H. Herzog and B. , History of Tuberculosis. RES, vol.65, issue.1, pp.5-15, 1998.

J. W. Mcgrath, Social Networks of Disease Spread in the Lower Illinois Valley: A Simulation Approach, Am. J. Phys. Anthropol, vol.77, issue.4, pp.483-496, 1988.

, The Arrow of Disease | DiscoverMagazine, 2018.

J. F. Murray, The White Plague: Down and Out, or Up and Coming?, J. Burns Amberson Lecture. American Review of Respiratory Disease, vol.140, pp.1788-1795, 1989.

, A sickly young woman sits covered up on a balcony; death (a ghostly skeleton clutching a scythe and an hourglass) is standing next to her; representing tuberculosis, p.31, 1912.

L. Dû and D. , Raccourcissement de La Durée de Traitement de La Tuberculose, Journal des Antiinfectieux, vol.2015, issue.3, pp.99-109

J. R. Glynn, Resurgence of Tuberculosis and the Impact of HIV Infection, Br. Med. Bull, vol.54, issue.3, pp.579-593, 1998.

, WHO | Global tuberculosis report, 2018.

, Comparative analysis of volatile organic compounds for the classification and identification of mycobacterial species, 2018.

J. I. Van, Z. Rahim, A. Mulder, M. J. Boeree, R. Simeone et al., Characterization of Mycobacterium Orygis as M. Tuberculosis Complex Subspecies., Characterization of Mycobacterium Orygis as M. Tuberculosis Complex Subspecies. Emerg Infect Dis, vol.18, pp.653-655, 2012.

P. J. Bishop and G. Neumann, The History of the Ziehl-Neelsen Stain, Tubercle, vol.1970, issue.2, pp.196-206

, Coloration de Ziehl-Neelsen. Wikipédia, 2018.

A. M. Cooper, Cell-Mediated Immune Responses in Tuberculosis, Annual Review of Immunology, vol.27, issue.1, pp.393-422, 2009.

P. J. Brennan and H. Nikaido, The Envelope of Mycobacteria, Annu. Rev. Biochem, vol.64, pp.29-63, 1995.

V. Jarlier and H. Nikaido, Mycobacterial Cell Wall: Structure and Role in Natural Resistance to Antibiotics, FEMS Microbiol. Lett, vol.123, issue.1-2, pp.11-18, 1994.

D. C. Crick, S. Mahapatra, and P. J. Brennan, Biosynthesis of the Arabinogalactan-Peptidoglycan Complex of Mycobacterium Tuberculosis, Glycobiology, vol.11, issue.9, pp.107-118, 2001.

, Mycobacteria Cell Wall: Mycobacteria Powerpoint Presentation -Articleblog, 2019.

P. J. Brennan, Structure, Function, and Biogenesis of the Cell Wall of Mycobacterium Tuberculosis, Tuberculosis (Edinb), vol.83, issue.1-3, pp.91-97, 2003.

K. J. Kieser, C. Baranowski, M. C. Chao, J. E. Long, C. M. Sassetti et al., Peptidoglycan Synthesis in Mycobacterium Tuberculosis Is Organized into Networks with Varying Drug Susceptibility, Proc. Natl. Acad. Sci. U.S.A, vol.112, issue.42, pp.13087-13092, 2015.

R. Veyron-churlet, S. Bigot, O. Guerrini, S. Verdoux, W. Malaga et al., The Biosynthesis of Mycolic Acids in Mycobacterium Tuberculosis Relies on Multiple Specialized Elongation Complexes Interconnected by Specific Protein-Protein Interactions, Journal of Molecular Biology, vol.353, issue.4, pp.847-858, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01883772

H. Marrakchi, M. Lanéelle, and M. Daffé, Mycolic Acids: Structures, Biosynthesis, and Beyond, vol.21, pp.67-85, 2014.

M. Jankute, J. A. Cox, J. Harrison, and G. S. Besra, Assembly of the Mycobacterial Cell Wall, Annu. Rev. Microbiol, vol.69, pp.405-423, 2015.

E. Schweizer and J. Hofmann, Microbial Type I Fatty Acid Synthases (FAS): Major Players in a, Network of Cellular FAS Systems. Microbiol. Mol. Biol. Rev, vol.68, issue.3, pp.501-517, 2004.

A. Bhatt, V. Molle, G. S. Besra, W. R. Jacobs, and L. Kremer, The Mycobacterium Tuberculosis FAS-II Condensing Enzymes: Their Role in Mycolic Acid Biosynthesis, Acid-Fastness, Pathogenesis and in Future Drug Development, Mol. Microbiol, vol.64, issue.6, pp.1442-1454, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02282906

T. Parish, G. Roberts, F. Laval, M. Schaeffer, M. Daffé et al., Functional Complementation of the Essential Gene FabG1 of Mycobacterium Tuberculosis by Mycobacterium Smegmatis FabG but Not Escherichia Coli FabG, J. Bacteriol, vol.189, issue.10, pp.3721-3728, 2007.

G. Poncet-montange, S. Ducasse-cabanot, A. Quemard, G. Labesse, and M. Cohen-gonsaud, Lack of Dynamics in the MabA Active Site Kills the Enzyme Activity: Practical Consequences for Drug-Design Studies, Acta Crystallogr. D Biol. Crystallogr, vol.63, pp.923-925, 2007.

R. Biswas, A. Dutta, D. Dutta, D. Hazra, D. R. Banerjee et al., Crystal Structure of Dehydratase Component HadAB Complex of Mycobacterial FAS-II Pathway, Biochem. Biophys. Res. Commun, vol.458, issue.2, pp.369-374, 2015.

S. Gavalda, M. Léger, B. Van-der-rest, A. Stella, F. Bardou et al., The Pks13/FadD32 Crosstalk for the Biosynthesis of Mycolic Acids in Mycobacterium Tuberculosis, J. Biol. Chem, vol.284, issue.29, pp.19255-19264, 2009.

M. Léger, S. Gavalda, V. Guillet, B. Van-der-rest, N. Slama et al., The Dual Function of the Mycobacterium Tuberculosis FadD32 Required for Mycolic Acid Biosynthesis, Chem. Biol, vol.16, issue.5, pp.510-519, 2009.

, Aspects cliniques et thérapeutiques de la tuberculose chez l'enfant et l'adulte -ScienceDirect, vol.27, 2018.

, Who puts the tubercle in tuberculosis?, Nature Reviews Microbiology, vol.24, 2018.

. Oms-|-rapport, , 2018.

, Tuberculose extrapulmonaire -Maladies infectieuses, 2018.

B. Fedoul, K. Chakour, M. E. Chaoui, and . Le, Mal de Pott: À Propos de 82 Cas, Pan Afr Med J, vol.8, 2011.

. History, |. Vaccine, . Calmette, and L. Guerin, TB Facts | TB, tests, drugs, statistics. (36) Tuberculose : vers un nouveau vaccin ?, 2019.

, OMS | Listes modèles OMS des médicaments essentiels, 2018.

S. R. Walker, Trends and Changes in Drug Research and Development, 2012.

J. Laborde, C. Deraeve, and V. Bernardes-génisson, Update of Antitubercular Prodrugs from a Molecular Perspective: Mechanisms of Action, Bioactivation Pathways, and Associated Resistance, ChemMedChem, vol.2017, issue.20, pp.1657-1676
URL : https://hal.archives-ouvertes.fr/hal-01939397

F. Bardou, C. Raynaud, C. Ramos, M. A. Lanéelle, and G. Lanéelle, Mechanism of Isoniazid Uptake in Mycobacterium Tuberculosis, Microbiology, pp.2539-2544, 1998.

Y. Zhang, B. Heym, B. Allen, D. Young, and S. Cole, The Catalase-Peroxidase Gene and Isoniazid Resistance of Mycobacterium Tuberculosis, Nature, vol.358, issue.6387, pp.591-593, 1992.

C. E. Cade, A. C. Dlouhy, K. F. Medzihradszky, S. P. Salas-castillo, and R. A. Ghiladi, Isoniazid-Resistance Conferring Mutations in Mycobacterium Tuberculosis KatG: Catalase, Peroxidase, and INH-NADH Adduct Formation Activities. Protein science : a publication of the, vol.19, pp.458-474, 2010.

S. Bala, Reclassification of Amycolatopsis Mediterranei DSM 46095 as Amycolatopsis Rifamycinica Sp, Nov. INTERNATIONAL JOURNAL OF SYSTEMATIC AND EVOLUTIONARY MICROBIOLOGY, vol.54, issue.4, pp.1145-1149, 2004.

M. Vilarem and . Un, ligand et activateur du récepteur des glucocorticoïdes. médecine/sciences, vol.14, p.451, 1998.

D. A. Mitchison, Understanding the Chemotherapy of Tuberculosis--Current Problems, J. Antimicrob. Chemother, vol.29, issue.5, pp.477-493, 1992.

J. Grosset, The Sterilizing Value of Rifampicin and Pyrazinamide in Experimental Short-Course Chemotherapy, Bull Int Union Tuberc, vol.53, issue.1, pp.5-12, 1978.

D. A. Mitchison and A. J. Nunn, Influence of Initial Drug Resistance on the Response to Short-Course Chemotherapy of Pulmonary Tuberculosis, Am. Rev. Respir. Dis, vol.133, issue.3, pp.423-430, 1986.

J. Perriot, É. Chambonnet, and A. Eschalier, Les Effets Indésirables Des Antituberculeux ; Prise En Charge, Revue des Maladies Respiratoires, vol.28, issue.4, pp.542-555, 2011.

H. M. Blumberg, W. J. Burman, R. E. Chaisson, C. L. Daley, S. C. Etkind et al., Infectious Diseases Society of America: Treatment of Tuberculosis. Am. J. Respir. Crit. Care Med, vol.167, issue.4, pp.603-662, 2003.

Y. Zhang, W. Shi, W. Zhang, and D. Mitchison, Mechanisms of Pyrazinamide Action and Resistance, Microbiol Spectr, vol.2013, issue.4, pp.1-12

R. L. Yeager, W. G. Munroe, F. I. Dessau, and . Pyrazinamide, Aldinamide) in the Treatment of Pulmonary Tuberculosis, Am Rev Tuberc, vol.1952, issue.5, pp.523-546

R. M. Mccune, R. Tompsett, . Fate, . Mycobacterium, . In et al., J Exp Med, vol.104, issue.5, pp.737-762, 1956.

Y. Zhang and D. Mitchison, The Curious Characteristics of Pyrazinamide: A Review, Int. J. Tuberc. Lung Dis, vol.7, issue.1, pp.6-21, 2003.

S. Ramón-garcía, V. Mick, E. Dainese, C. Martín, C. J. Thompson et al., Functional and Genetic Characterization of the Tap Efflux Pump in Mycobacterium Bovis BCG, Antimicrob Agents Chemother, vol.2012, issue.4, pp.2074-2083

A. Scorpio and Y. Zhang, Mutations in PncA, a Gene Encoding Pyrazinamidase/Nicotinamidase, Cause Resistance to the Antituberculous Drug Pyrazinamide in Tubercle Bacillus, Nature Medicine, vol.2, issue.6, pp.662-667, 1996.

Y. Zhang, A. Scorpio, H. Nikaido, and Z. Sun, Role of Acid PH and Deficient Efflux of Pyrazinoic Acid in Unique Susceptibility of Mycobacterium Tuberculosis to Pyrazinamide, J. Bacteriol, vol.181, issue.7, pp.2044-2049, 1999.

O. Zimhony, J. S. Cox, J. T. Welch, C. Vilchèze, and W. R. Jacobs, Pyrazinamide Inhibits the Eukaryotic-like Fatty Acid Synthetase I (FASI) of Mycobacterium Tuberculosis, Nat. Med, vol.6, issue.9, pp.1043-1047, 2000.

J. P. Thomas, C. O. Baughn, R. G. Wilkinson, and R. G. Shepherd, A New Synthetic Compound with Antituberculous Activity in Mice: Ethambutol (Dextro-2,2'-(Ethylenediimino)-Di-l-Butanol), Am. Rev. Respir. Dis, vol.83, pp.891-893, 1961.

K. Mikusová, R. A. Slayden, G. S. Besra, and P. J. Brennan, Biogenesis of the Mycobacterial Cell Wall and the Site of Action of Ethambutol, Antimicrob Agents Chemother, vol.39, issue.11, pp.2484-2489, 1995.

A. E. Belanger, G. S. Besra, M. E. Ford, K. Mikusová, J. T. Belisle et al., The EmbAB Genes of Mycobacterium Avium Encode an Arabinosyl Transferase Involved in Cell Wall Arabinan Biosynthesis That Is the Target for the Antimycobacterial Drug Ethambutol, Proc Natl Acad Sci, vol.93, issue.21, pp.11919-11924, 1996.

S. Chakraborty and K. Y. Rhee, Tuberculosis Drug Development: History and Evolution of the Mechanism-Based Paradigm. Cold Spring Harb Perspect Med, vol.5, 2015.

Y. Minato, J. M. Thiede, S. L. Kordus, E. J. Mcklveen, B. J. Turman et al., Mycobacterium Tuberculosis Folate Metabolism and the Mechanistic Basis for Para-Aminosalicylic Acid Susceptibility and Resistance, Antimicrob. Agents Chemother, vol.59, issue.9, pp.5097-5106, 2015.

J. Zheng, E. J. Rubin, P. Bifani, V. Mathys, V. Lim et al., Aminosalicylic Acid Is a Prodrug Targeting Dihydrofolate Reductase in Mycobacterium Tuberculosis?, J Biol Chem, vol.288, issue.32, pp.23447-23456, 2013.

S. S. Grant, S. Wellington, T. Kawate, C. A. Desjardins, M. R. Silvis et al., Baeyer-Villager Monooxygenases EthA and MymA Are Required for Activation of Inhibitors against Replicating and Non-Replicating Mycobacterium Tuberculosis, Cell Chem Biol, vol.23, issue.6, pp.666-677, 2016.

M. C. Henderson, L. K. Siddens, J. T. Morré, S. K. Krueger, and D. E. Williams, Metabolism of the Anti-Tuberculosis Drug Ethionamide by Mouse and Human FMO1, FMO2 and FMO3 and Mouse and Human Lung Microsomes, Toxicol. Appl. Pharmacol, vol.233, issue.3, pp.420-427, 2008.

. Vidal--cyclosérine, , 2018.

B. Phetsuksiri, A. R. Baulard, A. M. Cooper, D. E. Minnikin, J. D. Douglas et al., Antimycobacterial Activities of Isoxyl and New Derivatives through the Inhibition of Mycolic Acid Synthesis, Antimicrob. Agents Chemother, vol.43, issue.5, pp.1042-1051, 1999.

J. M. Belardinelli and H. R. Morbidoni, Mutations in the Essential FAS II ?-Hydroxyacyl ACP Dehydratase Complex Confer Resistance to Thiacetazone in Mycobacterium Tuberculosis and Mycobacterium Kansasii, Mol. Microbiol, vol.2012, issue.3, pp.568-579

. Vidal--linézolide, , 2018.

, A New Series of Phenazines (Rimino-Compounds) With High Antituberculosis Activity | Nature, vol.23, 2018.

. Vidal--clofazimine, , 2018.

. Vidal--imipénem, , 2018.

J. Hugonnet, L. W. Tremblay, H. I. Boshoff, C. E. Barry, and J. S. Blanchard, Meropenem-Clavulanate Is Effective Against Extensively Drug-Resistant Mycobacterium Tuberculosis, Science, vol.323, issue.5918, pp.1215-1218, 2009.

. Vidal--clarithromycine, , 2018.

R. Mahajan, Bedaquiline: First FDA-Approved Tuberculosis Drug in 40 Years, Int J Appl Basic Med Res, vol.2013, issue.1, pp.1-2

A. C. Haagsma, I. Podasca, A. Koul, K. Andries, J. Guillemont et al., Probing the Interaction of the Diarylquinoline TMC207 with Its Target Mycobacterial ATP Synthase, PLOS ONE, vol.6, issue.8, 2011.

K. Andries, P. Verhasselt, J. Guillemont, H. W. Göhlmann, J. Neefs et al., A Diarylquinoline Drug Active on the ATP Synthase of Mycobacterium Tuberculosis, Science, vol.307, issue.5707, pp.223-227, 2005.

H. Nagabushan and H. S. Roopadevi, Bedaquiline: A Novel Antitubercular Drug for Multidrug-Resistant Tuberculosis, J Postgrad Med, vol.60, issue.3, pp.300-302, 2014.

|. Delamanid and . Working, Group for New TB Drugs, 2018.

M. M. Pyle, Relative Numbers of Resistant Tubercle Bacilli in Sputa of Patients before and during Treatment with Streptomycin, Proc Staff Meet Mayo Clin, vol.22, issue.21, pp.465-473, 1947.

Y. Zhang and W. W. Yew, Mechanisms of Drug Resistance in Mycobacterium Tuberculosis, Int. J. Tuberc. Lung Dis, vol.13, issue.11, pp.1320-1330, 2009.

W. A. Denny and B. D. Palmer, The Nitroimidazooxazines (PA-824 and Analogs): Structure-Activity Relationship and Mechanistic Studies, Future Medicinal Chemistry, vol.2010, issue.8, pp.1295-1304

C. K. Stover, P. Warrener, D. R. Vandevanter, D. R. Sherman, T. M. Arain et al., A Small-Molecule Nitroimidazopyran Drug Candidate for the Treatment of Tuberculosis, Nature, issue.6789, p.962, 2000.

E. Schena, L. Nedialkova, E. Borroni, S. Battaglia, A. M. Cabibbe et al., Delamanid Susceptibility Testing of Mycobacterium Tuberculosis Using the Resazurin Microtitre Assay and the BACTEC TM MGIT TM 960 System, J. Antimicrob. Chemother, vol.71, issue.6, pp.1532-1539, 2016.

A. Koul, N. Dendouga, K. Vergauwen, B. Molenberghs, L. Vranckx et al., Diarylquinolines Target Subunit c of Mycobacterial ATP Synthase, Nat. Chem. Biol, vol.3, issue.6, pp.323-324, 2007.

A. C. Haagsma, R. Abdillahi-ibrahim, M. J. Wagner, K. Krab, K. Vergauwen et al., Selectivity of TMC207 towards Mycobacterial ATP Synthase Compared with That towards the Eukaryotic Homologue, Antimicrob Agents Chemother, vol.53, issue.3, pp.1290-1292, 2009.

, Bedaquiline, vol.25, 2019.

, Rifapentine | tuberculosis -List Results -ClinicalTrials, vol.17, 2019.

N. N. Bock, T. R. Sterling, C. D. Hamilton, C. Pachucki, Y. Wang et al., A Prospective, Randomized, Double-Blind Study of the Tolerability of Rifapentine 600, 900, and 1,200 Mg Plus Isoniazid in the Continuation Phase of Tuberculosis Treatment, Am J Respir Crit Care Med, vol.165, issue.11, pp.1526-1530, 2002.

. Admin and . Sanofi, La FDA approuve Priftin® (rifapentine, en comprimés) de Sanofi dans le traitement de l'infection tuberculeuse latente, vol.17, 2019.

A. S. Borisov, S. Bamrah-morris, G. J. Njie, C. A. Winston, D. Burton et al., Update of Recommendations for Use of Once-Weekly Isoniazid-Rifapentine Regimen to Treat Latent Mycobacterium Tuberculosis Infection, MMWR Morb Mortal Wkly Rep, vol.67, issue.25, pp.723-726, 2018.

, Figure 2. Process to identify new drug candidate, SQ109, from a focused

. Tb-r&d-update, Tuberculosis drug, SQ109, targets multiple enzymes and could treat other diseases while preventing resistance | Working Group for New TB Drugs, vol.29, 2019.

K. Tahlan, R. Wilson, D. B. Kastrinsky, K. Arora, V. Nair et al., SQ109 Targets MmpL3, a Membrane Transporter of Trehalose Monomycolate Involved in Mycolic Acid Donation to the Cell Wall Core of Mycobacterium Tuberculosis, Antimicrob. Agents Chemother, vol.56, issue.4, pp.1797-1809, 2012.

W. Li, A. Upadhyay, F. L. Fontes, E. J. North, Y. Wang et al., Novel Insights into the Mechanism of Inhibition of MmpL3, a Target of Multiple Pharmacophores in Mycobacterium Tuberculosis, Antimicrob. Agents Chemother, vol.58, issue.11, pp.6413-6423, 2014.

M. Brecik, I. Centárová, R. Mukherjee, G. S. Kolly, S. Huszár et al., DprE1 Is a Vulnerable Tuberculosis Drug Target Due to Its Cell Wall Localization, ACS Chemical Biology, vol.10, issue.7, pp.1631-1636, 2015.

V. Makarov, J. Neres, R. C. Hartkoorn, O. B. Ryabova, E. Kazakova et al., The 8-Pyrrole-Benzothiazinones Are Noncovalent Inhibitors of DprE1 from Mycobacterium Tuberculosis, Antimicrob Agents Chemother, vol.59, issue.8, pp.4446-4452, 2015.

, PanACEA Sutezolid Dose-finding and Combination Evaluation -Full Text View -ClinicalTrials, p.23, 2019.

M. R. Barbachyn, D. K. Hutchinson, S. J. Brickner, M. H. Cynamon, J. O. Kilburn et al., Identification of a Novel Oxazolidinone (U-100480) with Potent Antimycobacterial Activity, J. Med. Chem, vol.39, issue.3, pp.680-685, 1996.

R. S. Wallis, W. Jakubiec, V. Kumar, G. Bedarida, A. Silvia et al., Biomarker-Assisted Dose Selection for Safety and Efficacy in Early Development of PNU-100480 for Tuberculosis, Antimicrob. Agents Chemother, vol.55, issue.2, pp.567-574, 2011.

J. W. Alffenaar, T. Van-der-laan, S. Simons, T. S. Van-der-werf, P. J. Van-de-kasteele et al., Susceptibility of Clinical Mycobacterium Tuberculosis Isolates to a Potentially Less Toxic Derivate of Linezolid, PNU-100480, Antimicrob. Agents Chemother, vol.55, issue.3, pp.1287-1289, 2011.

K. N. Williams, S. J. Brickner, C. K. Stover, T. Zhu, A. Ogden et al., Addition of PNU-100480 to First-Line Drugs Shortens the Time Needed to Cure Murine Tuberculosis, Am. J. Respir. Crit. Care Med, vol.180, issue.4, pp.371-376, 2009.

Y. Choi, S. W. Lee, A. Kim, K. Jang, H. Nam et al., Tolerability and Pharmacokinetics of 21 Day Multiple Oral Administration of a New Oxazolidinone Antibiotic, LCB01-0371, in Healthy Male Subjects, J. Antimicrob. Chemother, vol.73, issue.1, pp.183-190, 2018.

T. S. Kim, J. H. Choe, Y. J. Kim, C. Yang, H. Kwon et al., Activity of LCB01-0371, a Novel Oxazolidinone, against Mycobacterium Abscessus, Antimicrob. Agents Chemother, issue.9, p.61, 2017.

Z. Zong, W. Jing, J. Shi, S. Wen, T. Zhang et al., Comparison of In Vitro Activity and MIC Distributions between the Novel Oxazolidinone Delpazolid and Linezolid against Multidrug-Resistant and Extensively Drug-Resistant Mycobacterium Tuberculosis in China, Antimicrob. Agents Chemother, issue.8, p.62, 2018.

, A Phase 2 Study to Evaluate Early Bactericidal Activity, Safety, Tolerability, and Pharmacokinetics of Multiple Oral Doses of Telacebec (Q203) -Full Text View -ClinicalTrials, 2019.

Q. C. Ltd and . Telacebec, , 2019.

N. P. Kalia, E. J. Hasenoehrl, N. B. Ab-rahman, V. H. Koh, M. L. Ang et al., Exploiting the Synthetic Lethality between Terminal Respiratory Oxidases to Kill Mycobacterium Tuberculosis and Clear Host Infection, Proc. Natl. Acad. Sci. U.S.A, vol.114, issue.28, pp.7426-7431, 2017.

. Vidal--auranofine, , 2019.

S. Thangamani, H. Mohammad, M. F. Abushahba, T. J. Sobreira, V. E. Hedrick et al., Antibacterial Activity and Mechanism of Action of Auranofin against Multi-Drug Resistant Bacterial Pathogens, Scientific Reports, vol.6, p.22571, 2016.

M. B. Harbut, C. Vilchèze, X. Luo, M. E. Hensler, H. Guo et al., Auranofin Exerts Broad-Spectrum Bactericidal Activities by Targeting Thiol-Redox Homeostasis, Proc Natl Acad Sci, vol.112, issue.14, pp.4453-4458, 2015.

C. P. Locher, S. M. Jones, B. L. Hanzelka, E. Perola, C. M. Shoen et al., A Novel Inhibitor of Gyrase B Is a Potent Drug Candidate for Treatment of Tuberculosis and Nontuberculosis Mycobacterial Infections, Antimicrob. Agents Chemother, vol.59, issue.3, pp.1455-1465, 2015.

Y. Lu, M. Zheng, B. Wang, L. Fu, W. Zhao et al., Clofazimine Analogs with Efficacy against Experimental Tuberculosis and Reduced Potential for Accumulation, Antimicrob. Agents Chemother, vol.55, issue.11, pp.5185-5193, 2011.

J. Xu, B. Wang, L. Fu, H. Zhu, S. Guo et al., Vitro and In Vivo Activities of the Riminophenazine TBI-166 against Mycobacterium Tuberculosis, vol.63, pp.2155-2173, 2019.

V. Makarov, O. B. Riabova, A. Yuschenko, N. Urlyapova, A. Daudova et al., Synthesis and Antileprosy Activity of Some Dialkyldithiocarbamates, J. Antimicrob. Chemother, vol.57, issue.6, pp.1134-1138, 2006.

V. Makarov, G. Manina, K. Mikusova, U. Möllmann, O. Ryabova et al., Benzothiazinones Kill Mycobacterium Tuberculosis by Blocking Arabinan Synthesis, Science, vol.324, issue.5928, pp.801-804, 2009.

K. Lv, X. You, B. Wang, Z. Wei, Y. Chai et al., Identification of Better Pharmacokinetic Benzothiazinone Derivatives as New Antitubercular Agents, ACS Med Chem Lett, vol.8, issue.6, pp.636-641, 2017.

T. Yuan and N. S. Sampson, Hit Generation in TB Drug Discovery: From Genome to Granuloma, Chem Rev, vol.118, issue.4, pp.1887-1916, 2018.

D. J. Hafkin, Updates in the Development of Delamanid, OPC-167832, and Otsuka's LAM Biomarker, vol.18

, A Phase 1 Study to Evaluate Safety, Tolerability, PK, and PK Interactions of TBA-7371 -Full Text View -ClinicalTrials, 2019.

C. Shoen, M. Destefano, B. Hafkin, and M. Cynamon, In Vitro and In Vivo Activities of Contezolid (MRX-I) against Mycobacterium Tuberculosis, Antimicrob. Agents Chemother, issue.8, p.62, 2018.

M. F. Gordeev and Z. Y. Yuan, New Potent Antibacterial Oxazolidinone (MRX-I) with an Improved Class Safety Profile, J. Med. Chem, vol.57, issue.11, pp.4487-4497, 2014.

, Contezolid Acefosamil Versus Linezolid for the Treatment of Acute Bacterial Skin and Skin Structure Infection -Full Text View -ClinicalTrials, Jul, vol.25, 2019.

H. S. Sutherland, A. S. Tong, P. J. Choi, A. Blaser, D. Conole et al., 5-Dialkoxypyridine Analogues of Bedaquiline Are Potent Antituberculosis Agents with Minimal Inhibition of the HERG Channel, Bioorg Med Chem, vol.3, issue.7, pp.1292-1307, 2019.

D. B. Aguirre, R. H. Bates, R. G. Rio, A. M. Losana, and S. R. García, Sanfetrinem or a Salt or Ester Thereof for Use in Treating Mycobacterial Infection. WO2018206466A1, 2018.

, GSK-286 | Groupe de travail sur les nouveaux médicaments antituberculeux, 2019.

, Analogues de spectinomycine avec des sucres à chaîne ramifiée en C-3, 2019.

R. Maier, E. Woitun, A. Reuter, W. Reuter, and B. Wetzel, Modification of Spectinomycin. 1. Synthesis of 4-Aminospectinomycins, J. Antibiot, vol.34, issue.1, pp.16-21, 1981.

E. Woitun, R. Maier, B. Wetzel, W. Reuter, and U. Lechner, Modification of Spectinomycin. 2. Derivatives of 4-Dihydro-4-Deoxy-4(R)-Aminospectinomycin, J. Antibiot, vol.34, issue.1, pp.22-27, 1981.

G. E. Zurenko, B. H. Yagi, J. J. Vavra, and B. B. Wentworth, In Vitro Antibacterial Activity of Trospectomycin (U-63366F, vol.32, issue.2, pp.216-223, 1988.

M. A. Borovinskaya, S. Shoji, J. M. Holton, K. Fredrick, and J. H. Cate, A Steric Block in Translation Caused by the Antibiotic Spectinomycin, ACS Chem. Biol, vol.2, issue.8, pp.545-552, 2007.

R. E. Lee, J. G. Hurdle, J. Liu, D. F. Bruhn, T. Matt et al., A New Class of Semisynthetic Anti-Tuberculosis Agents That Overcome Native Drug Efflux, Nat Med, vol.20, issue.2, pp.152-158, 2014.

D. B. Madhura, J. Liu, B. Meibohm, and R. E. Lee, Phase II Metabolic Pathways of Spectinamide Antitubercular Agents: A Comparative Study of the Reactivity of 4-Substituted Pyridines to Glutathione Conjugation, Medchemcomm, vol.2016, issue.1, pp.114-117

, cytochrome C reductase QcrB -Mycobacterium tuberculosis (strain ATCC 25177 / H37Ra) -qcrB gene & protein, vol.25, 2019.

, Inhibitor of the Respiratory Cytochrome bcc Complex for the Treatment of Drug-Resistant Tuberculosis | ACS Infectious Diseases, vol.25, 2019.

Y. Liu, Y. Gao, J. Liu, Y. Tan, Z. Liu et al., The Compound TB47 Is Highly Bactericidal against Mycobacterium Ulcerans in a Buruli Ulcer Mouse Model, Nature Communications, vol.10, issue.1, 2019.

Y. Takahashi, M. Igarashi, M. Okada, -. Anti, and . Drug, Anti-MDR-TB Drug, and Combination Anti-Tuberculosis Drug. US9040502B2, 2015.

J. Engohang-ndong, D. Baillat, M. Aumercier, F. Bellefontaine, G. S. Besra et al., Repressor of the TetR/CamR Family Implicated in Ethionamide Resistance in Mycobacteria, Octamerizes Cooperatively on Its Operator. Mol. Microbiol, vol.51, issue.1, pp.175-188, 2004.

M. Flipo, M. Desroses, N. Lecat-guillet, B. Dirié, X. Carette et al., Ethionamide Boosters: Synthesis, Biological Activity, and Structure-Activity Relationships of a Series of 1,2,4-Oxadiazole EthR Inhibitors, J. Med. Chem, vol.54, issue.8, pp.2994-3010, 2011.

N. Willand, B. Dirié, X. Carette, P. Bifani, A. Singhal et al., Synthetic EthR Inhibitors Boost Antituberculous Activity of Ethionamide, Nat. Med, vol.15, issue.5, pp.537-544, 2009.

N. Blondiaux, M. Moune, M. Desroses, R. Frita, M. Flipo et al., Reversion of Antibiotic Resistance in Mycobacterium Tuberculosis by Spiroisoxazoline SMARt-420, Science, vol.2017, issue.6330, pp.1206-1211

, Development of a Novel Lead that Targets M. tuberculosis Polyketide Synthase, vol.13, 2019.

G. F. Dos-santos-fernandes, P. C. De-souza, E. Moreno-viguri, M. Santivañez-veliz, R. Paucar et al., Synthesis, and Characterization of N-Oxide-Containing Heterocycles with in Vivo Sterilizing Antitubercular Activity, J Med Chem, vol.60, issue.20, pp.8647-8660, 2017.

M. Martínez-hoyos, E. Perez-herran, G. Gulten, L. Encinas, D. Álvarez-gómez et al., Antitubercular Drugs for an Old Target: GSK693 as a Promising InhA Direct Inhibitor, vol.8, pp.291-301, 2016.

S. P. Rao, S. B. Lakshminarayana, R. R. Kondreddi, M. Herve, L. R. Camacho et al., Indolcarboxamide Is a Preclinical Candidate for Treating Multidrug-Resistant Tuberculosis, Sci Transl Med, vol.5, issue.214, 2013.

, Synergistic Interactions of MmpL3 Inhibitors with Antitubercular Compounds In Vitro, vol.26, 2019.

P. Plankton, |. Bacteria, and . Encyclopedia, , 2019.

F. Wang, D. Sambandan, R. Halder, J. Wang, S. M. Batt et al., Identification of a Small Molecule with Activity against Drug-Resistant and Persistent Tuberculosis, Proc. Natl. Acad. Sci. U.S.A, vol.110, issue.27, pp.2510-2517, 2013.

. Vidal--bortézomib, , 2019.

W. Moreira, S. Santhanakrishnan, G. J. Ngan, C. B. Low, K. Sangthongpitag et al., Towards Selective Mycobacterial ClpP1P2 Inhibitors with Reduced Activity against the Human Proteasome, Antimicrob. Agents Chemother, issue.5, p.61, 2017.

B. V. Nikonenko, V. M. Reddy, M. Protopopova, E. Bogatcheva, L. Einck et al., Activity of SQ641, a Capuramycin Analog, in a Murine Model of Tuberculosis, Antimicrobial Agents and Chemotherapy, vol.53, issue.7, pp.3138-3139, 2009.

K. A. Sacksteder, M. Protopopova, C. E. Barry, K. Andries, and C. A. Nacy, Discovery and Development of SQ109: A New Antitubercular Drug with a Novel Mechanism of Action, Future Microbiol, vol.7, issue.7, pp.823-837, 2012.

V. Reddy, L. Einck, K. Andries, and C. Nacy, Vitro Interactions between New Antitubercular Drug Candidates SQ109 and TMC207. Antimicrobial agents and chemotherapy, vol.54, pp.2840-2846, 2010.

R. Tasneen, S. Li, C. A. Peloquin, D. Taylor, K. N. Williams et al., Sterilizing Activity of Novel TMC207-and PA-824-Containing Regimens in a Murine Model of Tuberculosis?, Antimicrob Agents Chemother, vol.55, issue.12, pp.5485-5492, 2011.

B. Nikonenko, V. M. Reddy, E. Bogatcheva, M. Protopopova, L. Einck et al., Therapeutic Efficacy of SQ641-NE against Mycobacterium Tuberculosis, Antimicrob Agents Chemother, vol.58, issue.1, pp.587-589, 2014.

E. Ballinger, J. Mosior, T. Hartman, K. Burns-huang, B. Gold et al., Opposing Reactions in Coenzyme A Metabolism Sensitize Mycobacterium Tuberculosis to Enzyme Inhibition, Science, vol.2019, issue.6426, p.363

C. Leblanc, T. Prudhomme, G. Tabouret, A. Ray, S. Burbaud et al., 4?-Phosphopantetheinyl Transferase PptT, a New Drug Target Required for Mycobacterium Tuberculosis Growth and Persistence In Vivo, PLoS Pathog, vol.8, issue.12, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02391857

S. Wellington, P. P. Nag, K. Michalska, S. E. Johnston, R. P. Jedrzejczak et al., A Small-Molecule Allosteric Inhibitor of Mycobacterium Tuberculosis Tryptophan Synthase, Nat. Chem. Biol, vol.13, issue.9, pp.943-950, 2017.

K. A. Abrahams, J. A. Cox, K. Fütterer, J. Rullas, F. Ortega-muro et al., Inhibiting Mycobacterial Tryptophan Synthase by Targeting the Inter-Subunit Interface, Sci Rep, vol.2017, issue.1, pp.1-15

|. Compounds and . Tb-alliance, , 2019.

J. Xu, J. Wang, J. Zhou, C. Xu, B. Huang et al., A Novel Protein Kinase Inhibitor IMB-YH-8 with Anti-Tuberculosis Activity. Sci Rep, vol.7, p.5093, 2017.

S. P. Parihar, R. Guler, R. Khutlang, D. M. Lang, R. Hurdayal et al., Statin Therapy Reduces the Mycobacterium Tuberculosis Burden in Human Macrophages and in Mice by Enhancing Autophagy and Phagosome Maturation, J Infect Dis, vol.209, issue.5, pp.754-763, 2014.

R. L. Leistikow, R. A. Morton, I. L. Bartek, I. Frimpong, K. Wagner et al., The Mycobacterium Tuberculosis DosR Regulon Assists in Metabolic Homeostasis and Enables Rapid Recovery from Nonrespiring Dormancy, Journal of Bacteriology, vol.192, issue.6, pp.1662-1670, 2010.

H. Zheng, C. J. Colvin, B. K. Johnson, P. D. Kirchhoff, M. Wilson et al., Inhibitors of Mycobacterium Tuberculosis DosRST Signaling and Persistence, Nat. Chem. Biol, vol.2017, issue.2, pp.218-225

. Oms-|-l, utilisation de l'artémisinine et de ses dérivés dans le traitement antipaludique, 2019.

W. H. Choi, Novel Pharmacological Activity of Artesunate and Artemisinin: Their Potential as Anti-Tubercular Agents, J Clin Med, vol.2017, issue.3

M. Morake, D. Coertzen, A. Ngwane, J. F. Wentzel, H. N. Wong et al., Preliminary Evaluation of Artemisinin-Cholesterol Conjugates as Potential Drugs for the Treatment of Intractable Forms of Malaria and Tuberculosis, ChemMedChem, vol.13, issue.1, pp.67-77, 2018.

S. Puckett, C. Trujillo, Z. Wang, H. Eoh, T. R. Ioerger et al., Glyoxylate Detoxification Is an Essential Function of Malate Synthase Required for Carbon Assimilation in Mycobacterium Tuberculosis, PNAS, vol.2017, issue.11, pp.2225-2232

K. S. Singh, R. Sharma, D. Keshari, N. Singh, and S. K. Singh, Down-Regulation of Malate Synthase in Mycobacterium Tuberculosis H37Ra Leads to Reduced Stress Tolerance, Persistence and Survival in Macrophages, Tuberculosis, vol.106, pp.73-81, 2017.

H. Huang, I. V. Krieger, M. K. Parai, V. B. Gawandi, and J. C. Sacchettini, Mycobacterium Tuberculosis Malate Synthase Structures with Fragments Reveal a Portal for Substrate/Product Exchange, J Biol Chem, vol.291, issue.53, pp.27421-27432, 2016.

R. Shukla, H. Shukla, and T. Tripathi, Structural and Energetic Understanding of Novel Natural Inhibitors of Mycobacterium Tuberculosis Malate Synthase, J. Cell. Biochem, 2018.

A. Trapero, A. Pacitto, V. Singh, M. Sabbah, A. G. Coyne et al., Fragment-Based Approach to Targeting Inosine-5'-Monophosphate Dehydrogenase (IMPDH) from Mycobacterium Tuberculosis, J. Med. Chem, issue.7, pp.2806-2822, 2018.

M. Makowska-grzyska, Y. Kim, S. K. Gorla, Y. Wei, K. Mandapati et al., Mycobacterium Tuberculosis IMPDH in Complexes with Substrates, Products and Antitubercular Compounds, p.10, 2015.

,

V. Singh, S. Donini, A. Pacitto, C. Sala, R. C. Hartkoorn et al., The Inosine Monophosphate Dehydrogenase, GuaB2, Is a Vulnerable New Bactericidal Drug Target for Tuberculosis, ACS Infect Dis, vol.2017, issue.1, pp.5-17

V. Usha, J. V. Hobrath, S. S. Gurcha, R. C. Reynolds, and G. S. Besra, Identification of Novel Mt-Guab2 Inhibitor Series Active against M. Tuberculosis, vol.7, 2012.

,

, WHO | WHO Preferred Product Characteristics for New Tuberculosis Vaccine, 2019.

, Global Clinical Portfolio of TB Vaccine Candidates, 2019.

R. S. Wallis and J. L. Johnson, Chapter 70 -Immunotherapy of Tuberculosis

H. S. Schaaf, A. I. Zumla, J. M. Grange, M. C. Raviglione, W. W. Yew et al., , pp.718-726, 2009.

, Phase III Clinical Study of Efficacy and Safety of Vaccae TM to Prevent Tuberculosis -Full Text View -ClinicalTrials, 2019.

L. Grode, C. A. Ganoza, C. Brohm, J. Weiner, B. Eisele et al., Safety and Immunogenicity of the Recombinant BCG Vaccine VPM1002 in a Phase 1 Open-Label Randomized Clinical Trial, Vaccine, vol.2013, issue.9, pp.1340-1348

S. K. Sharma, K. Katoch, R. Sarin, R. Balambal, N. K. Jain et al., Efficacy and Safety of Mycobacterium Indicus Pranii as an Adjunct Therapy in Category II Pulmonary Tuberculosis in a Randomized Trial, Sci Rep, vol.2017, issue.1, pp.1-12

A. K. Luabeya, B. M. Kagina, M. D. Tameris, H. Geldenhuys, S. T. Hoff et al., First-in-Human Trial of the Post-Exposure Tuberculosis Vaccine H56:IC31 in Mycobacterium Tuberculosis Infected and Non-Infected Healthy Adults, Vaccine, vol.33, issue.33, pp.4130-4140, 2015.

T. A. Olafsdottir, K. Lingnau, and E. Nagy, Jonsdottir, I. IC31, a Two-Component Novel Adjuvant Mixed with a Conjugate Vaccine Enhances Protective Immunity against Pneumococcal Disease in Neonatal Mice, Scand. J. Immunol, vol.69, issue.3, pp.194-202, 2009.

, Study to Evaluate H56:IC31 in Preventing Rate of TB Recurrence -Full Text View -ClinicalTrials, vol.29, 2019.

P. J. Cardona and I. Amat, Origin and Development of RUTI, a New Therapeutic Vaccine Against Mycobacterium Tuberculosis Infection, Archivos de Bronconeumología, vol.42, issue.1, pp.60110-60119, 2006.

P. Cardona and . Ruti, A New Chance to Shorten the Treatment of Latent Tuberculosis Infection, Tuberculosis (Edinb), vol.86, issue.3-4, pp.273-289, 2006.

J. Quigley, V. K. Hughitt, C. A. Velikovsky, R. A. Mariuzza, N. M. El-sayed et al., The Cell Wall Lipid PDIM Contributes to Phagosomal Escape and Host Cell Exit of Mycobacterium Tuberculosis, vol.8, pp.148-165, 2017.

J. Gonzalo-asensio, D. Marinova, C. Martin, N. Aguilo, and . Mtbvac, Attenuating the Human Pathogen of Tuberculosis (TB) Toward a Promising Vaccine against the TB Epidemic. Front Immunol, 2017.

A. Penn-nicholson, M. Tameris, E. Smit, T. A. Day, M. Musvosvi et al., Safety and Immunogenicity of the Novel Tuberculosis Vaccine ID93 + GLA-SE in BCG-Vaccinated Healthy Adults in South Africa: A Randomised, Double-Blind, Placebo-Controlled Phase 1 Trial, Lancet Respir Med, vol.6, issue.4, pp.30077-30085, 2018.

H. A. Fletcher and L. Schrager, TB Vaccine Development and the End TB Strategy: Importance and Current Status, Trans R Soc Trop Med Hyg, vol.110, issue.4, pp.212-218, 2016.

E. Stylianou, K. L. Griffiths, H. C. Poyntz, R. Harrington-kandt, M. D. Dicks et al., Improvement of BCG Protective Efficacy with a Novel Chimpanzee Adenovirus and a Modified Vaccinia Ankara Virus Both Expressing Ag85A. Vaccine, vol.33, pp.6800-6808, 2015.