L. Antunes, P. Visca, and K. J. Towner, Acinetobacter baumannii: evolution of a global pathogen, Pathog Dis, vol.71, pp.292-301, 2014.

L. Dijkshoorn, A. Nemec, and H. Seifert, An increasing threat in hospitals: multidrug-resistant Acinetobacter baumannii, Nat Rev Microbiol, vol.5, pp.939-51, 2007.

P. Fournier, D. Vallenet, V. Barbe, S. Audic, H. Ogata et al., Comparative Genomics of Multidrug Resistance in Acinetobacter baumannii, PLOS Genet, vol.2, p.7, 2006.

R. L. Nation, J. Li, O. Cars, W. Couet, M. N. Dudley et al., Framework for optimisation of the clinical use of colistin and polymyxin B: the Prato polymyxin consensus, Lancet Infect Dis, vol.15, pp.225-259, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02479001

E. I. Nielsen and L. E. Friberg, Pharmacokinetic-pharmacodynamic modeling of antibacterial drugs, Pharmacol Rev, vol.65, pp.1053-90, 2013.

M. Brill, A. N. Kristoffersson, C. Zhao, E. I. Nielsen, and L. E. Friberg, Semi-mechanistic pharmacokinetic-pharmacodynamic modelling of antibiotic drug combinations, Clin Microbiol Infect, vol.0, 2017.

D. J. Hong, J. O. Kim, H. Lee, E. Yoon, S. H. Jeong et al., In vitro antimicrobial synergy of colistin with rifampicin and carbapenems against colistin-resistant Acinetobacter baumannii clinical isolates, Diagn Microbiol Infect Dis, vol.86, pp.184-193, 2016.

J. R. Lenhard, J. B. Bulitta, T. D. Connell, N. King-lyons, C. B. Landersdorfer et al., Highintensity meropenem combinations with polymyxin B: new strategies to overcome carbapenem resistance in Acinetobacter baumannii, J Antimicrob Chemother, vol.72, pp.153-65, 2017.

G. G. Rao, N. S. Ly, J. B. Bulitta, R. L. Soon, S. Roman et al., Polymyxin B in combination with doripenem against heteroresistant Acinetobacter baumannii: pharmacodynamics of new dosing strategies, J Antimicrob Chemother, vol.71, pp.3148-56, 2016.

T. Lim, T. Tan, W. Lee, S. Sasikala, T. Tan et al., In-Vitro Activity of Polymyxin B, Rifampicin, Tigecycline Alone and in Combination against Carbapenem-Resistant Acinetobacter baumannii in Singapore, PLOS ONE, vol.6, p.18485, 2011.

J. Y. Song, S. Y. Kee, I. S. Hwang, Y. B. Seo, H. W. Jeong et al., In vitro activities of carbapenem/sulbactam combination, colistin, colistin/rifampicin combination and tigecycline against carbapenem-resistant Acinetobacter baumannii, J Antimicrob Chemother, vol.60, pp.317-339, 2007.

S. Bae, M. Kim, S. Park, H. S. Kim, H. Sung et al., In Vitro Synergistic Activity of Antimicrobial Agents in Combination against Clinical Isolates of Colistin-Resistant Acinetobacter baumannii, Antimicrob Agents Chemother, vol.60, pp.6774-6783, 2016.

X. Cai, Z. Yang, J. Dai, K. Chen, L. Zhang et al., Pharmacodynamics of tigecycline alone and in combination with colistin against clinical isolates of multidrug-resistant Acinetobacter baumannii in an in vitro pharmacodynamic model, Int J Antimicrob Agents, vol.49, pp.609-625, 2017.

M. Dizbay, D. K. Tozlu, M. Y. Cirak, Y. Isik, K. Ozdemir et al., In vitro synergistic activity of tigecycline and colistin against XDR-Acinetobacter baumannii, J Antibiot (Tokyo), vol.63, pp.51-54, 2009.

G. G. Rao, N. S. Ly, J. Diep, A. Forrest, J. B. Bulitta et al., Combinatorial pharmacodynamics of polymyxin B and tigecycline against heteroresistant Acinetobacter baumannii, Int J Antimicrob Agents, vol.48, pp.331-337, 2016.

Y. Zhang, F. Chen, E. Sun, R. Ma, C. Qu et al., In vitro antibacterial activity of combinations of fosfomycin, minocycline and polymyxin B on pan-drug-resistant Acinetobacter baumannii, Exp Ther Med, vol.5, pp.1737-1746, 2013.

D. R. Bowers, H. Cao, J. Zhou, K. R. Ledesma, D. Sun et al., Assessment of Minocycline and Polymyxin B Combination against Acinetobacter baumannii, Antimicrob Agents Chemother, vol.59, p.2720, 2015.

R. López-rojas, M. J. Mcconnell, M. E. Jiménez-mejías, J. Domínguez-herrera, F. Fernández-cuenca et al., Colistin Resistance in a Clinical Acinetobacter baumannii Strain Appearing after Colistin Treatment: Effect on Virulence and Bacterial Fitness, Antimicrob Agents Chemother, vol.57, pp.4587-4596, 2013.

V. Aranzana-climent, A. Chauzy, N. Grégoire, S. Marchand, W. Couet et al., In vitro activity of polymyxin B alone and in combination against colistin-resistant Acinetobacter baumannii, 2019.

A. Bleibtreu, P. Gros, C. Laouénan, O. Clermont, L. Nagard et al., Stress Resistance, and Extraintestinal Virulence in Escherichia coli, Infect Immun, vol.81, pp.2733-2775, 2013.

S. G. Wicha, C. Chen, O. Clewe, and U. Simonsson, A general pharmacodynamic interaction model identifies perpetrators and victims in drug interactions, Nat Commun, vol.8, p.2129, 2017.

J. A. Orwa, C. Govaerts, K. Gevers, E. Roets, A. Van-schepdael et al., Study of the stability of polymyxins B1, E1 and E2 in aqueous solution using liquid chromatography and mass spectrometry, J Pharm Biomed Anal, vol.29, p.16, 2002.

, Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard-Ninth Edition. CLSI document M07-A9, 2012.

M. Jacobs, N. Grégoire, W. Couet, and J. B. Bulitta, Distinguishing Antimicrobial Models with Different Resistance Mechanisms via Population Pharmacodynamic Modeling, PLoS Comput Biol, vol.12, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02141647

A. Beceiro, E. Llobet, J. Aranda, J. A. Bengoechea, M. Doumith et al., Phosphoethanolamine Modification of Lipid A in Colistin-Resistant Variants of Acinetobacter baumannii Mediated by the pmrAB Two-Component Regulatory System, Antimicrob Agents Chemother, vol.55, pp.3370-3379, 2011.

J. Barin, A. F. Martins, B. L. Heineck, A. L. Barth, and A. P. Zavascki, Hetero-and adaptive resistance to polymyxin B in OXA-23-producing carbapenem-resistant Acinetobacter baumannii isolates, Ann Clin Microbiol Antimicrob, vol.12, p.15, 2013.

A. Skiada, A. Markogiannakis, D. Plachouras, and G. L. Daikos, Adaptive resistance to cationic compounds in Pseudomonas aeruginosa, Int J Antimicrob Agents, vol.37, pp.187-93, 2011.

D. J. Ritchie and A. Garavaglia-wilson, A Review of Intravenous Minocycline for Treatment of Multidrug-Resistant Acinetobacter Infections, Clin Infect Dis, vol.59, pp.374-80, 2014.

E. Comets, K. Brendel, and F. Mentré, Computing normalised prediction distribution errors to evaluate nonlinear mixed-effect models: the npde add-on package for R, Comput Methods Programs Biomed, vol.90, pp.154-66, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00274332

S. L. Beal, Ways to Fit a PK Model with Some Data Below the Quantification Limit, J Pharmacokinet Pharmacodyn, vol.28, pp.481-504, 2001.

. Xiii and . Références,

P. Courvalin, Predictable and unpredictable evolution of antibiotic resistance, Journal of Internal Medicine, vol.264, pp.4-16, 2008.

, The bacterial challenge, time to react: a call to narrow the gap between multidrug-resistant bacteria in the EU and the development of new antibacterial agents, European Centers for Disease Control, 2009.

, Antibiotic resistance threats in the United States, Centers for Disease Control and Prevention, 2013.

, Antimicrobial Resistance: Tackling a crisis for the health and wealth of nations, The Review on Antimicrobial Resistance, 2014.

, WHO | Global priority list of antibiotic-resistant bacteria to guide research, discovery, and development of new antibiotics, 2019.

C. L. Ventola, The Antibiotic Resistance Crisis, P T, vol.40, pp.277-83, 2015.

, FDA Approved Drugs in Infections and Infectious Diseases | CenterWatch n.d, vol.16, 2019.

R. L. Nation, J. Li, O. Cars, W. Couet, M. N. Dudley et al., Framework for optimisation of the clinical use of colistin and polymyxin B: the Prato polymyxin consensus, The Lancet Infectious Diseases, vol.15, pp.225-259, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02479001

H. Derendorf and B. Meibohm, Modeling of pharmacokinetic/pharmacodynamic (PK/PD) relationships: concepts and perspectives, Pharm Res, vol.16, pp.176-85, 1999.

, Determination of minimum inhibitory concentrations (MICs) of antibacterial agents by broth dilution, Clinical Microbiology and Infection, vol.9, 2003.

, Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard-Ninth Edition. CLSI document M07-A9, 2012.

, The European Committee on Antimicrobial Susceptibility Testing. Breakpoint tables for interpretation of MICs and zone diameters. Version 8.1, 2018.

, Performance standards for antimicrobial susceptibility testing: 24th informational supplement M100-S24 n.d, p.110

, M07-A10: Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically, p.110

D. D. Khan, L. E. Friberg, and E. I. Nielsen, A pharmacokinetic-pharmacodynamic (PKPD) model based on in vitro time-kill data predicts the in vivo PK/PD index of colistin, J Antimicrob Chemother, 2016.

J. W. Mouton, M. N. Dudley, O. Cars, H. Derendorf, and G. L. Drusano, Standardization of pharmacokinetic/pharmacodynamic (PK/PD) terminology for anti-infective drugs: an update, J Antimicrob Chemother, vol.55, pp.601-608, 2005.

D. Andes and W. A. Craig, In vivo pharmacodynamic activity of the glycopeptide dalbavancin, Antimicrob Agents Chemother, vol.51, pp.1633-1675, 2007.

A. Barbour, F. Scaglione, and H. Derendorf, Class-dependent relevance of tissue distribution in the interpretation of anti-infective pharmacokinetic/pharmacodynamic indices, Int J Antimicrob Agents, vol.35, pp.431-439, 2010.

D. S. Burgess and C. R. Frei, Comparison of beta-lactam regimens for the treatment of gram-negative pulmonary infections in the intensive care unit based on pharmacokinetics/pharmacodynamics, J Antimicrob Chemother, vol.56, pp.893-901, 2005.

A. Kashuba, A. N. Nafziger, G. L. Drusano, and J. S. Bertino, Optimizing Aminoglycoside Therapy for Nosocomial Pneumonia Caused by Gram-Negative Bacteria, Antimicrobial Agents and Chemotherapy, vol.43, pp.623-632, 1999.

J. Turnidge, Pharmacodynamics and dosing of aminoglycosides, Infect Dis Clin North Am, vol.17, pp.503-531, 2003.

B. Vogelman, S. Gudmundsson, J. Leggett, J. Turnidge, S. Ebert et al., Correlation of antimicrobial pharmacokinetic parameters with therapeutic efficacy in an animal model, J Infect Dis, vol.158, pp.831-878, 1988.

A. Forrest, S. Chodosh, M. A. Amantea, D. A. Collins, and J. J. Schentag, Pharmacokinetics and pharmacodynamics of oral grepafloxacin in patients with acute bacterial exacerbations of chronic bronchitis, J Antimicrob Chemother, vol.40, pp.45-57, 1997.

A. Forrest, D. E. Nix, C. H. Ballow, T. F. Goss, M. C. Birmingham et al., Pharmacodynamics of intravenous ciprofloxacin in seriously ill patients, Antimicrobial Agents and Chemotherapy, vol.37, pp.1073-81, 1993.

G. L. Drusano, S. L. Preston, C. Fowler, M. Corrado, B. Weisinger et al., Relationship between fluoroquinolone area under the curve: minimum inhibitory concentration ratio and the probability of eradication of the infecting pathogen, in patients with nosocomial pneumonia, J Infect Dis, vol.189, pp.1590-1597, 2004.

S. L. Preston, G. L. Drusano, A. L. Berman, C. L. Fowler, A. T. Chow et al., Pharmacodynamics of levofloxacin: a new paradigm for early clinical trials, JAMA, vol.279, pp.125-134, 1998.

C. R. Rayner, A. Forrest, A. K. Meagher, M. C. Birmingham, and J. J. Schentag, Clinical pharmacodynamics of linezolid in seriously ill patients treated in a compassionate use programme, Clin Pharmacokinet, vol.42, pp.1411-1434, 2003.

D. Andes, M. L. Ogtrop, . Van, J. Peng, and W. A. Craig, Vivo Pharmacodynamics of a New Oxazolidinone (Linezolid), vol.46, pp.3484-3493, 2002.

D. Ogtrop-ml-van,-andes, T. J. Stamstad, B. Conklin, W. J. Weiss, and W. A. Craig, In Vivo Pharmacodynamic Activities of Two Glycylcyclines (GAR-936 and WAY 152,288) against Various Gram-Positive and Gram-Negative Bacteria, Antimicrobial Agents and Chemotherapy, vol.44, pp.943-952, 2000.

A. K. Meagher, J. A. Passarell, B. B. Cirincione, S. Wart, K. Liolios et al., Exposure-Response Analyses of Tigecycline Efficacy in Patients with Complicated Skin and Skin-Structure Infections, Antimicrobial Agents and Chemotherapy, vol.51, pp.1939-1984, 2007.

J. A. Passarell, A. K. Meagher, K. Liolios, B. B. Cirincione, S. A. Van-wart et al., Exposure-response analyses of tigecycline efficacy in patients with complicated intraabdominal infections, Antimicrob Agents Chemother, vol.52, pp.204-214, 2008.

R. Jain and L. H. Danziger, The macrolide antibiotics: a pharmacokinetic and pharmacodynamic overview, Curr Pharm Des, vol.10, pp.3045-53, 2004.

D. Andes, J. Anon, M. R. Jacobs, and W. A. Craig, Application of pharmacokinetics and pharmacodynamics to antimicrobial therapy of respiratory tract infections, Clin Lab Med, vol.24, pp.477-502, 2004.

F. Van-bambeke and P. M. Tulkens, Macrolides: pharmacokinetics and pharmacodynamics, Int J Antimicrob Agents, vol.18, issue.1, pp.17-23, 2001.

J. G. Hollander, J. D. Knudsen, J. W. Mouton, K. Fuursted, N. Frimodt-møller et al., Comparison of pharmacodynamics of azithromycin and erythromycin in vitro and in vivo, Antimicrob Agents Chemother, vol.42, pp.377-82, 1998.

C. H. Nightingale, Pharmacokinetics and pharmacodynamics of newer macrolides, Pediatr Infect Dis J, vol.16, pp.438-481, 1997.

A. Novelli, S. Fallani, M. I. Cassetta, S. Arrigucci, and T. Mazzei, In vivo pharmacodynamic evaluation of clarithromycin in comparison to erythromycin, J Chemother, vol.14, pp.584-90, 2002.

J. Shi, G. Montay, and V. O. Bhargava, Clinical pharmacokinetics of telithromycin, the first ketolide antibacterial, Clin Pharmacokinet, vol.44, pp.915-949, 2005.

J. D. Knudsen, K. Fuursted, S. Raber, F. Espersen, and N. Frimodt-moller, Pharmacodynamics of glycopeptides in the mouse peritonitis model of Streptococcus pneumoniae or Staphylococcus aureus infection, Antimicrob Agents Chemother, vol.44, pp.1247-54, 2000.

P. A. Moise-broder, A. Forrest, M. C. Birmingham, and J. J. Schentag, Pharmacodynamics of vancomycin and other antimicrobials in patients with Staphylococcus aureus lower respiratory tract infections, Clin Pharmacokinet, vol.43, pp.925-967, 2004.

S. M. Bhavnani, J. A. Passarell, J. S. Owen, J. S. Loutit, S. B. Porter et al., Pharmacokineticpharmacodynamic relationships describing the efficacy of oritavancin in patients with Staphylococcus aureus bacteremia, Antimicrob Agents Chemother, vol.50, pp.994-1000, 2006.

C. J. Boylan, K. Campanale, P. W. Iversen, D. L. Phillips, M. L. Zeckel et al., Pharmacodynamics of oritavancin (LY333328) in a neutropenic-mouse thigh model of Staphylococcus aureus infection, Antimicrob Agents Chemother, vol.47, pp.1700-1706, 2003.

C. Li, J. L. Kuti, C. H. Nightingale, and D. P. Nicolau, Population pharmacokinetic analysis and dosing regimen optimization of meropenem in adult patients, J Clin Pharmacol, vol.46, pp.1171-1179, 2006.

A. N. Kristoffersson, P. David-pierson, N. J. Parrott, O. Kuhlmann, T. Lave et al., Simulation-Based Evaluation of PK/PD Indices for Meropenem Across Patient Groups and Experimental Designs, Pharm Res, vol.33, pp.1115-1140, 2016.

W. J. Jusko, Pharmacodynamics of chemotherapeutic effects: dose-time-response relationships for phase-nonspecific agents, J Pharm Sci, vol.60, pp.892-897, 1971.

J. J. Campion, P. Chung, P. J. Mcnamara, W. B. Titlow, and M. E. Evans, Pharmacodynamic modeling of the evolution of levofloxacin resistance in Staphylococcus aureus, Antimicrob Agents Chemother, vol.49, pp.2189-99, 2005.

A. K. Meagher, A. Forrest, A. Dalhoff, H. Stass, and J. J. Schentag, Novel pharmacokineticpharmacodynamic model for prediction of outcomes with an extended-release formulation of ciprofloxacin, Antimicrob Agents Chemother, vol.48, pp.2061-2069, 2004.

E. I. Nielsen, A. Viberg, E. Löwdin, O. Cars, M. O. Karlsson et al., Semimechanistic Pharmacokinetic/Pharmacodynamic Model for Assessment of Activity of Antibacterial Agents from Time-Kill Curve Experiments, Antimicrob Agents Chemother, vol.51, pp.128-164, 2007.

N. Q. Balaban, J. Merrin, R. Chait, L. Kowalik, and S. Leibler, Bacterial Persistence as a Phenotypic Switch, Science, vol.305, pp.1622-1627, 2004.

J. B. Bulitta, J. C. Yang, L. Yohonn, N. S. Ly, S. V. Brown et al., Attenuation of Colistin Bactericidal Activity by High Inoculum of Pseudomonas aeruginosa Characterized by a New Mechanism-Based Population Pharmacodynamic Model, Antimicrob Agents Chemother, vol.54, pp.2051-62, 2010.

L. Zhuang, S. Sy, H. Xia, R. P. Singh, M. B. Mulder et al., Evaluation of in vitro synergy between vertilmicin and ceftazidime against Pseudomonas aeruginosa using a semi-mechanistic pharmacokinetic/pharmacodynamic model, Int J Antimicrob Agents, vol.45, pp.151-60, 2015.

Y. Yano, T. Oguma, H. Nagata, and S. Sasaki, Application of logistic growth model to pharmacodynamic analysis of in vitro bactericidal kinetics, J Pharm Sci, vol.87, pp.1177-83, 1998.

V. H. Tam, A. N. Schilling, and M. Nikolaou, Modelling time-kill studies to discern the pharmacodynamics of meropenem, J Antimicrob Chemother, vol.55, pp.699-706, 2005.

A. F. Mohamed, O. Cars, and L. E. Friberg, A pharmacokinetic/pharmacodynamic model developed for the effect of colistin on Pseudomonas aeruginosa in vitro with evaluation of population pharmacokinetic variability on simulated bacterial killing, J Antimicrob Chemother, vol.69, pp.1350-61, 2014.

M. Jacobs, N. Gr?goire, W. Couet, and J. B. Bulitta, Distinguishing Antimicrobial Models with Different Resistance Mechanisms via Population Pharmacodynamic Modeling, PLOS Computational Biology, vol.12, p.1004782, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02141647

P. D. Tamma, S. E. Cosgrove, and L. L. Maragakis, Combination Therapy for Treatment of Infections with Gram-Negative Bacteria, Clinical Microbiology Reviews, vol.25, pp.450-70, 2012.

A. Kumar, R. Zarychanski, B. Light, J. Parrillo, D. Maki et al., Early combination antibiotic therapy yields improved survival compared with monotherapy in septic shock: a propensity-matched analysis, Crit Care Med, vol.38, pp.1773-85, 2010.

S. T. Micek, E. C. Welch, J. Khan, M. Pervez, J. A. Doherty et al., Empiric Combination Antibiotic Therapy Is Associated with Improved Outcome against Sepsis Due to Gram-Negative Bacteria: a Retrospective Analysis, Antimicrob Agents Chemother, vol.54, pp.1742-1750, 2010.

N. Petrosillo, M. Giannella, M. Antonelli, M. Antonini, B. Barsic et al., Clinical Experience of Colistin-Glycopeptide Combination in Critically Ill Patients Infected with Gram-Negative Bacteria, Antimicrob Agents Chemother, vol.58, pp.851-859, 2014.

T. Tängdén, Combination antibiotic therapy for multidrug-resistant Gram-negative bacteria, Ups J Med Sci, vol.119, pp.149-53, 2014.

M. C. Berenbaum, What is synergy?, Pharmacol Rev, vol.41, pp.93-141, 1989.

W. R. Greco, G. Bravo, and J. C. Parsons, The search for synergy: a critical review from a response surface perspective, Pharmacol Rev, vol.47, pp.331-85, 1995.

N. Geary, Understanding synergy, Am J Physiol Endocrinol Metab, vol.304, pp.237-253, 2013.

J. Foucquier and M. Guedj, Analysis of drug combinations: current methodological landscape, Pharmacol Res Perspect, vol.3, 2015.

E. Käer, S. Loewe, and . Über-kombinationswirkungen, Archiv f experiment Pathol u Pharmakol, vol.127, pp.308-326, 1928.

C. I. Bliss, The Toxicity of Poisons Applied Jointly1, Annals of Applied Biology, vol.26, pp.585-615, 1939.

Y. Grabovsky and R. J. Tallarida, Isobolographic analysis for combinations of a full and partial agonist: curved isoboles, J Pharmacol Exp Ther, vol.310, pp.981-987, 2004.

R. J. Tallarida, An overview of drug combination analysis with isobolograms, J Pharmacol Exp Ther, vol.319, pp.1-7, 2006.

M. Ezechiá? and T. Cajthaml, New insight into isobolographic analysis for combinations of a full and partial agonist: Curved isoboles, Toxicology, pp.402-403, 2018.

G. G. Rao, J. Li, S. M. Garonzik, R. L. Nation, and A. Forrest, Assessment and modelling of antibacterial combination regimens, Clin Microbiol Infect, vol.24, pp.689-96, 2018.

G. Orhan, A. Bayram, Y. Zer, and I. Balci, Synergy Tests by E Test and Checkerboard Methods of Antimicrobial Combinations against Brucella melitensis, Journal of Clinical Microbiology, vol.43, p.140, 2005.

, Clinical Microbiology Procedures Handbook, Fourth Edition. American Society of Microbiology, 2016.

F. C. Odds, Synergy, antagonism, and what the chequerboard puts between them, J Antimicrob Chemother, vol.52, pp.1-1, 2003.

M. H. Hsieh, C. M. Yu, V. L. Yu, and J. W. Chow, Synergy assessed by checkerboard a critical analysis, Diagnostic Microbiology and Infectious Disease, vol.16, issue.93, p.90087, 1993.

K. H. Rand, H. J. Houck, P. Brown, and D. Bennett, Reproducibility of the microdilution checkerboard method for antibiotic synergy, Antimicrob Agents Chemother, vol.37, pp.613-618, 1993.

J. W. Mouton, D. Ogtrop-ml-van,-andes, and W. A. Craig, Use of Pharmacodynamic Indices To Predict Efficacy of Combination Therapy In Vivo, Antimicrobial Agents and Chemotherapy, vol.43, pp.2473-2481, 1999.

J. J. Schentag, L. C. Strenkoski-nix, D. E. Nix, and A. Forrest, Pharmacodynamic Interactions of Antibiotics Alone and in Combination, Clin Infect Dis, vol.27, pp.40-46, 1998.

. Hollander-jg-den, J. W. Mouton, and H. A. Verbrugh, Use of Pharmacodynamic Parameters To Predict Efficacy of Combination Therapy by Using Fractional Inhibitory Concentration Kinetics, Antimicrobial Agents and Chemotherapy, vol.42, pp.744-752, 1998.

M. Brill, A. N. Kristoffersson, C. Zhao, E. I. Nielsen, and L. E. Friberg, Semi-mechanistic pharmacokinetic-pharmacodynamic modelling of antibiotic drug combinations, Clinical Microbiology and Infection, vol.0, 2017.

W. R. Greco, H. S. Park, and Y. M. Rustum, Application of a New Approach for the Quantitation of Drug Synergism to the Combination of cis-Diamminedichloroplatinum and 1-?-d-Arabinofuranosylcytosine, Cancer Res, vol.50, pp.5318-5345, 1990.

G. L. Drusano, M. Neely, M. V. Guilder, A. Schumitzky, D. Brown et al., Analysis of Combination Drug Therapy to Develop Regimens with Shortened Duration of Treatment for Tuberculosis, PLOS ONE, vol.9, p.101311, 2014.

S. Wicha, W. Huisinga, and C. Kloft, Translational Pharmacometric Evaluation of Typical Antibiotic Broad-Spectrum Combination Therapies Against Staphylococcus Aureus Exploiting In Vitro Information, CPT Pharmacometrics Syst Pharmacol, vol.6, pp.512-534, 2017.

G. G. Rao, N. S. Ly, J. B. Bulitta, R. L. Soon, S. Roman et al., Polymyxin B in combination with doripenem against heteroresistant Acinetobacter baumannii: pharmacodynamics of new dosing strategies, J Antimicrob Chemother, vol.71, pp.3148-56, 2016.

R. Yadav, C. B. Landersdorfer, R. L. Nation, J. D. Boyce, and J. B. Bulitta, Novel Approach To Optimize Synergistic Carbapenem-Aminoglycoside Combinations against Carbapenem-Resistant Acinetobacter baumannii, Antimicrob Agents Chemother, vol.59, pp.2286-98, 2015.

C. B. Landersdorfer, N. S. Ly, H. Xu, B. T. Tsuji, and J. B. Bulitta, Quantifying Subpopulation Synergy for Antibiotic Combinations via Mechanism-Based Modeling and a Sequential Dosing Design, Antimicrobial Agents and Chemotherapy, vol.57, pp.2343-51, 2013.

N. S. Ly, J. B. Bulitta, G. G. Rao, C. B. Landersdorfer, P. N. Holden et al., Colistin and doripenem combinations against Pseudomonas aeruginosa: profiling the time course of synergistic killing and prevention of resistance, J Antimicrob Chemother, vol.70, pp.1434-1476, 2015.

N. S. Ly, Z. P. Bulman, J. B. Bulitta, C. Baron, G. G. Rao et al., Optimization of Polymyxin B in Combination with Doripenem To Combat Mutator Pseudomonas aeruginosa, Antimicrobial Agents and Chemotherapy, vol.60, pp.2870-80, 2016.

J. R. Lenhard, J. B. Bulitta, T. D. Connell, N. King-lyons, C. B. Landersdorfer et al., Highintensity meropenem combinations with polymyxin B: new strategies to overcome carbapenem resistance in Acinetobacter baumannii, J Antimicrob Chemother, vol.72, pp.153-65, 2017.

S. Sy, L. Zhuang, H. Xia, M. Beaudoin, V. J. Schuck et al., Prediction of in vivo and in vitro infection model results using a semimechanistic model of avibactam and aztreonam combination against multidrug resistant organisms, CPT: Pharmacometrics & Systems Pharmacology, vol.6, pp.197-207, 2017.

A. F. Mohamed, A. N. Kristoffersson, M. Karvanen, E. I. Nielsen, O. Cars et al., Dynamic interaction of colistin and meropenem on a WT and a resistant strain of Pseudomonas aeruginosa as quantified in a PK/PD model, J Antimicrob Chemother, 2016.

S. G. Wicha, C. Chen, O. Clewe, and U. Simonsson, A general pharmacodynamic interaction model identifies perpetrators and victims in drug interactions, Nature Communications, vol.8, p.2129, 2017.

M. Lee, W. Sheng, C. Hung, C. Yu, L. Lee et al., Mycobacterium abscessus Complex Infections in Humans, Emerging Infect Dis, vol.21, pp.1638-1684, 2015.

S. Teng, C. Chen, M. Lee, T. Lee, K. Chien et al., Matrix-Assisted Laser Desorption Ionization-Time of Flight Mass Spectrometry Can Accurately Differentiate between Mycobacterium masilliense (M. abscessus subspecies bolletti) and M. abscessus (Sensu Stricto), J Clin Microbiol, vol.51, pp.3113-3119, 2013.

J. Benwill and R. Wallace, Mycobacterium abscessus: challenges in diagnosis and treatment, Current Opinion in Infectious Diseases, vol.27, pp.506-516, 2014.

D. R. Prevots, P. A. Shaw, D. Strickland, L. A. Jackson, M. A. Raebel et al., Nontuberculous Mycobacterial Lung Disease Prevalence at Four Integrated Health Care Delivery Systems, Am J Respir Crit Care Med, vol.182, pp.970-976, 2010.

C. Lai, C. Tan, C. Chou, H. Hsu, C. Liao et al., Increasing Incidence of Nontuberculous Mycobacteria, Emerg Infect Dis, vol.16, pp.294-300, 2000.

M. Lee, W. Sheng, C. Hung, C. Yu, L. Lee et al., Complex Infections in Humans. Emerg Infect Dis, vol.21, pp.1638-1684, 2015.

F. Mougari, L. Guglielmetti, L. Raskine, I. Sermet-gaudelus, N. Veziris et al., Infections caused by Mycobacterium abscessus: epidemiology, diagnostic tools and treatment, Expert Rev Anti Infect Ther, vol.14, pp.1139-54, 2016.

R. Nessar, E. Cambau, J. M. Reyrat, A. Murray, and B. Gicquel, Mycobacterium abscessus: a new antibiotic nightmare, J Antimicrob Chemother, vol.67, pp.810-818, 2012.

M. Sfeir, M. Walsh, R. Rosa, L. Aragon, S. Y. Liu et al., Mycobacterium abscessus Complex Infections: A Retrospective Cohort Study, Open Forum Infect Dis, vol.5, p.22, 2018.

B. E. Ferro, J. Van-ingen, M. Wattenberg, D. Van-soolingen, and J. W. Mouton, Time-kill kinetics of antibiotics active against rapidly growing mycobacteria, J Antimicrob Chemother, vol.70, pp.811-818, 2015.

R. Greendyke and T. F. Byrd, Differential antibiotic susceptibility of Mycobacterium abscessus variants in biofilms and macrophages compared to that of planktonic bacteria, Antimicrob Agents Chemother, vol.52, pp.2019-2045, 2008.

A. Lefebvre, V. Dubée, M. Cortes, D. Dorchêne, M. Arthur et al., Bactericidal and intracellular activity of ?-lactams against Mycobacterium abscessus, J Antimicrob Chemother, vol.71, pp.1556-63, 2016.

. Cefoxitin and . Wikipedia, , 2019.

E. Story-roller, E. C. Maggioncalda, K. A. Cohen, and G. Lamichhane, Mycobacterium abscessus and ?-Lactams: Emerging Insights and Potential Opportunities, Front Microbiol, vol.9, 2018.

M. L. Grayson, S. E. Cosgrove, S. Crowe, W. Hope, J. S. Mccarthy et al., The Use of Antibiotics : A Clinical Review of Antibacterial, Antifungal, Antiparasitic, and Antiviral Drugs, Seventh Edition -Three Volume Set, 2017.

G. A. Jacoby and G. L. Archer, New Mechanisms of Bacterial Resistance to Antimicrobial Agents, New England Journal of Medicine, vol.324, pp.601-613, 1991.

G. A. Jacoby, AmpC beta-lactamases, Table of Contents, vol.22, pp.161-82, 2009.

C. C. Sanders and W. E. Sanders, Emergence of resistance to cefamandole: possible role of cefoxitininducible beta-lactamases, Antimicrob Agents Chemother, vol.15, pp.792-799, 1979.

, WHO | Global action plan on AMR. WHO n.d, 2019.

, G7 Health Ministers Declaration n

, Milan_2017_0/index.pdf (accessed July, vol.5, 2019.

J. About, , 2019.

J. Second and . Call, , 2019.

. Co-action, Developing combinations of CO-ACTIVE and non-antimicrobials n, 2019.

T. Velkov, K. D. Roberts, R. L. Nation, P. E. Thompson, and J. Li, Pharmacology of polymyxins: new insights into an 'old' class of antibiotics, Future Microbiol, vol.8, 2013.

L. Poirel, A. Jayol, and P. Nordmann, Polymyxins: Antibacterial Activity, Susceptibility Testing, and Resistance Mechanisms Encoded by Plasmids or Chromosomes, Clin Microbiol Rev, vol.30, pp.557-96, 2017.

R. L. Nation, T. Velkov, L. J. Colistin, and B. Polymyxin, Peas in a Pod, or Chalk and Cheese?, Clin Infect Dis, vol.59, pp.88-94, 2014.

P. J. Bergen, J. Li, C. R. Rayner, and R. L. Nation, Colistin Methanesulfonate Is an Inactive Prodrug of Colistin against Pseudomonas aeruginosa, Antimicrob Agents Chemother, vol.50, pp.1953-1961, 2006.

R. Aggarwal and A. Dewan, Comparison of nephrotoxicity of Colistin with Polymyxin B administered in currently recommended doses: a prospective study, Annals of Clinical Microbiology and Antimicrobials, vol.17, p.15, 2018.

A. P. Zavascki and R. L. Nation, Nephrotoxicity of Polymyxins: Is There Any Difference between Colistimethate and Polymyxin B?, Antimicrob Agents Chemother, p.61, 2017.

M. H. Rigatto, M. S. Oliveira, L. V. Perdigão-neto, A. S. Levin, C. M. Carrilho et al., Multicenter Prospective Cohort Study of Renal Failure in Patients Treated with Colistin versus Polymyxin B, Antimicrob Agents Chemother, vol.60, pp.2443-2452, 2016.

A. M. Sandri, C. B. Landersdorfer, J. Jacob, M. M. Boniatti, M. G. Dalarosa et al., Population Pharmacokinetics of Intravenous Polymyxin B in Critically Ill Patients: Implications for Selection of Dosage Regimens, Clin Infect Dis, vol.57, pp.524-555, 2013.

M. J. Mcconnell, L. Actis, and J. Pachón, Acinetobacter baumannii: human infections, factors contributing to pathogenesis and animal models, FEMS Microbiol Rev, vol.37, pp.130-55, 2013.

L. Dijkshoorn, A. Nemec, and H. Seifert, An increasing threat in hospitals: multidrug-resistant Acinetobacter baumannii, Nature Reviews Microbiology, vol.5, pp.939-51, 2007.

J. Y. Fagon, J. Chastre, Y. Domart, J. L. Trouillet, and C. Gibert, Mortality due to ventilator-associated pneumonia or colonization with Pseudomonas or Acinetobacter species: assessment by quantitative culture of samples obtained by a protected specimen brush, Clin Infect Dis, vol.23, pp.538-580, 1996.

J. Garnacho, J. Sole-violan, M. Sa-borges, E. Diaz, and J. Rello, Clinical impact of pneumonia caused by Acinetobacter baumannii in intubated patients: a matched cohort study, Crit Care Med, vol.31, pp.2478-82, 2003.

M. E. Falagas, P. Kopterides, and I. I. Siempos, Attributable mortality of Acinetobacter baumannii infection among critically ill patients, Clin Infect Dis, vol.43, p.389, 2006.

A. Abbo, Y. Carmeli, S. Navon-venezia, Y. Siegman-igra, and M. J. Schwaber, Impact of multi-drugresistant Acinetobacter baumannii on clinical outcomes, Eur J Clin Microbiol Infect Dis, vol.26, pp.793-800, 2007.

M. E. Falagas and P. I. Rafailidis, Attributable mortality of Acinetobacter baumannii: no longer a controversial issue, Crit Care, vol.11, p.134, 2007.

N. Lee, H. Lee, N. Ko, C. Chang, H. Shih et al., Clinical and economic impact of multidrug resistance in nosocomial Acinetobacter baumannii bacteremia, Infect Control Hosp Epidemiol, vol.28, pp.713-722, 2007.

H. Wisplinghoff, T. Bischoff, S. M. Tallent, H. Seifert, R. P. Wenzel et al., Nosocomial bloodstream infections in US hospitals: analysis of 24,179 cases from a prospective nationwide surveillance study, Clin Infect Dis, vol.39, pp.309-326, 2004.

J. L. García-garmendia, C. Ortiz-leyba, J. Garnacho-montero, F. J. Jiménez-jiménez, J. Monterrubio-villar et al., Mortality and the increase in length of stay attributable to the acquisition of Acinetobacter in critically ill patients, Crit Care Med, vol.27, pp.1794-1803, 1999.

T. Jang, S. Lee, C. Huang, C. Lee, and W. Chen, Risk factors and impact of nosocomial Acinetobacter baumannii bloodstream infections in the adult intensive care unit: a case-control study, J Hosp Infect, vol.73, pp.143-50, 2009.

J. Y. Jung, M. S. Park, S. E. Kim, B. H. Park, J. Y. Son et al., Risk factors for multi-drug resistant Acinetobacter baumannii bacteremia in patients with colonization in the intensive care unit, BMC Infect Dis, vol.10, p.228, 2010.

H. Seifert, A. Strate, and G. Pulverer, Nosocomial bacteremia due to Acinetobacter baumannii. Clinical features, epidemiology, and predictors of mortality, Medicine (Baltimore), vol.74, pp.340-349, 1995.

P. Fournier, D. Vallenet, V. Barbe, S. Audic, H. Ogata et al., Comparative Genomics of Multidrug Resistance in Acinetobacter baumannii, PLOS Genetics, vol.2, p.7, 2006.

M. Iacono, L. Villa, D. Fortini, R. Bordoni, F. Imperi et al., Whole-genome pyrosequencing of an epidemic multidrug-resistant Acinetobacter baumannii strain belonging to the European clone II group, Antimicrob Agents Chemother, vol.52, pp.2616-2641, 2008.

D. W. Wareham, D. C. Bean, P. Khanna, E. M. Hennessy, D. Krahe et al., Bloodstream infection due to Acinetobacter spp: epidemiology, risk factors and impact of multi-drug resistance, Eur J Clin Microbiol Infect Dis, vol.27, pp.607-619, 2008.

R. López-rojas, M. J. Mcconnell, M. E. Jiménez-mejías, J. Domínguez-herrera, F. Fernández-cuenca et al., Colistin Resistance in a Clinical Acinetobacter baumannii Strain Appearing after Colistin Treatment: Effect on Virulence and Bacterial Fitness, Antimicrob Agents Chemother, vol.57, pp.4587-4596, 2013.

N. Jaidane, T. Naas, W. Mansour, B. B. Radhia, S. Jerbi et al., Genomic analysis of in vivo acquired resistance to colistin and rifampicin in Acinetobacter baumannii, Int J Antimicrob Agents, vol.51, pp.266-275, 2018.

S. Pournaras, A. Poulou, K. Dafopoulou, Y. N. Chabane, I. Kristo et al., Growth Retardation, Reduced Invasiveness, and Impaired Colistin-Mediated Cell Death Associated with Colistin Resistance Development in Acinetobacter baumannii, Antimicrob Agents Chemother, vol.58, pp.828-860, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02334071

J. Rolain, S. M. Diene, M. Kempf, G. Gimenez, C. Robert et al., Real-Time Sequencing To Decipher the Molecular Mechanism of Resistance of a Clinical Pan-Drug-Resistant Acinetobacter baumannii Isolate from Marseille, France, Antimicrobial Agents and Chemotherapy, vol.57, pp.592-598, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01389302

D. J. Ritchie and A. Garavaglia-wilson, A review of intravenous minocycline for treatment of multidrug-resistant Acinetobacter infections, Clin Infect Dis, vol.59, issue.6, pp.374-380, 2014.

E. Bishburg and K. Bishburg, Minocycline--an old drug for a new century: emphasis on methicillinresistant Staphylococcus aureus (MRSA) and Acinetobacter baumannii, Int J Antimicrob Agents, vol.34, pp.395-401, 2009.

. Minocycline and . Wikipedia, , 2019.

S. L. Greig and L. J. Scott, Intravenous Minocycline: A Review in Acinetobacter Infections, Drugs, vol.76, pp.1467-76, 2016.

. Fda and . Minocin--minocycline, , 2019.

O. Lomovskaya, D. Sun, D. Rubio-aparicio, K. J. Nelson, V. Thamlikitkul et al., Absence of TetB Identifies Minocycline-Susceptible Isolates of Acinetobacter baumannii, International Journal of Antimicrobial Agents, vol.0, 2018.

L. Antunes, P. Visca, and K. J. Towner, Acinetobacter baumannii: evolution of a global pathogen, Pathog Dis, vol.71, pp.292-301, 2014.

E. I. Nielsen and L. E. Friberg, Pharmacokinetic-pharmacodynamic modeling of antibacterial drugs, Pharmacol Rev, vol.65, pp.1053-90, 2013.

D. J. Hong, J. O. Kim, H. Lee, E. Yoon, S. H. Jeong et al., In vitro antimicrobial synergy of colistin with rifampicin and carbapenems against colistin-resistant Acinetobacter baumannii clinical isolates. Diagnostic Microbiology and Infectious Disease, vol.86, pp.184-193, 2016.

T. Lim, T. Tan, W. Lee, S. Sasikala, T. Tan et al., In-Vitro Activity of Polymyxin B, Rifampicin, Tigecycline Alone and in Combination against Carbapenem-Resistant Acinetobacter baumannii in Singapore, PLOS ONE, vol.6, p.18485, 2011.

J. Y. Song, S. Y. Kee, I. S. Hwang, Y. B. Seo, H. W. Jeong et al., In vitro activities of carbapenem/sulbactam combination, colistin, colistin/rifampicin combination and tigecycline against carbapenem-resistant Acinetobacter baumannii, J Antimicrob Chemother, vol.60, pp.317-339, 2007.

S. Bae, M. Kim, S. Park, H. S. Kim, H. Sung et al., In Vitro Synergistic Activity of Antimicrobial Agents in Combination against Clinical Isolates of Colistin-Resistant Acinetobacter baumannii, Antimicrob Agents Chemother, vol.60, pp.6774-6783, 2016.

X. Cai, Z. Yang, J. Dai, K. Chen, L. Zhang et al., Pharmacodynamics of tigecycline alone and in combination with colistin against clinical isolates of multidrug-resistant Acinetobacter baumannii in an in vitro pharmacodynamic model, Int J Antimicrob Agents, vol.49, pp.609-625, 2017.

M. Dizbay, D. K. Tozlu, M. Y. Cirak, Y. Isik, K. Ozdemir et al., In vitro synergistic activity of tigecycline and colistin against XDR-Acinetobacter baumannii, J Antibiot, vol.63, pp.51-54, 2009.

G. G. Rao, N. S. Ly, J. Diep, A. Forrest, J. B. Bulitta et al., Combinatorial pharmacodynamics of polymyxin B and tigecycline against heteroresistant Acinetobacter baumannii, Int J Antimicrob Agents, vol.48, pp.331-337, 2016.

Y. Zhang, F. Chen, E. Sun, R. Ma, C. Qu et al., In vitro antibacterial activity of combinations of fosfomycin, minocycline and polymyxin B on pan-drug-resistant Acinetobacter baumannii, Exp Ther Med, vol.5, pp.1737-1746, 2013.

D. R. Bowers, H. Cao, J. Zhou, K. R. Ledesma, D. Sun et al., Assessment of Minocycline and Polymyxin B Combination against Acinetobacter baumannii, Antimicrobial Agents and Chemotherapy, vol.59, p.2720, 2015.

A. Bleibtreu, P. Gros, C. Laouénan, O. Clermont, L. Nagard et al., Stress Resistance, and Extraintestinal Virulence in Escherichia coli, Infect Immun, vol.81, pp.2733-2775, 2013.

J. A. Orwa, C. Govaerts, K. Gevers, E. Roets, A. Van-schepdael et al., Study of the stability of polymyxins B1, E1 and E2 in aqueous solution using liquid chromatography and mass spectrometry, Journal of Pharmaceutical and Biomedical Analysis, vol.29, pp.203-215, 2002.

M. Jacobs, N. Grégoire, W. Couet, and J. B. Bulitta, Distinguishing Antimicrobial Models with Different Resistance Mechanisms via Population Pharmacodynamic Modeling, PLoS Comput Biol, vol.12, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02141647

A. Beceiro, E. Llobet, J. Aranda, J. A. Bengoechea, M. Doumith et al., Phosphoethanolamine Modification of Lipid A in Colistin-Resistant Variants of Acinetobacter baumannii Mediated by the pmrAB Two-Component Regulatory System, Antimicrobial Agents and Chemotherapy, vol.55, pp.3370-3379, 2011.

J. Barin, A. F. Martins, B. L. Heineck, A. L. Barth, and A. P. Zavascki, Hetero-and adaptive resistance to polymyxin B in OXA-23-producing carbapenem-resistant Acinetobacter baumannii isolates, Ann Clin Microbiol Antimicrob, vol.12, p.15, 2013.

A. Skiada, A. Markogiannakis, D. Plachouras, and G. L. Daikos, Adaptive resistance to cationic compounds in Pseudomonas aeruginosa, Int J Antimicrob Agents, vol.37, pp.187-93, 2011.

D. J. Ritchie and A. Garavaglia-wilson, A Review of Intravenous Minocycline for Treatment of Multidrug-Resistant Acinetobacter Infections, Clin Infect Dis, vol.59, pp.374-80, 2014.

E. Comets, K. Brendel, and F. Mentré, Computing normalised prediction distribution errors to evaluate nonlinear mixed-effect models: the npde add-on package for R, Comput Methods Programs Biomed, vol.90, pp.154-66, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00274332

S. L. Beal, Ways to Fit a PK Model with Some Data Below the Quantification Limit, J Pharmacokinet Pharmacodyn, vol.28, pp.481-504, 2001.

R. H. Deurenberg, E. Bathoorn, M. A. Chlebowicz, N. Couto, M. Ferdous et al., Application of next generation sequencing in clinical microbiology and infection prevention, Journal of Biotechnology, vol.243, pp.16-24, 2017.

Z. Wang, M. Gerstein, and M. Snyder, RNA-Seq: a revolutionary tool for transcriptomics, Nat Rev Genet, vol.10, pp.57-63, 2009.

M. R. Pulido, M. García-quintanilla, M. L. Gil-marqués, and M. J. Mcconnell, Identifying targets for antibiotic development using omics technologies, Drug Discov Today, vol.21, pp.465-72, 2016.

I. Vranakis, I. Goniotakis, A. Psaroulaki, V. Sandalakis, Y. Tselentis et al., Proteome studies of bacterial antibiotic resistance mechanisms, J Proteomics, vol.97, pp.88-99, 2014.

C. Freiberg, H. Brötz-oesterhelt, and H. Labischinski, The impact of transcriptome and proteome analyses on antibiotic drug discovery, Curr Opin Microbiol, vol.7, pp.451-460, 2004.

R. Da-cunha, B. Fonseca, L. P. Calado, and C. , Antibiotic Discovery: Where Have We Come from, Where Do We Go?, Antibiotics (Basel), vol.8, 2019.

Y. Zhu, J. Zhao, M. Maifiah, T. Velkov, F. Schreiber et al., Metabolic Responses to Polymyxin Treatment in Acinetobacter baumannii ATCC 19606: Integrating Transcriptomics and Metabolomics with Genome-Scale Metabolic Modeling, MSystems, vol.4, pp.157-175, 2019.

N. Benson, Quantitative Systems Pharmacology and Empirical Models: Friends or Foes?, CPT: Pharmacometrics & Systems Pharmacology, vol.8, pp.135-142, 2019.

M. Ayrapetyan, T. C. Williams, and J. D. Oliver, Bridging the gap between viable but non-culturable and antibiotic persistent bacteria, Trends Microbiol, vol.23, pp.7-13, 2015.

W. Ungphakorn, P. Lagerbäck, E. I. Nielsen, and T. Tängdén, Automated time-lapse microscopy a novel method for screening of antibiotic combination effects against multidrug-resistant Gramnegative bacteria, Clin Microbiol Infect, vol.24, 2018.

, Pharmacodynamics of polymyxin B against extensively drug-resistant <em>Acinetobacter baumannii </em>in a hollow fibre infection model -ECCMID Live n, 2019.

J. M. Buyck, P. Plésiat, H. Traore, F. Vanderbist, P. M. Tulkens et al., Increased susceptibility of Pseudomonas aeruginosa to macrolides and ketolides in eukaryotic cell culture media and biological fluids due to decreased expression of oprM and increased outermembrane permeability, Clin Infect Dis, vol.55, pp.534-576, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01695413

J. X. Huang, M. Blaskovich, R. Pelingon, S. Ramu, A. Kavanagh et al., Mucin Binding Reduces Colistin Antimicrobial Activity, Antimicrob Agents Chemother, vol.59, pp.5925-5956, 2015.

C. Butnarasu, N. Barbero, D. Pacheco, P. Petrini, and S. Visentin, Mucin binding to therapeutic molecules: The case of antimicrobial agents used in cystic fibrosis, Int J Pharm, vol.564, pp.136-180, 2019.

S. Kirchner, J. L. Fothergill, E. A. Wright, C. E. James, E. Mowat et al., Use of Artificial Sputum Medium to Test Antibiotic Efficacy Against Pseudomonas aeruginosa in Conditions More Relevant to the Cystic Fibrosis Lung, Journal of Visualized Experiments : JoVE, 2012.

J. R. Lenhard, N. M. Smith, Z. P. Bulman, X. Tao, V. Thamlikitkul et al., High-Dose Ampicillin-Sulbactam Combinations Combat Polymyxin-Resistant Acinetobacter baumannii in a Hollow-Fiber Infection Model, Antimicrob Agents Chemother, vol.61, pp.1268-1284, 2017.

S. Pichereau, M. Pantrangi, W. Couet, C. Badiou, G. Lina et al., Simulated antibiotic exposures in an in vitro hollow-fiber infection model influence toxin gene expression and production in community-associated methicillin-resistant Staphylococcus aureus strain MW2, Antimicrob Agents Chemother, vol.56, pp.140-147, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02474493

Y. Aoki, M. Sundqvist, A. C. Hooker, and P. Gennemark, PopED lite: An optimal design software for preclinical pharmacokinetic and pharmacodynamic studies, Computer Methods and Programs in Biomedicine, vol.127, pp.126-169, 2016.