D. 'herelle and F. , Sur un microbe invisible antagoniste des bacilles dysentériques, Comptes Rendus Acad. Sci. Paris, vol.165, pp.373-375, 1917.

M. Kutateladze, Experience of the Eliava Institute in bacteriophage therapy, Virologica Sinica, vol.69, issue.1, pp.80-81, 2015.
DOI : 10.1093/jac/dkt453

S. Jennes, M. Merabishvili, P. Soentjens, K. W. Pang, T. Rose et al., Use of bacteriophages in the treatment of colistin-only-sensitive Pseudomonas aeruginosa septicaemia in a patient with acute kidney injury???a case report, Critical Care, vol.363, issue.1, p.129, 2017.
DOI : 10.1093/femsle/fnv242

R. T. Schooley, B. Biswas, J. J. Gill, A. Hernandez-morales, J. Lancaster et al.,

F. Rohwer and S. Benler, Development and use of personalized bacteriophage-based therapeutic cocktails to treat a patient with a disseminated resistant Acinetobacter baumannii infection, Antimicrob. Agents Chemother, p.61, 2017.

A. Wright, C. H. Hawkins, E. E. Anggard, D. R. Harper, S. Sultana et al., ; a preliminary report of efficacy, CrossRef] [PubMed] 6. Sarker,CrossRef] [PubMed] 7. Sarker, S.A.; Brussow, H. From bench to bed and back again: Phage therapy of childhood Escherichia coli diarrhea, pp.349-357, 2009.
DOI : 10.3201/eid0809.020063

S. Chibani-chennoufi, J. Sidoti, A. Bruttin, M. L. Dillmann, E. Kutter et al., Isolation of Escherichia coli Bacteriophages from the Stool of Pediatric Diarrhea Patients in Bangladesh, Journal of Bacteriology, vol.186, issue.24, pp.8287-8294, 2004.
DOI : 10.1128/JB.186.24.8287-8294.2004

D. Sordi, L. Khanna, V. Debarbieux, and L. , The Gut Microbiota Facilitates Drifts in the Genetic Diversity and Infectivity of Bacterial Viruses, Cell Host & Microbe, vol.22, issue.6, pp.801-808, 2017.
DOI : 10.1016/j.chom.2017.10.010

URL : https://hal.archives-ouvertes.fr/pasteur-01827316

M. Galtier, L. De-sordi, D. Maura, H. Arachchi, S. Volant et al., Bacteriophages to reduce gut carriage of antibiotic resistant uropathogens with low impact on microbiota composition, Environmental Microbiology, vol.350, issue.7, pp.2237-2245, 2016.
DOI : 10.1126/science.aad6791

URL : https://hal.archives-ouvertes.fr/pasteur-01538960

D. Maura, M. Galtier, C. Le-bouguenec, and L. Debarbieux, ABSTRACT, Antimicrobial Agents and Chemotherapy, vol.56, issue.12, pp.6235-6242
DOI : 10.1128/AAC.00602-12

M. Weiss, E. Denou, A. Bruttin, R. Serra-moreno, M. L. Dillmann et al., In vivo replication of T4 and T7 bacteriophages in germ-free mice colonized with Escherichia coli, Virology, vol.393, issue.1, pp.16-23, 2009.
DOI : 10.1016/j.virol.2009.07.020

J. Oh, A. L. Byrd, C. Deming, S. Conlan, N. C. Program et al., Biogeography and individuality shape function in the human skin metagenome, Nature, vol.2, issue.7520, pp.59-64, 2014.
DOI : 10.1080/STA-200066418

J. C. Venter, K. Remington, J. F. Heidelberg, A. L. Halpern, D. Rusch et al., Environmental Genome Shotgun Sequencing of the Sargasso Sea, Science, vol.304, issue.5667, pp.66-74, 2004.
DOI : 10.1126/science.1093857

C. N. Signori, F. Thomas, A. Enrich-prast, R. C. Pollery, and S. M. Sievert, Microbial diversity and community structure across environmental gradients in Bransfield Strait, Western Antarctic Peninsula, Frontiers in Microbiology, vol.49, p.647, 2014.
DOI : 10.1016/S0967-0637(01)00062-0

P. G. Falkowski, T. Fenchel, and E. Delong, The Microbial Engines That Drive Earth's Biogeochemical Cycles, Science, vol.103, issue.32, pp.1034-1039, 2008.
DOI : 10.1073/pnas.0605127103

J. D. Van-elsas, A. V. Semenov, R. Costa, and J. Trevors, Survival of Escherichia coli in the environment: fundamental and public health aspects, The ISME Journal, vol.191, issue.2, pp.173-183, 2011.
DOI : 10.1128/JB.00599-08

E. Denou, B. Berger, C. Barretto, J. M. Panoff, F. Arigoni et al., Gene Expression of Commensal Lactobacillus johnsonii Strain NCC533 during In Vitro Growth and in the Murine Gut, Journal of Bacteriology, vol.189, issue.22, pp.8109-8119, 2007.
DOI : 10.1128/JB.00991-07

J. P. Feugeas, J. Tourret, A. Launay, O. Bouvet, C. Hoede et al., : Correlations between Gene Expression and Gene Variability Reflect Growth Efficiencies, Molecular Biology and Evolution, vol.67, issue.10, pp.2515-2529, 2016.
DOI : 10.1016/j.mrrev.2012.12.003

C. Janoir, C. Deneve, S. Bouttier, F. Barbut, S. Hoys et al., ABSTRACT, Infection and Immunity, vol.81, issue.10, pp.3757-3769, 2013.
DOI : 10.1128/IAI.00515-13

J. D. Partridge, C. Scott, Y. Tang, R. K. Poole, and J. Green, Transcriptome Dynamics during the Transition from Anaerobic to Aerobic Conditions, Journal of Biological Chemistry, vol.145, issue.38, pp.27806-27815, 2006.
DOI : 10.1038/nature03239

J. A. Snyder, B. J. Haugen, E. L. Buckles, C. V. Lockatell, D. E. Johnson et al., Transcriptome of Uropathogenic Escherichia coli during Urinary Tract Infection, Infection and Immunity, vol.72, issue.11, pp.6373-6381, 2004.
DOI : 10.1128/IAI.72.11.6373-6381.2004

A. Stintzi, D. Marlow, K. Palyada, H. Naikare, R. Panciera et al., Use of Genome-Wide Expression Profiling and Mutagenesis To Study the Intestinal Lifestyle of Campylobacter jejuni, Infection and Immunity, vol.73, issue.3, pp.1797-1810, 2005.
DOI : 10.1128/IAI.73.3.1797-1810.2005

S. J. Labrie, J. E. Samson, and S. Moineau, Bacteriophage resistance mechanisms, Nature Reviews Microbiology, vol.153, issue.5, pp.317-327, 2010.
DOI : 10.1007/s00438-004-1048-y

T. Goldfarb, H. Sberro, E. Weinstock, O. Cohen, S. Doron et al., Sorek, R. BREX is a novel phage resistance system widespread in microbial genomes, EMBO J. 2015, vol.34, pp.169-183

S. Doron, S. Melamed, G. Ofir, A. Leavitt, A. Lopatina et al., Systematic discovery of antiphage defense systems in the microbial pangenome, Science, vol.95, issue.6379, p.359, 2018.
DOI : 10.1126/science.aad9822

D. Bryan, A. El-shibiny, Z. Hobbs, J. Porter, and E. M. Kutter, Bacteriophage T4 Infection of Stationary Phase E. coli: Life after Log from a Phage Perspective, Frontiers in Microbiology, vol.86, issue.fnw047, 1391.
DOI : 10.1016/S0092-8674(00)80089-6

P. Golec, J. Karczewska-golec, M. Los, and G. Wegrzyn, host: comparison of a mathematical model with the experimental data, FEMS Microbiology Letters, vol.184, issue.2, pp.156-161, 2014.
DOI : 10.1128/JB.184.7.1888-1894.2002

H. Hadas, M. Einav, I. Fishov, and A. Zaritsky, Bacteriophage T4 Development Depends on the Physiology of its Host Escherichia Coli, Microbiology, vol.143, issue.1, pp.179-185, 1997.
DOI : 10.1099/00221287-143-1-179

URL : http://mic.microbiologyresearch.org/deliver/fulltext/micro/143/1/mic-143-1-179.pdf?itemId=/content/journal/micro/10.1099/00221287-143-1-179&mimeType=pdf&isFastTrackArticle=

D. Nabergoj, P. Modic, and A. Podgornik, Effect of bacterial growth rate on bacteriophage population growth rate. Microbiol, 2017.
DOI : 10.1002/mbo3.558

URL : http://onlinelibrary.wiley.com/doi/10.1002/mbo3.558/pdf

C. P. Ricciuti, Host-virus interactions in Escherichia coli: Effect of stationary phase on viral release from MS2-infected bacteria, J. Virol, vol.10, pp.162-165, 1972.

B. Propst-ricciuti, The Effect of Host-Cell Starvation on Virus-induced Lysis by MS2 Bacteriophage, Journal of General Virology, vol.31, issue.3, pp.323-330, 1976.
DOI : 10.1099/0022-1317-31-3-323

J. J. Bull, C. S. Vegge, M. Schmerer, W. N. Chaudhry, and B. R. Levin, Phenotypic Resistance and the Dynamics of Bacterial Escape from Phage Control, PLoS ONE, vol.2, issue.4, p.94690
DOI : 10.1371/journal.pone.0094690.t001

URL : https://doi.org/10.1371/journal.pone.0094690

E. Chapman-mcquiston and X. L. Wu, Stochastic Receptor Expression Allows Sensitive Bacteria to Evade Phage Attack. Part I: Experiments, Biophysical Journal, vol.94, issue.11, pp.4525-4536, 2008.
DOI : 10.1529/biophysj.107.120212

B. R. Levin, S. Moineau, M. Bushman, and R. Barrangou, The Population and Evolutionary Dynamics of Phage and Bacteria with CRISPR???Mediated Immunity, PLoS Genetics, vol.255, issue.2, p.1003312
DOI : 10.1371/journal.pgen.1003312.s007

URL : https://doi.org/10.1371/journal.pgen.1003312

B. Labedan, Requirement for a fluid host cell membrane in injection of coliphage T5 DNA, J. Virol, vol.49, pp.273-275, 1984.

R. Daugelavicius, A. Gaidelyte, V. Cvirkaite-krupovic, and D. H. Bamford, On-line monitoring of changes in host cell physiology during the one-step growth cycle of Bacillus phage Bam35, Journal of Microbiological Methods, vol.69, issue.1, pp.174-179, 2007.
DOI : 10.1016/j.mimet.2006.12.023

S. Sillankorva, R. Oliveira, M. J. Vieira, I. Sutherland, and J. Azeredo, infection by bacteriophage ????S1: the influence of temperature, host growth phase and media, FEMS Microbiology Letters, vol.399, issue.1, pp.13-20, 2004.
DOI : 10.1128/AAC.39.3.645

URL : https://academic.oup.com/femsle/article-pdf/241/1/13/19122124/241-1-13.pdf

N. Dufour, O. Clermont, B. La-combe, J. Messika, S. Dion et al., clonal complex, Journal of Antimicrobial Chemotherapy, vol.2, issue.11, pp.3072-3080, 2016.
DOI : 10.1093/jac/dks261

URL : https://hal.archives-ouvertes.fr/pasteur-01539016

Y. Ohshima, F. Schumacher-perdreau, G. Peters, and G. Pulverer, The role of capsule as a barrier to bacteriophage adsorption in an encapsulated Staphylococcus simulans strain, Medical Microbiology and Immunology, vol.177, issue.4, pp.229-233, 1988.
DOI : 10.1007/BF00211222

D. R. Roach, D. R. Sjaarda, A. J. Castle, and A. M. Svircev, ABSTRACT, Applied and Environmental Microbiology, vol.79, issue.10, pp.3249-3256, 2013.
DOI : 10.1128/AEM.00067-13

A. M. Nanda, K. Thormann, and J. Frunzke, Impact of Spontaneous Prophage Induction on the Fitness of Bacterial Populations and Host-Microbe Interactions, Journal of Bacteriology, vol.197, issue.3, pp.410-419, 2015.
DOI : 10.1128/JB.02230-14

M. De-paepe, M. Leclerc, C. R. Tinsley, and M. A. Petit, Bacteriophages: an underestimated role in human and animal health?, Frontiers in Cellular and Infection Microbiology, vol.181, issue.9, p.39, 2014.
DOI : 10.1086/315239

URL : https://hal.archives-ouvertes.fr/hal-01204359

P. Manrique, B. Bolduc, S. T. Walk, J. Van-der-oost, W. M. De-vos et al., Healthy human gut phageome, Proc. Natl. Acad. Sci. USA 2016, pp.10400-10405
DOI : 10.1371/journal.pone.0035053

URL : http://www.pnas.org/content/113/37/10400.full.pdf

H. C. Flemming, J. Wingender, U. Szewzyk, P. Steinberg, S. A. Rice et al., Biofilms: an emergent form of bacterial life, Nature Reviews Microbiology, vol.10, issue.9, pp.563-575, 2016.
DOI : 10.1111/j.1365-2958.2005.04938.x

J. W. Costerton, P. S. Stewart, and E. P. Greenberg, Bacterial Biofilms: A Common Cause of Persistent Infections, Science, vol.284, issue.5418, pp.1318-1322, 1999.
DOI : 10.1126/science.284.5418.1318

L. Fernandez, A. Rodriguez, and P. Garcia, Phage or foe: an insight into the impact of viral predation on microbial communities, The ISME Journal, vol.5, issue.5, pp.1171-1179, 2018.
DOI : 10.1371/journal.pone.0015678

L. Vidakovic, P. K. Singh, R. Hartmann, C. D. Nadell, and K. Drescher, Dynamic biofilm architecture confers individual and collective mechanisms of viral protection, Nature Microbiology, vol.966, issue.1, pp.26-31, 2018.
DOI : 10.1007/978-1-62703-245-2_4

URL : https://www.nature.com/articles/s41564-017-0050-1.pdf

G. Majkowska-skrobek, A. Latka, R. Berisio, B. Maciejewska, F. Squeglia et al., Capsule-Targeting Depolymerase, Derived from Klebsiella KP36 Phage, as a Tool for the Development of Anti-Virulent Strategy, Viruses, vol.133, issue.12, p.324, 2016.
DOI : 10.1111/j.1432-1033.1990.tb15578.x

D. P. Pires, H. Oliveira, L. D. Melo, S. Sillankorva, and J. Azeredo, Bacteriophage-encoded depolymerases: their diversity and biotechnological applications, Applied Microbiology and Biotechnology, vol.28, issue.Pt 2, pp.2141-2151, 2016.
DOI : 10.1007/s40259-013-0081-y

M. Simmons, K. Drescher, C. D. Nadell, and V. Bucci, Phage mobility is a core determinant of phage???bacteria coexistence in biofilms, The ISME Journal, vol.42, issue.2, pp.531-543, 2018.
DOI : 10.1002/bit.20544

K. J. Brathwaite, P. Siringan, P. L. Connerton, and I. Connerton, Host adaption to the bacteriophage carrier state of Campylobacter jejuni, Research in Microbiology, vol.166, issue.6, pp.504-515, 2015.
DOI : 10.1016/j.resmic.2015.05.003

P. Siringan, P. L. Connerton, N. J. Cummings, and I. Connerton, Alternative bacteriophage life cycles: the carrier state of Campylobacter jejuni, Open Biology, vol.9, issue.7, 2014.
DOI : 10.1371/journal.pgen.1003269

URL : http://rsob.royalsocietypublishing.org/content/royopenbio/4/3/130200.full.pdf

S. Heilmann, K. Sneppen, and S. Krishna, Coexistence of phage and bacteria on the boundary of self-organized refuges, Proc. Natl. Acad. Sci. USA 2012, pp.12828-12833
DOI : 10.1371/journal.pbio.0040193

M. A. Brockhurst, A. Buckling, and P. B. Rainey, Spatial heterogeneity and the stability of host-parasite coexistence, Journal of Evolutionary Biology, vol.15, issue.2, pp.374-379, 2006.
DOI : 10.1046/j.1365-3059.2002.00683.x

S. J. Schrag and J. Mittler, Host-Parasite Coexistence: The Role of Spatial Refuges in Stabilizing Bacteria-Phage Interactions, The American Naturalist, vol.148, issue.2, pp.348-377, 1996.
DOI : 10.1086/285929

R. S. Eriksen, S. L. Svenningsen, K. Sneppen, and N. Mitarai, A growing microcolony can survive and support persistent propagation of virulent phages, Proc. Natl. Acad. Sci. USA 2018, pp.337-342
DOI : 10.1073/pnas.1708954115

R. J. Payne, D. Phil, and V. A. Jansen, Phage therapy: The peculiar kinetics of self-replicating pharmaceuticals, Clinical Pharmacology & Therapeutics, vol.68, issue.3, pp.225-230, 2000.
DOI : 10.1067/mcp.2000.109520

B. A. Wiggins and M. Alexander, Minimum bacterial density for bacteriophage replication: Implications for significance of bacteriophages in natural ecosystems, Appl. Environ. Microbiol, vol.49, pp.19-23, 1985.

A. Reyes, M. Wu, N. P. Mcnulty, F. L. Rohwer, and J. Gordon, Gnotobiotic mouse model of phage-bacterial host dynamics in the human gut, Proc. Natl. Acad. Sci. USA 2013, pp.20236-20241
DOI : 10.1073/pnas.1206136109

N. Yutin, K. S. Makarova, A. B. Gussow, M. Krupovic, A. Segall et al., Discovery of an expansive bacteriophage family that includes the most abundant viruses from the human gut, Nature Microbiology, vol.35, issue.1, pp.38-46, 2018.
DOI : 10.1093/nar/gkm265

G. P. Donaldson, S. M. Lee, and S. K. Mazmanian, Gut biogeography of the bacterial microbiota, Nature Reviews Microbiology, vol.56, issue.1, pp.20-32, 2016.
DOI : 10.1371/journal.pone.0053957

, Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome, Nature, vol.2012, issue.486, pp.207-214

G. He, R. A. Shankar, M. Chzhan, A. Samouilov, P. Kuppusamy et al., Noninvasive measurement of anatomic structure and intraluminal oxygenation in the gastrointestinal tract of living mice with spatial and spectral EPR imaging, Proc. Natl. Acad. Sci, pp.4586-4591, 1999.
DOI : 10.1088/0031-9155/43/7/017

M. Koziolek, M. Grimm, D. Becker, V. Iordanov, H. Zou et al., Investigation of pH and Temperature Profiles in the GI Tract of Fasted Human Subjects Using the Intellicap?? System, Journal of Pharmaceutical Sciences, vol.104, issue.9, pp.2855-2863, 2015.
DOI : 10.1002/jps.24274

T. V. Maier, M. Lucio, L. H. Lee, N. C. Verberkmoes, C. J. Brislawn et al., ABSTRACT, mBio, vol.8, issue.5, pp.1343-1360, 2017.
DOI : 10.1128/mBio.01343-17

B. Marteyn, N. P. West, D. F. Browning, J. A. Cole, J. G. Shaw et al., Modulation of Shigella virulence in response to available oxygen in vivo, Nature, vol.168, issue.7296, pp.355-358, 2010.
DOI : 10.1007/s004240050455

URL : https://hal.archives-ouvertes.fr/pasteur-00493767

Y. Wang, E. Holmes, E. M. Comelli, G. Fotopoulos, G. Dorta et al., Topographical variation in metabolic signatures of human gastrointestinal biopsies revealed by high-resolution magic-angle spinning 1H NMR spectroscopy

, J. Proteome Res, vol.6, pp.3944-3951, 2007.

L. Hoyles, A. L. Mccartney, H. Neve, G. R. Gibson, J. D. Sanderson et al., Characterization of virus-like particles associated with the human faecal and caecal microbiota, Research in Microbiology, vol.165, issue.10, pp.803-812, 2014.
DOI : 10.1016/j.resmic.2014.10.006

C. L. Monaco, D. B. Gootenberg, G. Zhao, S. A. Handley, M. S. Ghebremichael et al., Altered Virome and Bacterial Microbiome in Human Immunodeficiency Virus-Associated Acquired Immunodeficiency Syndrome, Cell Host & Microbe, vol.19, issue.3, pp.311-322, 2016.
DOI : 10.1016/j.chom.2016.02.011

J. M. Norman, S. A. Handley, M. T. Baldridge, L. Droit, C. Y. Liu et al., Disease-Specific Alterations in the Enteric Virome in Inflammatory Bowel Disease, Disease-specific alterations in the enteric virome in inflammatory bowel disease, pp.447-460, 2015.
DOI : 10.1016/j.cell.2015.01.002

G. Zhao, T. Vatanen, L. Droit, A. Park, A. D. Kostic et al., Intestinal virome changes precede autoimmunity in type I diabetes-susceptible children, Proc. Natl. Acad. Sci. USA 2017, pp.6166-6175
DOI : 10.1038/ismej.2011.139

M. Vayssier-taussat, E. Albina, C. Citti, J. F. Cosson, M. A. Jacques et al., Shifting the paradigm from pathogens to pathobiome: new concepts in the light of meta-omics, Frontiers in Cellular and Infection Microbiology, vol.24, issue.138, p.29, 2014.
DOI : 10.1177/0022034512449466

URL : https://hal.archives-ouvertes.fr/hal-01153583

S. J. Ott, G. H. Waetzig, A. Rehman, J. Moltzau-anderson, R. Bharti et al., Efficacy of Sterile Fecal Filtrate Transfer for Treating Patients With Clostridium difficile Infection, Gastroenterology, vol.152, issue.4, pp.799-811, 2017.
DOI : 10.1053/j.gastro.2016.11.010

T. Zuo, S. H. Wong, K. Lam, R. Lui, K. Cheung et al., infection is associated with treatment outcome, Gut, vol.7, issue.(Suppl 1), pp.634-643, 2018.
DOI : 10.1136/gutjnl-2017-313952

J. J. Barr, R. Auro, M. Furlan, K. L. Whiteson, M. L. Erb et al., Bacteriophage adhering to mucus provide a non-host-derived immunity, Proc. Natl. Acad. Sci. USA 2013, pp.10771-10776
DOI : 10.1016/0022-2836(89)90464-6

D. Sordi, L. Lourenço, and M. , Debarbieux, L. I will survive: A tale of bacteriophage-bacteria coevolution in the gut, Gut Microbes, 2018.

L. Debarbieux, Bacterial sensing of bacteriophages in communities: the search for the Rosetta stone, Current Opinion in Microbiology, vol.20, issue.20, pp.125-130
DOI : 10.1016/j.mib.2014.05.015

URL : https://hal.archives-ouvertes.fr/pasteur-01539083

M. K. Mirzaei and C. Maurice, M??nage ?? trois in the human gut: interactions between host, bacteria and phages, Nature Reviews Microbiology, vol.102, issue.7, pp.397-408
DOI : 10.1073/pnas.0504062102

M. Galtier, L. De-sordi, A. Sivignon, A. De-vallee, D. Maura et al., strains as a promising new treatment for Crohn's disease, Journal of Crohn's and Colitis, vol.11, pp.840-847, 2017.
DOI : 10.1093/ecco-jcc/jjw224

E. Morello, E. Saussereau, D. Maura, M. Huerre, L. Touqui et al., Pulmonary Bacteriophage Therapy on Pseudomonas aeruginosa Cystic Fibrosis Strains: First Steps Towards Treatment and Prevention, PLoS ONE, vol.72, issue.Pt 10, 2011.
DOI : 10.1371/journal.pone.0016963.t001

URL : https://hal.archives-ouvertes.fr/pasteur-01425539

G. K. Vinner, G. T. Vladisavljevic, M. R. Clokie, and D. J. Malik, Microencapsulation of Clostridium difficile specific bacteriophages using microfluidic glass capillary devices for colon delivery using pH triggered release, PLOS ONE, vol.23, issue.2, p.186239, 2017.
DOI : 10.1371/journal.pone.0186239.s001

A. M. Comeau, F. Tetart, S. N. Trojet, M. F. Prere, and H. M. Krisch, Phage-Antibiotic Synergy (PAS): ??-Lactam and Quinolone Antibiotics Stimulate Virulent Phage Growth, PLoS ONE, vol.50, issue.8, p.799, 2007.
DOI : 10.1371/journal.pone.0000799.g005

URL : https://hal.archives-ouvertes.fr/hal-00180524

F. Oechslin, P. Piccardi, S. Mancini, J. Gabard, P. Moreillon et al., infection in endocarditis and reduces virulence, Journal of Infectious Diseases, vol.215, pp.703-712, 2017.
DOI : 10.1093/infdis/jiw632

C. Torres-barcelo, F. I. Arias-sanchez, M. Vasse, J. Ramsayer, O. Kaltz et al., A Window of Opportunity to Control the Bacterial Pathogen Pseudomonas aeruginosa Combining Antibiotics and Phages, PLoS ONE, vol.25, issue.2, p.106628
DOI : 10.1371/journal.pone.0106628.g004

URL : https://hal.archives-ouvertes.fr/hal-02015241

W. N. Chaudhry, J. Concepcion-acevedo, T. Park, S. Andleeb, J. J. Bull et al., Synergy and Order Effects of Antibiotics and Phages in Killing Pseudomonas aeruginosa Biofilms, PLOS ONE, vol.43, issue.4, p.168615, 2017.
DOI : 10.1371/journal.pone.0168615.s007

E. M. Ryan, M. Y. Alkawareek, R. F. Donnelly, and B. Gilmore, FEMS Immunology & Medical Microbiology, vol.37, issue.2, pp.395-398, 2012.
DOI : 10.1016/S0022-5193(03)00262-5

V. Verma, K. Harjai, and S. Chhibber, Restricting ciprofloxacin-induced resistant variant formation in biofilm of Klebsiella pneumoniae B5055 by complementary bacteriophage treatment, Journal of Antimicrobial Chemotherapy, vol.49, issue.7, pp.1212-1218, 2009.
DOI : 10.1128/AAC.49.7.2874-2878.2005

R. C. Allen, K. R. Pfrunder-cardozo, D. Meinel, A. Egli, and A. R. Hall, ABSTRACT, mBio, vol.8, issue.5, pp.1341-1358, 2017.
DOI : 10.1128/mBio.01341-17

D. R. Roach, C. Y. Leung, M. Henry, E. Morello, D. Singh et al., Synergy between the Host Immune System and Bacteriophage Is Essential for Successful Phage Therapy against an Acute Respiratory Pathogen, Cell Host & Microbe, vol.22, issue.1, pp.38-47, 2017.
DOI : 10.1016/j.chom.2017.06.018

URL : https://hal.archives-ouvertes.fr/pasteur-01827320