cancer, Br. J. Cancer, vol.114, issue.2, pp.138-145, 2016.

S. J. Cohen, C. Punt, and N. Iannotti, Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer, J. Clin. Oncol, vol.26, pp.3213-3221, 2008.

M. G. Krebs, R. Sloane, and L. Priest, Evaluation and prognostic signifi cance of circulating tumor cells in patients with non-small-cell lung cancer, J Clin Oncol, vol.29, issue.12, pp.1556-63, 2011.

C. Bayarri-lara, Circulating tumor cells identify early recurrence in patients with non-small-cell lung cancer undergoing radical resection, PLoS ONE, vol.11, issue.2, p.148659, 2016.

E. A. Punnoose, S. Atwal, and W. Liu, Evaluation of circulating tumor cells and circulating tumor DNA in non-small-cell lung cancer: association with clinical endpoints in a Phase II clinical trial of pertuzumab and erlotinib, Clin. Cancer Res, vol.18, issue.8, p.2391, 2012.

J. Wang, J. Huang, and K. Wang, Prognostic significance of circulating tumor cells in non-small-cell lung cancer patients: a meta-analysis, PLoS One, vol.8, p.78070, 2013.

A. Matikas, K. N. Syrigos, and S. Agelaki, Circulating Biomarkers in Non-Small-Cell Lung Cancer: Current Status and Future Challenges, Clin Lung Cancer, vol.17, pp.507-523, 2016.

M. Hashimoto, F. Tanaka, and K. Yoneda, Significant increase in circulating tumour cells in pulmonary venous blood during surgical manipulation in patients with primary lung cancer, Interact Cardiovasc Thorac Surg, vol.18, pp.775-83, 2014.

V. Hofman, C. Bonnetaud, and M. I. Ilie, Preoperative circulating tumor cell detection using the isolation by size of epithelial tumor cell method for patients with lung cancer is a new prognostic biomarker, Clin Cancer Res, vol.17, pp.827-862, 2011.

W. Sienel, R. Seen-hibler, and W. Mutschler, Tumour cells in the tumour draining vein of patients with non-small cell lung cancer: detection rate and clinical significance, Eur J Cardiothorac Surg, vol.23, pp.451-457, 2003.

J. M. Hou, M. G. Krebs, and L. Lancashire, Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer, J. Clin. Oncol, vol.30, issue.5, pp.525-532, 2012.

Y. Cheng, X. Liu, and Y. Fan, Circulating tumor cell counts/change for outcome prediction in patients with extensive-stage small-cell-lung cancer, Future Oncol, vol.12, issue.6, pp.789-799, 2016.

P. Gazzaniga, Circulating tumor cells detection has independent prognostic impact in high-risk non-muscle invasive bladder cancer, Int J Cancer, vol.135, pp.1978-1982, 2014.

K. Schulze, Presence of EpCAM-positive circulating tumor cells as biomarker for systemic disease strongly correlates to survival in patients with hepatocellular carcinoma, Int J Cancer, vol.133, pp.2165-2171, 2013.

Y. K. Vashist, Disseminated tumor cells in bone marrow and the natural course of resected esophageal cancer, Ann Surg, vol.255, pp.1105-1112, 2012.

A. C. Nichols, Detection of circulating tumor cells in advanced head and neck cancer using the CellSearch system, Head Neck, vol.34, pp.1440-1444, 2012.

L. Han, Prognostic value of circulating tumor cells in patients with pancreatic cancer: a meta-analysis, Tumour Biol, vol.35, pp.2473-2480, 2014.

C. Rao, Circulating melanoma cells and survival in metastatic melanoma, Int J Oncol, vol.38, pp.755-760, 2011.

J. Sastre, Circulating tumor cell count is a prognostic factor in metastatic colorectal cancer patients receiving first-line chemotherapy plus bevacizumab: a Spanish Cooperative Group for the Treatment of Digestive Tumors study, Oncologist, vol.17, pp.947-955, 2012.

J. Tol, Circulating tumour cells early predict progression-free and overall survival in advanced colorectal cancer patients treated with chemotherapy and targeted agents, Ann Oncol, vol.21, pp.1006-1012, 2010.

M. C. Miller, G. V. Doyle, and L. Terstappen, Significance of circulating tumor cells detected by the CellSearch system in patients with metastatic breast colorectal and prostate cancer, J. Oncol, p.617421, 2010.

D. F. Hayes, M. Cristofanilli, G. T. Budd, M. J. Ellis, A. Stopeck et al., Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival, Clin Cancer Res, vol.12, pp.4218-4224, 2006.

S. Nakamura, Multi-center study evaluating circulating tumor cells as a surrogate for response to treatment and overall survival in metastatic breast cancer, Breast Cancer, vol.17, pp.199-204, 2010.

M. C. Liu, Circulating tumor cells: a useful predictor of treatment efficacy in metastatic breast cancer, J Clin Oncol, vol.27, pp.5153-5159, 2009.

G. T. Budd, Circulating tumor cells versus imaging-predicting overall survival in metastatic breast cancer, Clin Cancer Res, vol.12, pp.6403-6409, 2006.

M. Cristofanilli, Circulating tumor cells: a novel prognostic factor for newly diagnosed metastatic breast cancer, J Clin Oncol, vol.23, pp.1420-1430, 2005.

D. Olmos, Circulating tumour cell (CTC) counts as intermediate end points in castration-resistant prostate cancer (CRPC): a single-centre experience, Ann Oncol, vol.20, pp.27-33, 2009.

G. Attard, Selective inhibition of CYP17 with abiraterone acetate is highly active in the treatment of castration-resistant prostate cancer, J Clin Oncol, vol.27, pp.3742-3748, 2009.

E. S. Lianidou, Circulating tumor cells in breast cancer: detection systems, molecular characterization, and future challenges, Clin Chem, vol.57, pp.1242-1255, 2011.

F. C. Bidard, Circulating tumor cells in breast cancer, Mol Oncol, vol.10, pp.418-430, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-00749450

J. Y. Pierga, D. Hajage, and T. Bachelot, High independent prognostic and predictive

J. Lin, Polyclonality of BRAF mutations in primary melanoma and the selection of mutant alleles during progression, Br J Cancer, vol.104, pp.464-468, 2011.

A. L. Reid and L. Et, Detection of BRAF-V600E and V600K in melanoma circulating tumour cells by droplet digital PCR, Clin Biochem, vol.48, pp.999-1002, 2015.

J. Atkins and M. B. , Treatment of BRAF-mutant melanoma: the role of vemurafenib and other therapies, Clin Pharmacol Ther, vol.95, pp.24-31, 2014.

F. Breitenbuecher, Development of a highly sensitive and specific method for detection of circulating tumor cells harboring somatic mutations in non-small-cell lung cancer patients, PLoS One, vol.9, p.85350, 2014.

T. K. Sundaresan, Detection of T790M, the acquired resistance EGFR mutation, by tumor biopsy versus noninvasive blood-based analyses, Clin Cancer Res, vol.22, pp.1103-1113, 2016.

E. Pailler, Detection of circulating tumor cells harboring a unique ALK rearrangement in ALK-positive non-small-cell lung cancer, J Clin Oncol, vol.31, pp.2273-81, 2013.

X. Ni, Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients, Proc Natl Acad Sci, vol.110, pp.21083-21091, 2013.

A. Marchetti, Assessment of EGFR mutations in circulating tumor cell preparations from NSCLC patients by next generation sequencing: toward a real-time liquid biopsy for treatment, PLoS ONE, vol.9, p.103883, 2014.

M. Kuwano, Overcoming drug resistance to receptor tyrosine kinase inhibitors: learning from lung cancer, Pharmacol Ther, vol.161, pp.97-110, 2016.

H. Schneck, Analysing the mutational status of PIK3CA in circulating tumor cells from metastatic breast cancer patients, Mol Oncol, vol.7, pp.976-986, 2013.

S. R. Johnston, Enhancing endocrine therapy for hormone receptor-positive advanced breast cancer: cotargeting signaling pathways, J Natl Cancer Inst, vol.107, p.10, 2015.

W. De-roock, D. Vriendt, V. Normanno, N. Ciardiello, F. et al., mutations: implications for targeted therapies in metastatic colorectal cancer, Lancet Oncol, vol.12, pp.594-603, 2011.

S. Meng, D. Tripathy, and S. Shete, HER2 gene amplification can be acquired as breast cancer progresses, Proc Natl Acad Sci U S A, vol.101, pp.9393-9401, 2004.

A. Hernández-blanquisett, Current and emerging therapies of HER2positive metastatic breast cancer, Breast, vol.29, pp.170-177, 2016.

N. H. Turner, D. Leo, and A. , HER2 discordance between primary and metastatic breast cancer: assessing the clinical impact, Cancer Treat Rev, vol.39, pp.947-957, 2013.

K. Pachmann, O. Camara, and T. Kroll, Efficacy control of therapy using circulating epithelial tumor cells (CETC) as "liquid biopsy": Trastuzumab in HER2/neu-positive breast carcinoma, J Cancer Res Clin Oncol, vol.137, pp.1317-1344, 2011.

M. Ignatiadis, J. Y. Pierga, and M. Campion, CTCs and HER2-positive CTCs detection by CellSearch® in non-metastatic breast cancer: an international ring study to assess interreader variability, Cancer Res, vol.71, pp.4-07, 2011.

A. Prat and J. Baselga, Dual human epidermal growth factor receptor 2 (HER2) blockade and hormonal therapy for the treatment of primary HER2-positive breast cancer: one more step toward chemotherapy-free therapy, J Clin Oncol, vol.31, pp.1703-1709, 2013.

V. Georgoulias, V. Bozionelou, and S. Agelaki, Trastuzumab decreases the incidence of clinical relapses in patients with early breast cancer presenting chemotherapy-resistant CK19mRNA-positive circulating tumor cells: results of a randomized phase II study, Ann Oncol, vol.23, pp.1744-50, 2012.

N. Xenidis, Clinical relevance of circulating CK-19 mRNA-positive cells detected during the adjuvant tamoxifen treatment in patients with early breast cancer, Ann Oncol, vol.18, pp.1623-1631, 2007.

A. Pantel and K. , Circulating tumor cells: liquid biopsy of cancer, 2013.

, Clin Chem, vol.59, pp.110-118

G. Heller, TMPRSS2-ERG status in circulating tumor cells as a predictive biomarker of sensitivity in castration-resistant prostate cancer patients treated with abiraterone acetate, Eur Urol, vol.60, pp.897-904, 2011.

J. D. Kuhlmann, ERCC1-positive circulating tumor cells in the blood of ovarian cancer patients as a predictive biomarker for platinum resistance, Clin Chem, vol.60, pp.1282-1289, 2014.

E. S. Antonarakis, Androgen receptor splice variant 7 and efficacy of taxane chemotherapy in patients with metastatic castration-resistant prostate cancer, JAMA Oncol, vol.1, pp.582-591, 2015.

W. Onstenk, Efficacy of cabazitaxel in castration-resistant prostate cancer is independent of the presence of AR-V7 in circulating tumor cells, Eur Urol, vol.68, pp.939-945, 2015.

E. S. Antonarakis, resistance to enzalutamide and abiraterone in prostate cancer, N Engl J Med, vol.371, pp.1028-1038

M. Jamal-hanjani, Tracking genomic cancer evolution for precision medicine: The lung TRACERx study, PLoS Biol, vol.12, p.1001906, 2014.

G. Middleton, The National Lung Matrix Trial: Translating the biology of stratification in advanced non-small-cell lung cancer, Ann. Oncol, vol.26, pp.2464-2469, 2015.

E. Pailler, N. Auger, C. R. Lindsay, P. Vielh, A. Islas-morris-hernandez et al., High level of chromosomal instability in circulating tumor cells of ROS1-rearranged non-small-cell lung cancer, Ann. Oncol, vol.26, pp.1408-1415, 2015.

B. Polzer, Molecular profiling of single circulating tumor cells with diagnostic intention, EMBO Mol. Med, vol.6, pp.1371-1386, 2014.

M. Suhaimi, N. A. Foong, Y. M. Lee, D. Y. Phyo, W. M. Cima et al., Non-invasive sensitive detection of KRAS and BRAF mutation in circulating tumor cells of colorectal cancer patients

, Oncol, vol.9, pp.850-860, 2015.

B. Mostert, KRAS and BRAF mutation status in circulating colorectal tumor cells and their correlation with primary and metastatic tumor tissue, Int. J. Cancer, vol.133, pp.130-141, 2013.

D. T. Miyamoto, R. J. Lee, S. L. Stott, D. T. Ting, B. S. Wittner et al., Androgen receptor signaling in circulating tumor cells as a marker of hormonally responsive prostate cancer, Cancer Discov, vol.2, pp.995-1003, 2012.

M. Yu, Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition, Science, vol.339, pp.580-584, 2013.

A. E. Dago, Rapid phenotypic and genomic change in response to therapeutic pressure in prostate cancer inferred by high content analysis of single circulating tumor cells, PLoS One, vol.9, p.101777, 2014.

T. Fojo, S. Mailankody, and A. Lo, Unintended consequences of expensive cancer therapeutics-the pursuit of marginal indications and a me-too mentality that stifles innovation and creativity: the John Conley Lecture, JAMA Otolaryngol. Head Neck Surg, vol.140, pp.1225-1236, 2014.

K. T. , Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations, N. Engl. J. Med, vol.367, pp.1694-1703, 2012.

G. Hrustanovic, V. Olivas, E. Pazarentzos, A. Tulpule, S. Asthana et al.,

, RAS-MAPK dependence underlies a rational polytherapy strategy in EML4-ALK-positive lung cancer, Nat Med, vol.21, issue.9, pp.1038-1085, 2015.

P. M. Enriquez-navas, Exploiting evolutionary principles to prolong tumor control in preclinical models of breast cancer, Sci. Transl. Med, vol.8, pp.327-351, 2016.

R. A. Gatenby, Adaptive therapy, Cancer Res, vol.69, pp.4894-4903, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01887358

A. S. Silva, Y. Kam, Z. P. Khin, S. E. Minton, R. J. Gillies et al., Evolutionary approaches to prolong progression-free survival in breast cancer, Cancer Res, vol.72, pp.6362-70, 2012.

M. P. Morelli, M. J. Overman, A. Dasari, S. Kazmi, T. Mazard et al.,

, Characterizing the patterns of clonal selection in circulating tumor DNA from patients with colorectal cancer refractory to antiEGFR treatment, Ann Oncol, vol.26, pp.731-737, 2015.

A. C. Seghers, S. Wilgenhof, C. Lebbé, and B. Neyns, Successful rechallenge in two patients with BRAF-V600-mutant melanoma who experienced previous progression during treatment with a selective BRAF inhibitor, Melanoma Res, vol.22, pp.466-72, 2012.

D. Thakur, M. Salangsang, F. Landman, A. S. Sellers, W. R. Pryer et al., Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance, Nature, vol.494, pp.251-256, 2013.

W. E. Evans,

V. Mary, Pharmacogenomics: Translating Functional Genomics into Rational Therapeutics. Relling Science, vol.286, issue.5439, pp.487-491, 1999.

J. Robert, On the use of pharmacogenetics in cancer treatment and clinical trials

, Eur J Cancer, vol.50, issue.15, pp.2532-2575, 2014.

J. N. Patel, Cancer pharmacogenomics, challenges in implementation, and patient-focused perspectives, Pharmgenomics Pers Med, vol.9, pp.65-77, 2016.

. On-ikediobi, Somatic pharmacogenomics in cancer, Pharmacogenomics J, 2008.

, , vol.8, pp.305-319

. Kelly-k-filipski, Pharmacogenomics in oncology care. Front Genet, vol.5, p.73, 2014.

M. , Biomarqueurs du cancer bronchique non à petites cellules : du concept à la pratique clinique, La Lettre du Cancérologue, pp.210-233, 2008.

J. Romanetto, Intérêts des biomarqueurs biologiques dans les essais cliniques

, Université Henri Poincaré-Nancy, vol.1, 2011.

C. Serdjebi, Pharmacogénétique des analogues nucléosidiques : Cytidine déaminase et issue clinique. Aix-Marseille Université. 25 septembre, 2015.

M. Billaud, L'invention de la médecine personnalisée, Med Sci (Paris), vol.31, pp.797-803, 2015.

,

F. Eberlé, Cancer : comprendre la médecine personnalisée. Le Figaro Santé. 6 novembre, 2014.

, Les défis de l'immunothérapie en oncologie

C. Granier, Immunothérapies des cancers : rationnel et avancées récentes, Rev Med Interne, 2016.

A. Bellesoeur, Biomarqueurs des immunothérapies anti-PD-1/PD-L1 : facteurs cliniques, histologiques et immunohistochimiques associés au statut PD-L1. Université Paris Descartes. 20 juin, 2016.

M. Graas, Effets secondaires des nouvelles thérapies

L. Létinier, Immunothérapie anti-cancéreuse

, Immunothérapie : tirer parti de l'immunité, p.348

, Ose pharma, vol.29

B. Nassim, Évaluation de l'acte de recherche ou de quantification du gène de fusion BCR-ABL par RT-PCR dans le diagnostic et le suivi thérapeutique des leucémies myéloïdes chroniques et des leucémies lymphoblastiques aiguës, 2017.

A. Turhan, Biologie de la protéine de fusion BCR-ABL : progrès récents, Hématologie, vol.8, issue.1, pp.35-45, 2002.

M. E. Aachab, Les Thérapies ciblées et leur place en onco-hématologie

, Partie II-Autres molécules informationnelles. Chapitre 8-Facteurs de croissance

K. Vari-khajapeer and R. Baskaran, Hsp90 Inhibitors for the Treatment of Chronic Myeloid Leukemia, Leukemia Research and Treatment, vol.2015

É. Lonchamp and F. Nowak, Les Tests de Genetique Moléculaire Pour l'accès Aux Thérapies Ciblées en 2012, 2012.

, Les thérapies ciblées orales. Onconews numéro 10. Octobre 216

, Le prix des médicaments anticancéreux. Institut National du Cancer

P. Casassus, Glivec®): une révolution dans le traitement de la leucémie myéloïde chronique (LMC). Médecine thérapeutique, vol.11, pp.428-436, 2005.

D. M. Hyman, Implementing Genome-Driven Oncology, Cell, vol.168, issue.4, pp.584-599, 2009.
DOI : 10.1016/j.cell.2016.12.015

URL : http://europepmc.org/articles/pmc5463457?pdf=render

C. L. Sawyers, , 2013.

M. , Les inhibiteurs de tyrosine kinase de l'EGFR dans le traitement du CBNPC, N° 8-C2. Octobre, vol.24, pp.188-197, 2007.

A. Chrétien, Fonctionnalité de la signalisation en aval des récepteurs HER : Implication de la réponse cellulaire et tumorale aux thérapies ciblées, 2011.

A. M. Krasinskas, EGFR Signaling in Colorectal Carcinoma, Pathology Research International, vol.2011

,

J. , Kras et cancer colorectal : un pas de géant vers la médecine personnalisée

. Et-biologie-spécialisée, , vol.24, pp.196-209, 2009.

C. Domblides, Traitement des progressions tumorales dans le cancer du poumon muté EGFR : à propos d'une cohorte de patients métastatiques suivis à l'Institut Bergonié, 2015.

T. Valentin, D. Bonnet, and R. Guimbaud, Thérapeutiques ciblées « pour les nuls » : modes d'action des anti-EGFR et des anti-VEGF, Hepato Gastro, vol.18, pp.4-17, 2011.

F. Viret and A. Gonçalves, Cancer colorectaux métastatiques et thérapies ciblées anti-EGFR, MEDECINE/SCIENCES, vol.25, issue.1, pp.13-20, 2009.
DOI : 10.1051/medsci/2009251s13

URL : https://www.medecinesciences.org/articles/medsci/pdf/2009/03/medsci2009251sp13.pdf

C. Audigier-valette, Les inhibiteurs de tyrosine kinase de l'EGFR dans le CBNPC : sont-ils tous identiques ? La Lettre du Cancérologue ?, pp.6-2014

S. Baldacci and A. Cortot, Cancers bronchiques non à petites cellules EGFR mutés, Revue des Maladies Respiratoires Actualités, vol.8, pp.373-381, 2016.

H. Gandomani, Colorectal cancer in the world: incidence, mortality and risk factors, Biomedical Research and Therapy, vol.4, issue.10, pp.1656-1675

,

D. Chan, Epidermal growth factor receptor (EGFR) inhibitors for metastatic colorectal cancer. Cochrane Database Syst Rev, vol.6, p.7047, 2017.

. France, Epidemiologie-des-cancers/Les-cancers-les-plus-frequents/Cancer-colorectal

. Volker-heinemann, Clinical relevance of EGFR-and KRAS-status in colorectal cancer patients treated with monoclonal antibodies directed against the EGFR, Cancer Treatment Reviews, vol.35, issue.3, pp.262-271, 2009.

,

C. Dreyer, Les thérapies ciblées et leurs indications dans les tumeurs solides. La Revue de médecine interne, vol.30, pp.416-424, 2009.

A. Harlé and J. L. Merlin, Biomarqueurs prédictifs de réponse aux thérapies ciblées en oncologie, Ann Biol Clin, vol.71, issue.1, pp.89-97, 2013.

A. Laurenty, J. Selves, and R. Guimbaud, Biomarqueurs des cancers colorectaux utiles en pratique clinique, 2014.

T. André, Les indications et le maniement des anticorps anti-EGFR dans les cancers colorectaux, Hépato-Gastro, vol.15, 2008.

, Commission de la Transparence. Cétuximab. Avis 2 décembre, HAS, 2015.

. Cunningham, Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer, N Engl J Med, vol.351, issue.4, pp.337-382, 2004.

, Commission de la Transparence. Cétuximab. Avis 16 mars, HAS, 2005.

B. Landi, Cétuximab (Erbitux®) dans le traitement du cancer colorectal métastatique

, La lettre de l'hépato-gastroentérologue-n° 2-vol. VIII-mars-avril, pp.81-82, 2005.

, Commission de la Transparence. Panitumumab. Avis 8 mars, HAS, 2017.

«. Http,

J. Y. Douillard, K. S. Oliner, and S. Siena, Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer, N Engl J Med, vol.369, pp.1023-1057, 2013.

C. H. Köhne, First-line panitumumab plus irinotecan/5-fluorouracil/leucovorin treatment in patients with metastatic colorectal cancer, J Cancer Res Clin Oncol, 2012.

, , vol.138, pp.65-72

M. Peeters, Analysis of KRAS/NRAS Mutations in a Phase III Study of Panitumumab with FOLFIRI Compared with FOLFIRI Alone as Second-line Treatment for Metastatic Colorectal Cancer, Clin Cancer Res, vol.21, issue.24, pp.5469-79, 2015.

, Commission de la Transparence. Panitumumab. Avis 8 avril, HAS, 2008.

E. Van-cutsem, Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer, J Clin Oncol, vol.25, issue.13, pp.1658-64, 2007.

, IFCT. Biomarqueurs et cancers du poumon : l'IFCT

, Cancérologie Thoracique) et Roche travaillent ensemble pour offrir plus de chances aux malades, 2010.

J. Simon and . Hollingsworth, Precision medicine in oncology drug development: a pharma perspective, Drug Discov Today, vol.20, issue.12, pp.1455-63, 2015.

I. , LUNG CANCER FACT SHEET. ASLC Highlights Promising Advances. 30 novembre, 2015.

, Lung Cancer Europe. LuCE Report on lung cancer, 2016.

, Séries Guide pour les Patients ESMO. Cancer du poumon non à petites cellules

, Rapport d'orientation. Pertinence du dépistage du cancer broncho-pulmonaire en, HAS

. France, Point de situation sur les données disponibles. Analyse critique des études contrôlées randomisées, 2016.

, Mutations de l'EGFR dans le cancer du poumon : mise en évidence d'une cible moléculaire permettant un accès spécifique aux thérapies ciblées, 2010.

M. Therezien, Place des thérapies ciblées dans la prise en charge du Cancer Bronchique Non à Petites Cellules (CBNPC), 2016.

, Tumeurs du poumon, primitives et secondaires, Pneumologie. Item, vol.306, 2017.

, Globocan. « www.iacr.com

K. Jéhannin-ligier, Rapport technique. Projection de l'incidence et de la mortalité par cancer en France métropolitaine en 2017, 2017.

, Cancer bronchique non à petites cellules / Référentiel national de RCP. Version interactive. Décembre, 2014.

I. Elghissassi, La place des inhibiteurs des tyrosine kinases de l'EGFR dans les cancers bronchiques non à petites cellules avancés, La Lettre du Cancérologue ?, pp.10-2011

H. Guide, Test compagnon associé à une thérapie ciblée : définitions et méthode d'évaluation. Février, 2014.

W. Jacot, Les Thérapies Ciblées les plus utilisées dans les Traitements AntiCancéreux. ICM. 14 octobre, 2014.

M. Fukuoka, Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS), J Clin Oncol, vol.29, pp.2866-2874, 2011.

M. Maemondo, Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR, N Eng J Med, vol.362, issue.25, pp.2380-2388, 2010.

T. Mitsudomi, Gefitinib versus cisplatin plus docetaxel in patients with nonsmall-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial, Lancet Oncol, vol.11, issue.2, pp.121-128, 2010.

T. S. Mok, Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma, 2009.

, N Eng J Med, vol.361, issue.10, pp.947-957

R. Rosell, Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC), Lancet Oncol, vol.13, issue.3, pp.239-246, 2012.

C. Zhou, Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG0802): a multi-centre, open-label, randomised, phase 3 study, Lancet Oncol, vol.12, issue.8, pp.735-742, 2011.

L. V. Sequist, Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations, J Clin Oncol, vol.31, 2013.

P. Fumoleau, Les nouvelles cibles thérapeutiques. Les nouvelles thérapeutiques ciblées. Cancer du sein: Compte-rendu du cours supérieur francophone de cancérologie, pp.233-266

M. Chapeau, Chimiothérapie orale et soins de support : Enquête sur les besoins des pharmaciens d'officine. Création du lien ville-hôpital, 2016.

R. Pérez-soler, Determinants of Tumor Response and Survival With Erlotinib in Patients With Non-Small-Cell Lung Cancer, Journal of Clinical Oncology, vol.22, pp.3238-3247, 2004.

L. Paz-ares, Clinical outcomes in non-small-cell lung cancer patients with EGFR mutations: pooled analysis, J Cell Mol Med, vol.14, issue.1-2, pp.51-69, 2010.

J. Cadranel, Intérêt des biomarqueurs pour prédire l'efficacité des inhibiteurs de tyrosine kinase anti-EGFR, données du projet ERMETIC, La Lettre du Pneumologue ?

, XIII-n° 2-mars-avril 2010, pp.66-67

F. Capuzzo, Erlotinib as maintenance treatment in advanced non-small-cell lung cancer: a multicentre, randomised, placebo-controlled phase 3 study, The Lancet Oncology, vol.11, issue.6, pp.70112-70113, 2010.

C. Costa, The impact of EGFR T790M mutations and BIM mRNA expression on outcome in patients with EGFR-mutant NSCLC treated with erlotinib or chemotherapy in the randomized phase III EURTAC trial, Clin Cancer Res, vol.20, issue.7, pp.2001-2011, 2014.

, Communication directe aux Professionnels de la santé, Tarceva®

, Commission de la transparence, HAS

J. Y. Douillard, Molecular predictors of outcome with gefitinib and docetaxel in previously treated non-small-cell lung cancer: data from the randomized phase III INTEREST trial, J Clin Oncol, vol.28, issue.5, pp.744-52, 2010.

P. Dielenseger, L'afatinib, un inhibiteur de la famille ErbB Données pratiques d'utilisation dans le traitement du cancer bronchique non à petites cellules, Bulletin Infirmier du Cancer, vol.14, pp.42-46

, Commission de la Transparence. Afatinib. 25 mai, HAS, 2016.

, Synthèse d'avis de la Commission de la Transparence, Afatinib. Février, 2014.

, Commission de la Transparence. Osimertinib. 13 septembre, HAS, 2017.

, Commission de la Transparence. Osimertinib. 21 septembre, HAS, 2016.

, Synthèse d'avis de la commission de la Transparence, HAS

F. Hermann, Illuminating ideas : Innovative clinical trial design

«. Roche,

P. Dielenseger, Les essais cliniques en oncologie, Bulletin Infirmier du Cancer

A. Rapport, Conduite des essais cliniques de médicaments en onco/hématologie ciblés, guidés par la génomique, 2014.

R. Simon, Genomic Alteration-Driven Clinical Trial Designs in Oncology, Ann Intern Med, vol.165, issue.4, pp.270-278, 2016.

G. Dominique, Particularité de la recherche clinique en cancérologie

B. Tranchand, Méthodologie des essais cliniques en cancérologie, La Lettre du Pharmacologue, vol.22

U. Derhaschnig, Combined integrated protocol/basket trial design for a first-inhuman trial, Orphanet J Rare Dis, vol.11, issue.1, p.134, 2016.

D. Zardavas, Clinical Trials of Precision Medicine through Molecular Profiling: Focus on Breast Cancer. Am Soc Clin Oncol Educ Book, vol.2015

W. Li, Estimation of treatment effect in two-stage confirmatory oncology trials of personalized medicines, Statistics in Medicine, pp.1843-1861, 2017.

J. Menis, New clinical research strategies in thoracic oncology: clinical trial design, adaptive, basket and umbrella trials, new end-points and new evaluations of response, Eur Respir Rev, vol.23, issue.133, pp.367-78, 2014.

K. M. Cunanan, An efficient basket trial design, Stat Med, vol.36, issue.10, pp.1568-1579, 2017.

L. A. Renfro, Statistical controversies in clinical research: basket trials, umbrella trials, and other master protocols: a review and examples, Ann Oncol, vol.28, issue.1, pp.34-43, 2017.

H. J. West, Novel Precision Medicine Trial Designs: Umbrellas and Baskets, JAMA

, Oncol, vol.3, issue.3, p.423, 2017.

S. Ornes, Core Concept: Basket trial approach capitalizes on the molecular mechanisms of tumors, Proc Natl Acad Sci, vol.113, issue.26, pp.7007-7008, 2016.

R. Simon, The Bayesian basket design for genomic variant-driven phase II trials

, Semin Oncol, vol.43, issue.1, pp.13-18, 2016.

A. J. Redig, Basket trials and the evolution of clinical trial design in an era of genomic medicine, J Clin Oncol, vol.33, issue.9, pp.975-982, 2015.

D. V. Catenacci, Next-generation clinical trials: Novel strategies to address the challenge of tumor molecular heterogeneity, Mol Oncol, vol.9, issue.5, pp.967-96, 2015.

R. A. Beckman, Adaptive Design for a Confirmatory Basket Trial in Multiple Tumor Types Based on a Putative Predictive Biomarker, Clin Pharmacol Ther, vol.100, issue.6, pp.617-625, 2016.

C. Chen, Statistical Design and Considerations of a Phase 3 Basket Trial for Simultaneous Investigation of Multiple Tumor Types in One Study, Statistics in Biopharmaceutical Research, vol.8, pp.2016-2019

J. J. Tao, Basket Studies: Redefining Clinical Trials in the Era of Genome-Driven Oncology, Annu Rev Med, vol.69, pp.319-331, 2018.

, Basket study': Clinical trial design explores responses to drugs based on specific mutations in patients' tumors

. Sciencedaily, , 2015.

J. Stallard, Clinical Trial Shows Promise of "Basket Studies" for Cancer Drugs

, Memorial Sloan Kettering Cancer Center, 2015.

I. Pauporté, Les essais cliniques en cancérologie : spécificités, structures et financements. DIU Chef de Projet / infirmiers & techniciens en recherche clinique. Institut National du Cancer. Département de Recherche Clinique, vol.13

G. Vassal, Biomarker-driven access to crizotinib in ALK, MET or ROS1 positive malignancies in adults and children : feasability of the French National AcSé Program, 2014.

G. Vassal, Biomarker-driven access to crizotinib in ALK, MET, or ROS1 positive (+) malignancies in adults and children: The French National AcSé Program, Journal of Clinical Oncology, vol.36, pp.15-2504, 2018.

, Final results of the Acsé-crizotinib national programme : Demonstration of equal and safe access to a targeted therapy beyond its initial indications. Higlights. Gustave Roussy at ASCO, 2018.

, Commission de la Transparence, HAS

J. Blay, VM) in non-melanoma V600 and non-V600 BRAF mutated cancers: first results of the ACSE trial, Annals of Oncology, vol.27, 2016.

X. Troussard, Advanced Patients with Hairy Cell Leukemia (HCL): Results of the Acsé Phase II Trial, vol.130, p.156, 2018.

J. Blay, Biomarker-driven access to vemurafenib in BRAF-positive cancers: Second study of the French National AcSé Program, Journal of Clinical Oncology, vol.34, pp.11620-11620, 2016.

J. Mazieres, P3.02a-034 Vemurafenib in Patients with Non-Small Cell Lung Cancer (NSCLC) Harboring BRAF Mutation. Preliminary Results of the AcSé Trial, Journal of Thoracic Oncology, vol.12, issue.1, pp.1182-1183

F. Nowak, Rôle de l'INCa dans le soutien de la recherche et des groupes coopérateurs. Institut National du Cancer, Symposium « Immunopathologie et cancers ». 27 mars, 2018.

O. Michel, , 2017.

A. Gonçalves, Nouvelles approches des essais thérapeutiques à l'heure de la médecine personnalisée

D. M. Hyman, Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations, N Engl J Med, vol.373, pp.726-762, 2015.

E. Diamond, Vemurafenib for BRAF V600-Mutant Erdheim-Chester Disease and Langerhans Cell Histiocytosis, JAMA Oncology, vol.4, issue.3, 2017.

D. Michael-hyman, VE-BASKET, a first-in-kind, phase II, histologyindependent "basket" study of vemurafenib (VEM) in nonmelanoma solid tumors harboring BRAF V600 mutations (V600m), Journal of Clinical Oncology, vol.32, pp.2533-2533, 2014.

E. L. Diamond, Vemurafenib in Patients with Erdheim-Chester Disease (ECD) and Langerhans Cell Histiocytosis (LCH) Harboring BRAFV600 Mutations: A Cohort of the

V. Histology-independent and . Study, Blood, vol.128, issue.22, p.480, 2018.

, FDA Announces First Approval of Targeted Therapy Based on Basket Study. Memorial Sloan Kettering Cancer Center, 2017.

P. Schöffski, Activity and safety of crizotinib in patients with advanced clear-cell sarcoma with MET alterations: European Organization for Research and Treatment of Cancer phase II trial 90101 'CREATE'. Ann Oncol, vol.28, pp.3000-3008, 2017.

P. Schöffski, Activity and safety of crizotinib in patients with alveolar soft part sarcoma with rearrangement of TFE3: European Organization for Research and Treatment of Cancer (EORTC) phase II trial 90101 'CREATE'. Ann Oncol, vol.29, pp.758-765, 2018.

P. Schöffski, The tyrosine kinase inhibitor crizotinib does not have clinically meaningful activity in heavily pre-treated patients with advanced alveolar rhabdomyosarcoma with FOXO rearrangement: European Organisation for Research and Treatment of Cancer phase 2 trial 90101 'CREATE', Eur J Cancer, vol.94, pp.156-167, 2018.

P. Schöffski, Crizotinib in patients with advanced, inoperable inflammatory myofibroblastic tumours with and without anaplastic lymphoma kinase gene alterations (European Organisation for Research and Treatment of Cancer 90101 CREATE): a multicentre, single-drug, prospective, non-randomised phase 2 trial, Lancet Respir Med, 2018.

, , vol.6, pp.30116-30120

P. Schöffski, Crizotinib achieves objective responses and long-lasting disease control in patients with metastatic papillary renal cell carcinoma type 1 (pRCC1) with somatic MET mutations. EORTC phase II trial 90101 'CREATE, AACR Annual Meeting, 2016.

C. Abstract, , 2016.

A. Drilon, STARTRK-2: A global phase 2, open-label, basket study of entrectinib in patients with locally advanced or metastatic solid tumors harboring TRK, ROS1, or ALK gene fusions, Supplement), vol.060, issue.77, 2017.

P. Knox, Ignyta Announces Results for Potential New Cancer Treatment, 2016.

A. Drilon, D. Braud, F. G. Siena, and S. , Entrectinib, an oral pan-Trk, ROS1, and ALK inhibitor in TKI-naïve patients with advanced solid tumors harboring gene rearrangements, Presented at the 2016 AACR Annual Meeting

, Ignyta's-Entrectinib-Suggests-Potential-Best-in-Class

L. Astor, Potential Seen for Entrectinib for Specific Patients With Sarcoma Harboring Genetic Rearrangements. Targeted Oncology, 2017.

P. Multani, Poster 080 #2798661 Preliminary evidence of clinical response to entrectinib in three sarcoma patients, CTOS Annual Meeting, 2017.

D. Liu, Entrectinib: an orally available, selective tyrosine kinase inhibitor for the treatment of NTRK, ROS1, and ALK fusion-positive solid tumors, Ther Clin Risk Manag, vol.14, pp.1247-1252, 2018.

D. J. Slamon, Use of Chemotherapy plus a Monoclonal Antibody against HER2 for Metastatic Breast Cancer That Overexpresses HER2, N Engl J Med, vol.344, p.783, 2001.

,

D. M. Hyman, HER kinase inhibition in patients with HER2-and HER3-mutant cancers, Nature, vol.554, issue.7691, pp.189-194, 2018.

E. W. Alley, Clinical safety and activity of pembrolizumab in patients with malignant pleural mesothelioma (KEYNOTE-028): preliminary results from a nonrandomised, open-label, phase 1b trial, Lancet Oncol, vol.18, issue.5, pp.623-630, 2017.

P. A. Ott, Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with recurrent carcinoma of the anal canal, Ann Oncol, vol.28, issue.5, pp.1036-1041, 2017.

C. Hsu, Safety and Antitumor Activity of Pembrolizumab in Patients With Programmed Death-Ligand 1-Positive Nasopharyngeal Carcinoma: Results of the KEYNOTE-028 Study, J Clin Oncol, vol.35, issue.36, pp.4050-4056, 2017.

J. M. 427o-mehnert, Pembrolizumab for patients with PD-L1-positive advanced carcinoid or pancreatic neuroendocrine tumors: results from the KEYNOTE-028 study, Annals of Oncology, vol.28, 2017.

A. David, ATIM-35. Results of the phase IB KEYNOTE-028 multi-cohort trial of pembrolizumab monotherapy in patients with recurrent PD-L1-positive glioblastoma multiforme (GBM), Neuro-Oncology, vol.18, pp.25-26, 2016.

A. Varga, Pembrolizumab in patients (pts) with PD-L1-positive (PD-L1+) advanced ovarian cancer: Updated analysis of KEYNOTE-028, Journal of Clinical Oncology, vol.35, pp.5513-5513, 2017.

Y. J. Bang, 525 POSTER : Safety and efficacy of pembrolizumab (MK-3475) in patients (pts) with advanced biliary tract cancer: Interim results of KEYNOTE-028, European Journal of Cancer, vol.51, 2015.

P. A. Ott, Pembrolizumab in Patients With Extensive-Stage Small-Cell Lung Cancer: Results From the Phase Ib KEYNOTE-028 Study, J Clin Oncol, vol.35, issue.34, pp.3823-3829, 2017.

T. Doi, Safety and Antitumor Activity of the Anti-Programmed Death-1 Antibody Pembrolizumab in Patients With Advanced Esophageal Carcinoma, J Clin Oncol, vol.36, issue.1, pp.61-67, 2018.

J. S. Frenel, Safety and Efficacy of Pembrolizumab in Advanced, Programmed Death Ligand 1-Positive Cervical Cancer: Results From the Phase Ib KEYNOTE-028 Trial, J Clin Oncol, vol.35, issue.36, pp.4035-4041, 2017.

O. Bh, Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced colorectal carcinoma. PLoS One, vol.12, p.189848, 2017.

R. B. Cohen, Pembrolizumab for the Treatment of Advanced Salivary Gland Carcinoma: Findings of the Phase 1b KEYNOTE-028 Study, Am J Clin Oncol, 2018.

H. S. Rugo, Safety and Antitumor Activity of Pembrolizumab in Patients with Estrogen Receptor-Positive/Human Epidermal Growth Factor Receptor 2-Negative Advanced Breast Cancer, Clin Cancer Res, vol.24, issue.12, pp.2804-2811, 2018.

P. A. Ott, Safety and Antitumor Activity of Pembrolizumab in Advanced Programmed Death Ligand 1-Positive Endometrial Cancer: Results From the KEYNOTE-028

. Study, , vol.35, pp.2535-2541, 2017.

A. R. Hansen, Pembrolizumab for advanced prostate adenocarcinoma: findings of the KEYNOTE-028 study, Ann Oncol, vol.29, issue.8, pp.1807-1813, 2018.

M. Stenger, Pembrolizumab in MSI-H or dMMR Solid Tumors

, Tissue/Site-Agnostic' Approval by FDA. The Asco post, 2018.

E. D. Slosberg, Signature program: a platform of basket trials, Oncotarget, vol.9, pp.21383-21395, 2018.

B. P. Kang, The Signature Program: Bringing the Protocol to the Patient, Clin Pharmacol Ther, vol.98, issue.2, pp.124-126, 2015.

C. Melton, Novel Trial Program Puts Patients First, Targeted Oncology, 2015.

T. Berberabe, Signature Trial Program Aims for Rapid Delivery of Novel Therapies

&. Oncologylive and . Vol, , vol.17, 2016.

A. Drilon, Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children, N Engl J Med, vol.378, issue.8, pp.731-739, 2018.

Y. Chen and P. Chi, Basket trial of TRK inhibitors demonstrates efficacy in TRK fusion-positive cancers, J Hematol Oncol, vol.11, p.78, 2018.

R. Simon, New Designs for Basket Clinical Trials in Oncology, Journal of Biopharmaceutical Statistics, 2017.

S. Dangi-garimella, New Oncology Clinical Trial Designs: What Works and What Doesn't? Evidence-Based Oncology, The American Society of Clinical Oncology Annual Meeting, 2015.

N. Meredith, Basket Study" May Open Doors for, Clinical Trials for Mesothelioma Patients. Mesothelioma Counsel. Posted on Thursday, 2015.

L. Musial, Basket Trials" Mark An Exciting New Era In Cancer Research and Treatment. Formerly of Roswell Park Comprehensive Cancer Center, Tuesday, 2015.

K. Ruddy, Basket Studies: The Next Frontier In Breast Cancer Research. Breast Health, 2015.

M. Garland, Memorial Sloan Kettering Cancer Center Announces Innovative Basket Study. The Life Raft Group, 2015.

A. V. Biankin, S. Piantadosi, and S. J. Hollingsworth, Patient-centric trials for therapeutic development in precision oncology, Nature, vol.526, issue.7573, pp.361-70, 2015.