, Mycobacterium tuberculosis Beijing Genotype Emerging in Vietnam, Emerging Infectious, vol.593, issue.3, pp.302-307, 2000.

A. Bahrmand, L. P. Reza, A. Titov, S. Hadizadeh-tasbiti, and E. A. Yari,

. Graviss, High-Level Rifampin Resistance Correlates with Multiple Mutations in the rpoB 597, 2009.

, Gene of Pulmonary Tuberculosis Isolates from the Afghanistan Border of Iran, Journal of Clinical 598 Microbiology, vol.47, issue.9, pp.2744-50

Y. Balabanova, B. Radiulyte, E. Davidaviciene, R. Hooper, and O. Ignatyeva, , p.600

F. A. Nikolayevskyy and . Drobniewski, Risk Factors for Drug-Resistant Tuberculosis 601 Patients in Lithuania, European Respiratory Journal, vol.39, issue.5, pp.1266-69, 2002.

,

P. J. Bifani, Z. Barun-mathema, S. L. Liu, B. Moghazeh, and . Shopsin, , p.604

J. Driscoll, Identification of a W Variant Outbreak of Mycobacterium tuberculosis 605 via Population-Based Molecular Epidemiology, JAMA, vol.282, issue.24, pp.2321-2348, 1999.

,

S. Bostanabad, A. R. Zaker, S. Bahrmand, F. Poorazar, A. Abdolrahimi et al., , p.608

M. Massomi and L. P. Titov, Mutations in Codon 315 of the KatG Gene 609 Associated with High-Level Resistance to Isoniazid, 2007.

F. Brossier, M. Boudinet, and V. Jarlier, Stéphanie Petrella, and Wladimir Sougakoff, Comparative Study of Enzymatic Activities of New KatG Mutants from Low-and High-Level, vol.611, 2016.

. Isoniazid-resistant, Clinical Isolates of Mycobacterium tuberculosis, Tuberculosis, vol.100, pp.15-24

T. N. Buu, M. N. Huyen, N. T. Lan, H. T. Quy, N. V. Hen et al.,

D. Cobelens and . Van-soolingen, The Beijing Genotype Is Associated with Young Age, p.616, 2009.

, Multidrug-Resistant Tuberculosis in Rural Vietnam, 2009.

P. J. Campbell, P. Glenn, R. D. Morlock, T. L. Sikes, B. Dalton et al.,

D. P. Starks, L. S. Hooks, B. B. Cowan, J. E. Plikaytis, and . Posey, Molecular Detection of Mutations Associated with First-and Second-Line Drug Resistance 621, vol.620, 2011.

, Compared with Conventional Drug Susceptibility Testing of Mycobacterium tuberculosis, p.622

, Antimicrobial Agents and Chemotherapy, vol.55, issue.5, pp.2032-2073

W. Cheunoy, M. Haile, A. Chaiprasert, and T. Prammananan,

M. Fernström, E. Vondracek, S. Chryssanthou, B. Hoffner, and . Petrini, Drug 625 Resistance and Genotypic Analysis of Mycobacterium tuberculosis Strains from Thai 626, 2009.

, Tuberculosis Patients, Microbiologica, et Immunologica Scandinavica, vol.627, issue.4, pp.286-90

H. Cox, T. Suzanne, D. Kubica, Y. Doshetov, and . Kebede, Sabine Rüsch-Gerdess, p.629

S. Niemann, The Beijing Genotype and Drug Resistant Tuberculosis in the Aral Sea 630 Region of Central Asia, Respiratory Research, vol.6, p.134, 2005.

. Cuevas-córdoba, S. Betzaida, A. Oyuki-xochihua-gonzález, and . Cuellar,

R. Domínguez and . Zenteno-cuevas, Characterization of PncA Gene Mutations in 634, 2013.

. Pyrazinamide-resistant, Mycobacterium tuberculosis Isolates from Mexico, Infection, Genetics 635 and Evolution, vol.19, pp.330-364

C. Demay, B. Liens, and T. Burguière, , p.637

C. Mokrousov, T. Sola, N. Zozio, and . Rastogi, , 2012.

, Available International Multimarker Database for Studying Mycobacterium tuberculosis Genetic 639

M. Diversity and . Epidemiology, Infection, Genetics and Evolution, Special Issue on 640 Molecular evolution, epidemiology and pathogenesis of Mycobacterium tuberculosis and other 641 mycobacteria, vol.12, pp.755-66

J. Domínguez, E. C. Boettger, D. Cirillo, F. Cobelens, K. D. Eisenach et al., Clinical Implications of Molecular Drug Resistance Testing for Mycobacterium 644 tuberculosis: A TBNET/RESIST-TB Consensus Statement, Tuberculosis and Lung Disease, vol.20, issue.1, pp.24-42, 2016.

N. Dookie, S. Rambaran, N. Padayatchi, S. Mahomed, and K. Naidoo, Evolution of Drug Resistance in Mycobacterium tuberculosis: A Review on the Molecular 649 Determinants of Resistance and Implications for Personalized Care, Journal of Antimicrobial, vol.648, 2018.

, Chemotherapy, vol.73, issue.5, pp.1138-51

F. Drobniewski, Y. Balabanova, V. Nikolayevsky, M. Ruddy, and S. Kuznetzov, , p.652

S. Zakharova, A. Melentyev, and I. Fedorin, Drug-Resistant 653, 2005.

, Clinical Virulence, and the Dominance of the Beijing Strain Family in Russia, Tuberculosis, vol.654, issue.22, pp.2726-2757

J. D. Embden, T. Van, K. Van-gorkom, R. Kremer, B. A. Jansen et al., Genetic Variation and Evolutionary Origin of the Direct Repeat Locus of Mycobacterium 657 tuberculosis Complex Bacteria, Journal of Bacteriology, vol.656, issue.9, pp.2393-2401, 2000.

, 659 "FAQ_TB_policy_recommendations_guidelines.Pdf, 2000.

M. R. Farhat, R. Karen, M. F. Jacobson, D. Franke, A. Kaur et al., Gyrase Mutations Are Associated with Variable Levels of, p.663, 2016.

, Fluoroquinolone Resistance in Mycobacterium tuberculosis, Journal of Clinical Microbiology, vol.54, issue.3, pp.727-760

A. Faustini, C. Hall, and . Perucci, Risk Factors for Multidrug Resistant Tuberculosis in Europe: 666 A Systematic Review, Thorax, vol.61, issue.2, pp.158-63, 2006.

I. Filliol, J. R. Driscoll, D. Van-soolingen, B. N. Kreiswirth, and K. Kremer, Georges, vol.668

D. Valétudie and . Duc-anh, Snapshot of Moving and Expanding Clones of 669, 2003.

, Mycobacterium tuberculosis and Their Global Distribution Assessed by Spoligotyping in an 670 705

J. Kamerbeek, . Schouls, . Kolk, . Van-agterveld, S. Van-soolingen et al., Simultaneous Detection and Strain Differentiation of Mycobacterium tuberculosis for 708 Diagnosis and Epidemiology, Journal of Clinical Microbiology, vol.35, issue.4, pp.907-921, 1997.

J. L. Kandler, T. Dalton, M. N. Ezewudo, L. Cowan, S. P. Burns et al., , p.711

P. Cegielski and J. E. Posey, Validation of Novel Isoniazid 712 Resistance Mutations Not Detectable by Common Molecular Tests, 2018.

,

S. Khann, E. T. Mao, Y. Prasad-rajendra, and S. Satyanarayana, Sharath Burugina, p.715

A. M. Nagaraja and . Kumar, Linkage of Presumptive Multidrug Resistant 716, 2013.

, MDR-TB) Patients to Diagnostic and Treatment Services in Cambodia, Tuberculosis, vol.717, issue.4

C. U. Köser and J. M. Bryant, Iñaki Comas, Silke Feuerriegel, Stefan Niemann, p.719

J. Gagneux, S. J. Parkhill, and . Peacock, Comment on: Characterization of the 720, 2014.

, EmbB Gene in Mycobacterium tuberculosis Isolates from Barcelona and Rapid Detection of Main 721

, Mutations Related to Ethambutol Resistance Using a Low-Density DNA Array, Journal, vol.722

, Antimicrobial Chemotherapy, vol.69, issue.8, pp.2298-99

A. Krüüner, S. E. Hoffner, and H. Sillastu,

S. Svenson, T. Ghebremichael, G. Koivula, and . Källenius, , 2001.

, Resistant Pulmonary Tuberculosis in Estonia, Journal of Clinical Microbiology, vol.39, issue.9, pp.3339-3384

,

R. W. Lau, . Pak-leung, . Ho, Y. T. Richard, . Kao et al.,

K. Cheng, . Yuen, K. W. Stephen, X. Tsui, W. Chen et al., Molecular Characterization of Fluoroquinolone Resistance in Mycobacterium tuberculosis: 730 Functional Analysis of GyrA Mutation at Position 74, Antimicrobial Agents and Chemotherapy, vol.729, issue.2, pp.608-622, 2011.

A. S. Lee, H. K. Irene, . Lim, L. H. Lynn, S. Tang et al., High Frequency of 733 Mutations in the RpoB Gene in Rifampin-Resistant Clinical Isolates of Mycobacterium 734 tuberculosis from Singapore, Journal of Clinical Microbiology, vol.43, issue.4, pp.2026-2053, 2005.

, , 2005.

P. Lempens, C. J. Meehan, K. Vandelannoote, and K. Fissette, , p.737

L. Deun, B. C. Rigouts, and J. De, Isoniazid Resistance Levels of 738 Mycobacterium tuberculosis Can Largely Be Predicted by High-Confidence Resistance-739, 2018.

, Conferring Mutations, Scientific Reports, vol.8, issue.1, p.3246

V. Lisdawati, N. Puspandari, L. Rif'ati, T. Soekarno, M. Melatiwati et al., , vol.742

N. Ratnasari, I. Izzatun, and . Parwati, Molecular Epidemiology Study of Mycobacterium 743 114 tuberculosis and Its Susceptibility to Anti-Tuberculosis Drugs in Indonesia, BMC Infectious, vol.744, 2015.

D. Machado, J. Perdigão, and J. Ramos,

M. Boettger and . Viveiros, High-Level Resistance to Isoniazid and Ethionamide in 747, 2013.

, Multidrug-Resistant Mycobacterium tuberculosis of the Lisboa Family Is Associated with InhA, vol.748

N. E. Maningi, T. Luke, J. D. Daum, . Rodriguez, M. Halima et al., John Osei, p.759

G. W. Sekyere, J. P. Fischer, P. Chambers, and . Bernard-fourie, , 2018.

, Extensively-Drug Resistant Mycobacterium tuberculosis in South Africa: A Molecular Analysis of 761 Historical Isolates | Request PDF, ResearchGate, 2018.

S. B. Marahatta, J. Kaewkungwal, P. Ramasoota, and P. Singhasivanon, Risk Factors, p.766, 2012.

, Multidrug Resistant Tuberculosis in Central Nepal: A Pilot Study, Kathmandu University Medical 767 Journal, vol.8, issue.4, pp.392-97

F. Maruri, T. R. Sterling, A. W. Kaiga, A. Blackman, and Y. F. Van-der-heijden, , p.769

C. Mayer, E. Cambau, and A. Aubry, A Systematic Review of, 2012.

, Gyrase Mutations Associated with Fluoroquinolone-Resistant Mycobacterium tuberculosis and a 771

, Proposed Gyrase Numbering System, Journal of Antimicrobial Chemotherapy, vol.67, issue.4, pp.819-850

,

W. Mcdermott and R. Tompsett, Activation of Pyrazinamide and Nicotinamide in Acidic 774 Environments in Vitro, American Review of Tuberculosis, vol.70, issue.4, pp.748-54, 1954.

P. Miotto, D. M. Cirillo, and G. B. Migliori, Drug Resistance InMycobacterium 776 tuberculosis: Molecular Mechanisms Challenging Fluoroquinolones and Pyrazinamide Eff, p.777, 2015.

. Ectiveness, CHEST, vol.147, issue.4, pp.1135-1178

I. Mokrousov, T. Otten, T. Zozio, E. Turkin, V. Nazemtseva et al., , p.779

B. Vishnevsky, O. Narvskaya, and N. Rastogi, At Baltic Crossroads: A Molecular 780 Snapshot of Mycobacterium tuberculosis Population Diversity in Kaliningrad, Russia, p.781, 2018.

, Immunology & Medical Microbiology, vol.55, issue.1, pp.13-22

G. P. Morlock, T. Jack, W. R. Crawford, S. E. Butler, D. Brim et al., , p.784

C. L. Woodley and R. C. Cooksey, Phenotypic Characterization of PncA 785 Mutants of Mycobacterium tuberculosis, Antimicrobial Agents and Chemotherapy, vol.44, issue.9, pp.2291-786, 2000.

G. Mulisa, T. Workneh, N. Hordofa, M. Suaudi, G. Abebe et al., Multidrug-Resistant Mycobacterium tuberculosis and Associated Risk Factors in Oromia 789 Region of Ethiopia, International Journal of Infectious Diseases, vol.788, pp.57-61, 2015.

,

H. Nguyen, N. Quang, L. Viet-nguyen, and . Contamin, , p.792

N. Van-nguyen and . Nguyen, Quadruple-First Line Drug Resistance in 793, 2017.

, Mycobacterium tuberculosis in Vietnam: What Can We Learn from Genes?, Genetics 794 and Evolution, vol.50, pp.55-61

Q. Nguyen and . Huy, Genetic Determinants and Evolution of Drug Resistance in Mycobacterium 796 tuberculosis in Vietnam: Toward New Diagnostic Tools, p.797, 2016.

C. Vietnam, Myanmar showed lower proportion of EAI than that observed in Laos (0.03%, 38.5%, 60%, and 48.4% respectively) but higher proportion of Beijing (74.1% , 38.5 %, 30 % and 31.9% respectively), This family was associated with older population, lower rates of TB transmission and mildly virulent

, Beijing family was associated with young people, urban areas [11, 180], greater virulence, drug resistance and highly transmissible, pp.181-183

, Conversely to population based sampling (TBPS), the Beijing family was more preponderant in hospital based sampling (DRS 2016-2017), and presumptive MDR-TB cases, 2010.

, This result is confirmed by the origin of the DRS isolates and presumptive MDR-TB cases (data not shown). Furthermore, the EAI family was more detected in age group ³ 35 years old, whereas Beijing family was also strongly present in younger age group (lower than 35 years old, 34.5% of Beinjing isolates). Furthermore, the analysis carried References 1. World Health Organization, % and 41 % respectively. This finding highlights the association of Beijing family with drug resistant TB and presumptive MDR-TB cases and thus the risk of increase and spread of this family in the country, 2017.

I. Law, P. Sylavanh, S. Bounmala, F. Nzabintwali, P. Paboriboune et al., The first national tuberculosis prevalence survey of Lao PDR, Trop Med Int Health, vol.20, pp.1146-54, 2010.

, Global tuberculosis report, 2012.

V. Iem, S. Somphavong, Y. Buisson, N. Steenkeste, F. Breysse et al., Resistance of Mycobacterium tuberculosis to antibiotics in Lao PDR: first multicentric study conducted in 3 hospitals, BMC Infectious Diseases, vol.13, p.275, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00972350

M. Zignol, A. M. Cabibbe, A. S. Dean, P. Glaziou, N. Alikhanova et al., Genetic sequencing for surveillance of drug resistance in tuberculosis in highly endemic countries: a multi-country population-based surveillance study. The Lancet Infectious Diseases, vol.18, pp.30073-30075, 2018.

R. Ramazanzadeh and K. Sayhemiri, Prevalence of Beijing family in Mycobacterium tuberculosis in world population: Systematic Review and Meta-Analysis, International Journal of Mycobacteriology, vol.3, pp.41-46, 2014.

H. Dou, F. Tseng, C. Lin, C. Sun, J. Tsai et al., Molecular epidemiology and evolutionary genetics of Mycobacterium tuberculosisin Taipei, BMC Infectious Diseases, vol.8, p.170, 2008.

K. Faksri, F. Drobniewski, V. Nikolayevskyy, T. Brown, T. Prammananan et al., Epidemiological trends and clinical comparisons of Mycobacterium tuberculosis lineages in Thai TB meningitis, Tuberculosis, vol.91, pp.594-600, 2011.

D. D. Anh, M. W. Borgdorff, L. N. Van, N. T. Lan, T. Van-gorkom et al., Mycobacterium tuberculosis Beijing genotype emerging in Vietnam, Emerg Infect Dis, vol.6, pp.302-307, 2000.

S. Phyu, R. Stavrum, T. Lwin, Ø. S. Svendsen, T. Ti et al., Predominance of Mycobacterium tuberculosis EAI and Beijing Lineages in Yangon, Myanmar, J Clin Microbiol, vol.47, pp.335-379, 2009.

V. Nguyen, M. Choisy, D. H. Nguyen, T. Tran, K. Pham et al., High Prevalence of Beijing and EAI4-VNM Genotypes among M. tuberculosis Isolates in Northern Vietnam: Sampling Effect, Rural and Urban Disparities, PLOS ONE, vol.7, p.45553, 2012.

H. Dong, Z. Liu, B. Lv, Y. Zhang, J. Liu et al., Spoligotypes of Mycobacterium tuberculosis from Different Provinces of China, J Clin Microbiol, vol.48, pp.4102-4108, 2010.

T. M. Daniel, The history of tuberculosis, Respiratory Medicine, vol.100, 2006.

A. Wright and M. Zignol, Anti-tuberculosis drug resistance in the world: fourth global report: the World Health Organization/International Union Against Tuberculosis and Lung Disease (WHO/UNION) Global Project on Anti-Tuberculosis Drug Resistance Surveillance, 2002.

S. Geneva, World Health Organization, 2008.

, WHO | Guidelines for the programmatic management of DR-TB. Companion Handbook to the WHO Guidelines for the Programmatic Management of Drug-Resistant Tuberculosis. Geneva: World Health Organization, 2014.

, WHO-Guidelines for Treatment of DS-TB Patient Care. Guidelines for Treatment of Drugsusceptible Tuberculosis and Patient Care, 2017.

B. Malhotra and W. Health, Guidelines for intensified tuberculosis case-finding and isoniazid preventative therapy for people living with HIV in resource-constrained settings, Department of HIV/AIDS : Stop TB Department, 2011.

O. Oxlade, D. Falzon, and D. Menzies, The impact and cost-effectiveness of strategies to detect drug-resistant tuberculosis, European Respiratory Journal, vol.39, pp.626-660, 2012.

L. M. Parsons, Á. Somoskövi, C. Gutierrez, E. Lee, C. N. Paramasivan et al., Laboratory Diagnosis of Tuberculosis in Resource-Poor Countries: Challenges and Opportunities, Clinical Microbiology Reviews, vol.24, pp.314-50, 2011.

H. D. Isenberg, Clinical Microbiology Procedures Handbook. American Society of Microbiology, 1992.

, Implementing tuberculosis diagnostics: policy framework. World Health Organization, WHO-Implementing TB diagnostics: policy framework

, Genève (Suisse): Word Health Organization, 2015.

, Stop TB Department, World Health Organization. Tuberculosis laboratory biosafety manual, Communicable Diseases Cluster, 2012.

, WHO | Guidelines for surveillance of DR in TB -5th edit. WHO | Guidelines for surveillance of drug resistance in tuberculosis, 2018.

K. Ängeby, P. Juréen, G. Kahlmeter, S. E. Hoffner, and T. Schön, WHO policy statement: molecular line probe assays. WHO | WHO policy statement: molecular line probe assays for rapid screening of patients at risk of multidrug-resistant tuberculosis, Bull World Health Organ, vol.90, pp.693-701, 2008.

, WHO-Xpert mtb/rif implementation manual. Xpert mtb/rif implementation manual: technical and operational "how-to, Geneva: World Health Organization, 2014.

M. Tomasicchio, G. Theron, E. Pietersen, E. Streicher, D. Stanley-josephs et al., The diagnostic accuracy of the MTBDRplus and MTBDRsl assays for drug-resistant TB detection when performed on sputum and culture isolates, Scientific Reports, vol.6, p.17850, 2016.

E. Sanchez-padilla, M. Merker, P. Beckert, F. Jochims, T. Dlamini et al., Detection of Drug-Resistant Tuberculosis by Xpert MTB/RIF in Swaziland, New England Journal of Medicine, vol.372, pp.1181-1183, 2015.

, Report of a Technical Expert Consultation: Non-inferiority analysis of Xpert MTB/RIF Ultra compared to Xpert MTB/RIF, WHO | WHO Meeting Report of a Technical Expert Consultation. WHO | WHO Meeting, 2018.

, WHO | The use of molecular LPA for the detection of resistance to SLDs. WHO | The use of molecular line probe assay for the detection of resistance to second-line anti-tuberculosis drugs, 2018.

, Global tuberculosis report, 2016.

L. Chaidir, C. Ruesen, B. E. Dutilh, A. R. Ganiem, A. Andryani et al., Use of whole genome sequencing to predict Mycobacterium tuberculosis drug resistance in Indonesia, Journal of Global Antimicrobial Resistance, 2018.

H. L. Aung, T. Tun, D. Moradigaravand, C. U. Köser, W. W. Nyunt et al., Whole-genome sequencing of multidrug-resistant Mycobacterium tuberculosis isolates from Myanmar, J Glob Antimicrob Resist, vol.6, pp.113-120, 2016.
DOI : 10.1016/j.jgar.2016.04.008

URL : https://doi.org/10.1016/j.jgar.2016.04.008

W. Ssengooba, C. J. Meehan, D. Lukoye, G. W. Kasule, K. Musisi et al., Whole genome sequencing to complement tuberculosis drug resistance surveys in Uganda. Infection, Genetics and Evolution, vol.40, pp.8-16, 2016.

C. Ruesen, A. L. Riza, A. Florescu, L. Chaidir, C. Editoiu et al., Linking minimum inhibitory concentrations to whole genome sequence-predicted drug resistance in Mycobacterium tuberculosis strains from Romania, Scientific Reports, vol.8, p.9676, 2018.

A. A. Witney, K. A. Gould, A. Arnold, D. Coleman, R. Delgado et al., Clinical Application of Whole-Genome Sequencing To Inform Treatment for Multidrug-Resistant Tuberculosis Cases, Journal of Clinical Microbiology, vol.53, pp.1473-83, 2015.

E. Cambau and M. Drancourt, Steps towards the discovery of Mycobacterium tuberculosis by Robert Koch, 1882, Clinical Microbiology and Infection, vol.20, pp.196-201, 2014.

M. Gengenbacher and S. Kaufmann, Mycobacterium tuberculosis: Success through dormancy, FEMS Microbiol Rev, vol.36, pp.514-546, 2012.
DOI : 10.1111/j.1574-6976.2012.00331.x

URL : https://academic.oup.com/femsre/article-pdf/36/3/514/18128985/36-3-514.pdf

E. Tortoli, P. Cichero, C. Piersimoni, M. T. Simonetti, G. Gesu et al., Use of BACTEC MGIT 960 for Recovery of Mycobacteria from Clinical Specimens: Multicenter Study, J Clin Microbiol, vol.37, pp.3578-82, 1999.

Á. Somoskövi and P. Magyar, Comparison of the Mycobacteria Growth Indicator Tube with MB Redox, Löwenstein-Jensen, and Middlebrook 7H11 Media for Recovery of Mycobacteria in Clinical Specimens, J Clin Microbiol, vol.37, pp.1366-1375, 1999.

G. Naveen and B. V. Peerapur, Comparison of the Lowenstein-Jensen Medium, the Middlebrook 7H10 Medium and MB/BacT for the Isolation of Mycobacterium Tuberculosis (MTB) from Clinical Specimens, J Clin Diagn Res, vol.6, pp.1704-1713, 2012.

H. Adler, C. Straub, and R. Frei, Comparison of BacT/ALERT 3D, Lowenstein-Jensen medium and Middlebrook 7H10/7H11 biplate for recovering mycobacteria from clinical specimens, Eur J Clin Microbiol Infect Dis, vol.24, pp.499-500, 2005.

D. Kaur, M. E. Guerin, H. ?kovierová, P. J. Brennan, and M. Jackson, Biogenesis of the cell wall and other glycoconjugates of Mycobacterium tuberculosis, Adv Appl Microbiol, vol.69, pp.23-78, 2009.

S. T. Cole, R. Brosch, J. Parkhill, T. Garnier, C. Churcher et al., Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequence, Nature, vol.393, pp.537-581, 1998.

T. M. Walker, C. L. Ip, R. H. Harrell, J. T. Evans, G. Kapatai et al., Whole-genome sequencing to delineate Mycobacterium tuberculosis outbreaks: a retrospective observational study, Lancet Infect Dis, vol.13, pp.137-183, 2013.

R. D. Fleischmann, D. Alland, J. A. Eisen, L. Carpenter, O. White et al., WholeGenome Comparison of Mycobacterium tuberculosis Clinical and Laboratory Strains, J Bacteriol, vol.184, pp.5479-90, 2002.

M. Merker, T. A. Kohl, A. Roetzer, L. Truebe, E. Richter et al., Whole Genome Sequencing Reveals Complex Evolution Patterns of Multidrug-Resistant Mycobacterium tuberculosis Beijing Strains in Patients, PLOS ONE, vol.8, p.82551, 2013.

E. N. Ilina, E. A. Shitikov, L. N. Ikryannikova, D. G. Alekseev, D. E. Kamashev et al.,

, Comparative Genomic Analysis of Mycobacterium tuberculosis Drug Resistant Strains from Russia, PLOS ONE, vol.8, p.56577, 2013.

N. Casali, V. Nikolayevskyy, Y. Balabanova, O. Ignatyeva, I. Kontsevaya et al., Microevolution of extensively drug-resistant tuberculosis in Russia, Genome Res, vol.22, pp.735-780, 2012.

M. Coscolla and S. Gagneux, Consequences of genomic diversity in Mycobacterium tuberculosis, Semin Immunol, vol.26, pp.431-475, 2014.

T. D. Vultos, O. Mestre, J. Rauzier, M. Golec, N. Rastogi et al., Evolution and Diversity of Clonal Bacteria: The Paradigm of Mycobacterium tuberculosis, PLOS ONE, vol.3, 2008.
URL : https://hal.archives-ouvertes.fr/pasteur-00364652

A. Roetzer, R. Diel, T. A. Kohl, C. Rückert, U. Nübel et al., Whole Genome Sequencing versus Traditional Genotyping for Investigation of a Mycobacterium tuberculosis Outbreak: A Longitudinal Molecular Epidemiological Study, PLOS Medicine, vol.10, 2013.

S. Gagneux and P. M. Small, Global phylogeography of Mycobacterium tuberculosis and implications for tuberculosis product development, Lancet Infect Dis, vol.7, pp.328-365, 2007.

S. Gagneux, K. Deriemer, T. Van, M. Kato-maeda, B. C. Jong et al., Variable host-pathogen compatibility in Mycobacterium tuberculosis, PNAS, vol.103, pp.2869-73, 2006.

M. C. Gutierrez, S. Brisse, R. Brosch, M. Fabre, B. Omaïs et al., Ancient Origin and Gene Mosaicism of the Progenitor of Mycobacterium tuberculosis, PLOS Pathogens, vol.1, p.5, 2005.
URL : https://hal.archives-ouvertes.fr/inserm-00080315

R. Firdessa, S. Berg, E. Hailu, E. Schelling, B. Gumi et al., Mycobacterial Lineages Causing Pulmonary and Extrapulmonary Tuberculosis, Ethiopia. Emerg Infect Dis, vol.19, pp.460-463, 2013.

B. Tessema, J. Beer, M. Merker, F. Emmrich, U. Sack et al., Molecular epidemiology and transmission dynamics of Mycobacterium tuberculosis in Northwest Ethiopia: new phylogenetic lineages found in Northwest Ethiopia, BMC Infectious Diseases, vol.13, p.131, 2013.

R. Brosch, S. V. Gordon, M. Marmiesse, P. Brodin, C. Buchrieser et al., A new evolutionary scenario for the Mycobacterium tuberculosis complex, Proc Natl Acad Sci USA, vol.99, pp.3684-3693, 2002.

I. Comas, M. Coscolla, T. Luo, S. Borrell, K. E. Holt et al., Out-of-Africa migration and Neolithic co-expansion of Mycobacterium tuberculosis with modern humans, Nat Genet, vol.45, pp.1176-82, 2013.

J. E. Galagan, Genomic insights into tuberculosis, Nature Reviews Genetics, vol.15, pp.307-327, 2014.
DOI : 10.1038/nrg3664

C. Demay, B. Liens, T. Burguière, V. Hill, D. Couvin et al., SITVITWEB -A publicly available international multimarker database for studying Mycobacterium tuberculosis genetic diversity and molecular epidemiology. Infection, Genetics and Evolution, vol.12, pp.755-66, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-00691750

K. Brudey, J. R. Driscoll, L. Rigouts, W. M. Prodinger, A. Gori et al., Mycobacterium tuberculosis complex genetic diversity: mining the fourth international spoligotyping database (SpolDB4) for classification, population genetics and epidemiology, BMC Microbiol, vol.6, p.23, 2006.

L. Qian, J. Van-embden, A. Van-der-zanden, E. F. Weltevreden, H. Duanmu et al., Retrospective Analysis of the Beijing Family of Mycobacterium tuberculosis in Preserved Lung Tissues, J Clin Microbiol, vol.37, pp.471-475, 1999.

D. Van-soolingen, L. Qian, P. E. De-haas, J. T. Douglas, H. Traore et al., Predominance of a single genotype of Mycobacterium tuberculosis in countries of east Asia, J Clin Microbiol, vol.33, pp.3234-3242, 1995.

N. Reiling, S. Homolka, K. Walter, J. Brandenburg, L. Niwinski et al., Clade-specific virulence patterns of Mycobacterium tuberculosis complex strains in human primary macrophages and aerogenically infected mice, MBio, vol.4, 2013.

D. Brites and S. Gagneux, Co-evolution of Mycobacterium tuberculosis and Homo sapiens, Immunol Rev, vol.264, pp.6-24, 2015.

S. Gagneux, Host-pathogen coevolution in human tuberculosis, Philos Trans R Soc Lond B Biol Sci, vol.367, pp.850-859, 2012.

M. Huyen, T. N. Buu, E. Tiemersma, N. Lan, N. H. Dung et al., Tuberculosis Relapse in Vietnam Is Significantly Associated With Mycobacterium tuberculosis Beijing Genotype Infections, The Journal of Infectious Diseases, vol.207, pp.1516-1540, 2013.

N. Casali, V. Nikolayevskyy, Y. Balabanova, S. R. Harris, O. Ignatyeva et al., Evolution and transmission of drug resistant tuberculosis in a Russian population, Nat Genet, vol.46, pp.279-86, 2014.

S. Ribeiro, L. L. Gomes, E. P. Amaral, M. Andrade, F. M. Almeida et al., Mycobacterium tuberculosis Strains of the Modern Sublineage of the Beijing Family Are More Likely To Display Increased Virulence than Strains of the Ancient Sublineage, J Clin Microbiol, vol.52, pp.2615-2639, 2014.

M. Klopper, R. M. Warren, C. Hayes, G. Van-pittius, N. C. Streicher et al., Emergence and Spread of Extensively and Totally Drug-Resistant Tuberculosis, South Africa. Emerg Infect Dis, vol.19, pp.449-55, 2013.

, Global tuberculosis report, 2018.

, WHO treatment guidelines for DR-TB: update. WHO treatment guidelines for drug-resistant tuberculosis: 2016 update, 2016.

J. C. Palomino and A. Martin, Drug Resistance Mechanisms in Mycobacterium tuberculosis, Antibiotics (Basel), vol.3, pp.317-357, 2014.

A. Silva, P. E. Palomino, and J. C. , Molecular basis and mechanisms of drug resistance in Mycobacterium tuberculosis: classical and new drugs, J Antimicrob Chemother, vol.66, 2011.

S. H. Gillespie, Evolution of Drug Resistance in Mycobacterium tuberculosis: Clinical and Molecular Perspective, Antimicrob Agents Chemother, vol.46, pp.267-74, 2002.

J. D. Fonseca, G. M. Knight, and T. D. Mchugh, The complex evolution of antibiotic resistance in Mycobacterium tuberculosis, International Journal of Infectious Diseases, vol.32, pp.94-100, 2015.

. Silva-pea-da, A. V. Groll, A. Martin, and J. C. Palomino, Efflux as a mechanism for drug resistance in Mycobacterium tuberculosis, FEMS Immunology & Medical Microbiology, vol.63, pp.1-9, 2011.

G. E. Louw, R. M. Warren, G. Van-pittius, N. C. Mcevoy, C. Van-helden et al., A Balancing Act: Efflux/Influx in Mycobacterial Drug Resistance, Antimicrob Agents Chemother, vol.53, pp.3181-3190, 2009.

I. Chopra, A. J. O'neill, and K. Miller, The role of mutators in the emergence of antibiotic-resistant bacteria, Drug Resistance Updates, vol.6, pp.137-182, 2003.

J. L. Martinez and F. Baquero, Mutation Frequencies and Antibiotic Resistance, Antimicrobial Agents and Chemotherapy, vol.44, pp.1771-1778, 2000.

S. V. Ramaswamy, R. Reich, S. Dou, L. Jasperse, X. Pan et al., Single Nucleotide Polymorphisms in Genes Associated with Isoniazid Resistance in Mycobacterium tuberculosis, Antimicrob Agents Chemother, vol.47, pp.1241-50, 2003.

M. H. Hazbón, M. Brimacombe, B. Del-valle, M. Cavatore, M. Guerrero et al., Population Genetics Study of Isoniazid Resistance Mutations and Evolution of Multidrug-Resistant Mycobacterium tuberculosis, Antimicrob Agents Chemother, vol.50, pp.2640-2649, 2006.

C. Vilchèze and W. R. Jacobs, The Mechanism of Isoniazid Killing: Clarity Through the Scope of Genetics, Annual Review of Microbiology, vol.61, pp.35-50, 2007.

A. Banerjee, E. Dubnau, A. Quemard, V. Balasubramanian, K. S. Um et al., Mycobacterium tuberculosis, vol.263, pp.227-257, 1994.

M. H. Larsen, C. Vilchèze, L. Kremer, G. S. Besra, L. Parsons et al., Overexpression of inhA, but not kasA, confers resistance to isoniazid and ethionamide in Mycobacterium smegmatis, M. bovis BCG and M. tuberculosis, vol.46, pp.453-66, 2002.

D. A. Mitchison, Basic Mechanisms of Chemotherapy, CHEST, vol.76, pp.771-80, 1979.

K. Konno, F. M. Feldmann, and W. Mcdermott, Pyrazinamide Susceptibility and Amidase Activity of Tubercle Bacilli, Am Rev Respir Dis, vol.95, pp.461-470, 1967.

Y. Zhang and D. Mitchison, The curious characteristics of pyrazinamide: a review, Int J Tuberc Lung Dis, vol.7, pp.6-21, 2003.

A. Scorpio, P. Lindholm-levy, L. Heifets, R. Gilman, S. Siddiqi et al., Characterization of pncA mutations in pyrazinamide-resistant Mycobacterium tuberculosis

, Antimicrob Agents Chemother, vol.41, pp.540-543, 1997.

M. G. Whitfield, H. M. Soeters, R. M. Warren, T. York, S. L. Sampson et al., A Global Perspective on Pyrazinamide Resistance: Systematic Review and Meta-Analysis, PLOS ONE, vol.10, p.133869, 2015.

K. Takayama and J. O. Kilburn, Inhibition of synthesis of arabinogalactan by ethambutol in Mycobacterium smegmatis, Antimicrob Agents Chemother, vol.33, pp.1493-1502, 1989.

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, pp.2484-2493, 1995.

A. Telenti, W. J. Philipp, S. Sreevatsan, C. Bernasconi, K. E. Stockbauer et al., The emb operon, a gene cluster of Mycobacterium tuberculosis involved in resistance to ethambutol, Nature Medicine, vol.3, pp.567-70, 1997.

S. Sreevatsan, K. E. Stockbauer, X. Pan, B. N. Kreiswirth, S. L. Moghazeh et al., Ethambutol resistance in Mycobacterium tuberculosis: critical role of embB mutations, Antimicrob Agents Chemother, vol.41, pp.1677-81, 1997.

D. Moazed and H. F. Noller, Interaction of antibiotics with functional sites in 16S ribosomal RNA, Nature, vol.327, pp.389-94, 1987.

M. Finken, P. Kirschner, A. Meier, A. Wrede, and E. C. Böttger, Molecular basis of streptomycin resistance in Mycobacterium tuberculosis: alterations of the ribosomal protein S12 gene and point mutations within a functional 16S ribosomal RNA pseudoknot, Molecular Microbiology, vol.9, pp.1239-1285, 1993.

S. B. Georghiou, M. Magana, R. S. Garfein, D. G. Catanzaro, A. Catanzaro et al., Evaluation of Genetic Mutations Associated with Mycobacterium tuberculosis Resistance to Amikacin, Kanamycin and Capreomycin: A Systematic Review, PLOS ONE, vol.7, p.33275, 2012.

H. E. Takiff, L. Salazar, C. Guerrero, W. Philipp, W. M. Huang et al., Cloning and nucleotide sequence of Mycobacterium tuberculosis gyrA and gyrB genes and detection of quinolone resistance mutations, Antimicrob Agents Chemother, vol.38, pp.773-80, 1994.

A. Fàbrega, S. Madurga, E. Giralt, and J. Vila, Mechanism of action of and resistance to quinolones, Microb Biotechnol, vol.2, pp.40-61, 2009.

A. Aubry, X. Pan, L. M. Fisher, V. Jarlier, and E. Cambau, Mycobacterium tuberculosis DNA Gyrase: Interaction with Quinolones and Correlation with Antimycobacterial Drug Activity, Antimicrob Agents Chemother, vol.48, pp.1281-1289, 2004.
DOI : 10.1128/aac.48.4.1281-1288.2004

URL : https://aac.asm.org/content/48/4/1281.full.pdf

Y. Zhang and W. Yew, Mechanisms of drug resistance in Mycobacterium tuberculosis: update 2015, 2015.

N. Dookie, S. Rambaran, N. Padayatchi, S. Mahomed, and K. Naidoo, Evolution of drug resistance in Mycobacterium tuberculosis: a review on the molecular determinants of resistance and implications for personalized care, J Antimicrob Chemother, vol.73, pp.1138-51, 2018.

S. Muralidhar and L. Srivastava, Evaluation of three methods to determine the antimicrobial susceptibility of Mycobacterium tuberculosis, Indian J Med Res, vol.120, pp.463-470, 2004.

G. Canetti, W. Fox, A. Khomenko, H. T. Mahler, N. K. Menon et al., Advances in techniques of testing mycobacterial drug sensitivity, and the use of sensitivity tests in tuberculosis control programmes, Bull World Health Organ, vol.41, pp.21-43, 1969.

S. Tyagi and F. R. Kramer, Molecular Beacons: Probes that Fluoresce upon Hybridization, Nature Biotechnology, vol.14, pp.303-311, 1996.
DOI : 10.1038/nbt0396-303

S. Tyagi, D. P. Bratu, and F. R. Kramer, Multicolor molecular beacons for allele discrimination, Nature Biotechnology, vol.16, pp.49-53, 1998.
DOI : 10.1038/nbt0198-49

A. S. Piatek, S. Tyagi, A. C. Pol, A. Telenti, L. P. Miller et al., Molecular beacon sequence analysis for detecting drug resistance in Mycobacterium tuberculosis, Nature Biotechnology, vol.16, pp.359-63, 1998.
DOI : 10.1038/nbt0498-359

A. S. Piatek, A. Telenti, M. R. Murray, H. El-hajj, W. R. Jacobs et al., Genotypic Analysis of Mycobacterium tuberculosis in Two Distinct Populations Using Molecular Beacons: Implications for Rapid Susceptibility Testing, Antimicrobial Agents and Chemotherapy, vol.44, pp.103-113, 2000.

D. Helb, M. Jones, E. Story, C. Boehme, E. Wallace et al., Rapid Detection of Mycobacterium tuberculosis and Rifampin Resistance by Use of On-Demand, Near-Patient Technology, Journal of Clinical Microbiology, vol.48, pp.229-266, 2010.

R. Blakemore, E. Story, D. Helb, J. Kop, P. Banada et al., Evaluation of the Analytical Performance of the Xpert MTB/RIF Assay, Journal of Clinical Microbiology, vol.48, pp.2495-501, 2010.

S. D. Lawn and M. P. Nicol, Xpert® MTB/RIF assay: development, evaluation and implementation of a new rapid molecular diagnostic for tuberculosis and rifampicin resistance, Future Microbiol, vol.6, pp.1067-82, 2011.

J. Kamerbeek, L. Schouls, A. Kolk, M. Van-agterveld, D. Van-soolingen et al., Simultaneous detection and strain differentiation of Mycobacterium tuberculosis for diagnosis and epidemiology, J Clin Microbiol, vol.35, pp.907-921, 1997.

P. F. Barnes and M. D. Cave, Molecular Epidemiology of Tuberculosis, New England Journal of Medicine, vol.349, pp.1149-56, 2003.

I. Filliol, J. R. Driscoll, D. Van-soolingen, B. N. Kreiswirth, K. Kremer et al., Snapshot of Moving and Expanding Clones of Mycobacterium tuberculosis and Their Global Distribution Assessed by Spoligotyping in an International Study, J Clin Microbiol, vol.41, pp.1963-70, 2003.

I. Vitola, J. Driscoll, B. Kreiswirth, N. Kurepina, P. Bennett et al., Identifying Mycobacterium tuberculosis Complex Strain Families using Spoligotypes, Infect Genet Evol, vol.6, pp.491-504, 2006.

R. Cox and S. M. Mirkin, Characteristic enrichment of DNA repeats in different genomes, Proc Natl Acad Sci U S A, vol.94, pp.5237-5279, 1997.

Y. Nakamura, M. Leppert, P. O'connell, R. Wolff, T. Holm et al., Variable number of tandem repeat (VNTR) markers for human gene mapping, Science, vol.235, pp.1616-1638, 1987.

Y. Nakamura, M. Carlson, K. Krapcho, M. Kanamori, and R. White, New approach for isolation of VNTR markers, Am J Hum Genet, vol.43, pp.854-863, 1988.

S. A. Woods and C. St, A family of dispersed repeats in Mycobacterium leprae, Molecular Microbiology, vol.4, pp.1745-51, 1990.

P. W. Hermans, D. Van-soolingen, and J. D. Van-embden, Characterization of a major polymorphic tandem repeat in Mycobacterium tuberculosis and its potential use in the epidemiology of Mycobacterium kansasii and Mycobacterium gordonae, J Bacteriol, vol.174, pp.4157-65, 1992.

D. Van-soolingen, P. E. De-haas, P. W. Hermans, P. M. Groenen, and J. D. Van-embden, Comparison of various repetitive DNA elements as genetic markers for strain differentiation and epidemiology of Mycobacterium tuberculosis, J Clin Microbiol, vol.31, pp.1987-95, 1993.

F. Bigi, M. Romano, A. Alito, and A. Cataldi, Cloning of a novel polymorphic GC-rich repetitive DNA from Mycobacterium bovis, Research in Microbiology, vol.146, issue.96, pp.81057-81063, 1995.

W. J. Philipp, S. Poulet, K. Eiglmeier, L. Pascopella, V. Balasubramanian et al., An integrated map of the genome of the tubercle bacillus, Mycobacterium tuberculosis H37Rv, and comparison with Mycobacterium leprae, Proc Natl Acad Sci U S A, vol.93, pp.3132-3139, 1996.

P. Supply, J. Magdalena, S. Himpens, and C. Locht, Identification of novel intergenic repetitive units in a mycobacterial two-component system operon, Molecular Microbiology, vol.26, pp.991-1003, 1997.

P. Supply, E. Mazars, S. Lesjean, V. Vincent, B. Gicquel et al., Variable human minisatellite-like regions in the Mycobacterium tuberculosis genome, Molecular Microbiology, vol.36, pp.762-71, 2000.

E. Mazars, S. Lesjean, A. Banuls, M. Gilbert, V. Vincent et al., High-resolution minisatellite-based typing as a portable approach to global analysis of Mycobacterium tuberculosis molecular epidemiology, Proc Natl Acad Sci U S A, vol.98, pp.1901-1907, 2001.

P. Supply, S. Lesjean, E. Savine, K. Kremer, . Soolingen-d-van et al., Automated HighThroughput Genotyping for Study of Global Epidemiology of Mycobacterium tuberculosis Based on Mycobacterial Interspersed Repetitive Units, J Clin Microbiol, vol.39, pp.3563-71, 2001.

P. Supply, C. Allix, S. Lesjean, M. Cardoso-oelemann, S. Rüsch-gerdes et al., Proposal for Standardization of Optimized Mycobacterial Interspersed Repetitive Unit-VariableNumber Tandem Repeat Typing of Mycobacterium tuberculosis, J Clin Microbiol, vol.44, pp.4498-510, 2006.

M. Gauthier, F. Bidault, A. Mosnier, N. Bablishvili, N. Tukvadze et al., HighThroughput Mycobacterial Interspersed Repetitive-Unit-Variable-Number Tandem-Repeat Genotyping for Mycobacterium tuberculosis Epidemiological Studies, J Clin Microbiol, vol.53, pp.498-503, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01911349

C. Allix-béguec, D. Harmsen, T. Weniger, P. Supply, and S. Niemann, Evaluation and Strategy for Use of MIRU-VNTRplus, a Multifunctional Database for Online Analysis of Genotyping Data and Phylogenetic Identification of Mycobacterium tuberculosis Complex Isolates, J Clin Microbiol, vol.46, pp.2692-2701, 2008.

. Nguyen-v-a.-t, H. Q. Nguyen, T. T. Vu, N. Nguyen, .. T. Duong et al., Reduced turn-around time for Mycobacterium tuberculosis drug susceptibility testing with a proportional agar microplate assay, Clinical Microbiology and Infection, vol.21, pp.1084-92, 2015.

H. Q. Nguyen, N. V. Nguyen, L. Contamin, T. Tran, T. T. Vu et al., Quadruple-first line drug resistance in Mycobacterium tuberculosis in Vietnam: What can we learn from genes? Infection, Genetics and Evolution, vol.50, pp.55-61, 2017.

H. Van-deutekom, S. P. Hoijng, P. De-haas, M. W. Langendam, A. Horsman et al., Clustered tuberculosis cases: do they represent recent transmission and can they be detected earlier?, Am J Respir Crit Care Med, vol.169, pp.806-816, 2004.

S. Sreevatsan, X. Pan, K. E. Stockbauer, N. D. Connell, B. N. Kreiswirth et al., Restricted structural gene polymorphism in the Mycobacterium tuberculosis complex indicates evolutionarily recent global dissemination, Proc Natl Acad Sci USA, vol.94, pp.9869-74, 1997.

J. N. Torres, L. V. Paul, T. C. Rodwell, T. C. Victor, A. M. Amallraja et al., Novel katG mutations causing isoniazid resistance in clinical M. tuberculosis isolates, Emerg Microbes Infect, vol.4, p.42, 2015.

Q. H. Nguyen, Genetic determinants and evolution of drug resistance in Mycobacterium tuberculosis in Vietnam: toward new diagnostic tools, 2016.
URL : https://hal.archives-ouvertes.fr/tel-01560449

C. U. Köser, J. M. Bryant, I. Comas, S. Feuerriegel, S. Niemann et al., Comment on: Characterization of the embB gene in Mycobacterium tuberculosis isolates from Barcelona and rapid detection of main mutations related to ethambutol resistance using a low-density DNA array, J Antimicrob Chemother, vol.69, pp.2298-2307, 2014.

M. R. Farhat, K. R. Jacobson, M. F. Franke, D. Kaur, A. Sloutsky et al., Gyrase Mutations Are Associated with Variable Levels of Fluoroquinolone Resistance in Mycobacterium tuberculosis, J Clin Microbiol, vol.54, pp.727-760, 2016.

P. Miotto, D. M. Cirillo, and G. B. Migliori, Drug Resistance inMycobacterium tuberculosis: Molecular Mechanisms Challenging Fluoroquinolones and Pyrazinamide Eff ectiveness, CHEST, vol.147, pp.1135-1178, 2015.

I. C. Shamputa, L. Jugheli, N. Sadradze, E. Willery, F. Portaels et al., Mixed infection and clonal representativeness of a single sputum sample in tuberculosis patients from a penitentiary hospital in Georgia, Respiratory Research, vol.7, p.99, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00089426

J. Van-embden, T. Van-gorkom, K. Kremer, R. Jansen, B. Van-der-zeijst et al., Genetic Variation and Evolutionary Origin of the Direct Repeat Locus of Mycobacterium tuberculosis Complex Bacteria, J Bacteriol, vol.182, pp.2393-401, 2000.

N. V. Nhung, N. B. Hoa, D. N. Sy, C. M. Hennig, and A. S. Dean, The Fourth National Anti-Tuberculosis Drug Resistance Survey in Viet Nam, 2015.

Y. Zhao, S. Xu, L. Wang, D. P. Chin, S. Wang et al., National Survey of DrugResistant Tuberculosis in China, New England Journal of Medicine, vol.366, pp.2161-70, 2012.

, report-national-tuberculosis-drs, 2006.

T. Ti, T. Lwin, T. T. Mar, W. Maung, P. Noe et al., National anti-tuberculosis drug resistance survey, 2002.

Q. H. Nguyen, L. Contamin, T. Nguyen, and A. Bañuls, Insights into the processes that drive the evolution of drug resistance in Mycobacterium tuberculosis, Evol Appl, vol.11, pp.1498-511, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01912302

X. Ma, H. Wang, Y. Deng, Z. Liu, Y. Xu et al., rpoB Gene Mutations and Molecular Characterization of Rifampin-Resistant Mycobacterium tuberculosis Isolates from Shandong Province, China. J Clin Microbiol, vol.44, pp.3409-3421, 2006.

C. Mani, N. Selvakumar, S. Narayanan, and P. R. Narayanan, Mutations in the rpoB Gene of Multidrug-Resistant Mycobacterium tuberculosis Clinical Isolates from India, J Clin Microbiol, vol.39, pp.2987-90, 2001.

. Miranda-ss-de, A. L. Kritski, I. Filliol, C. Mabilat, G. Panteix et al., Mutations in the rpoB gene of rifampicin-resistant Mycobacterium tuberculosis strains isolated in Brazil and France. Memórias do Instituto Oswaldo Cruz, vol.96, pp.247-50, 2001.

J. Sheng, J. Li, G. Sheng, H. Yu, H. Huang et al., Characterization of rpoB mutations associated with rifampin resistance in Mycobacterium tuberculosis from eastern China, Journal of Applied Microbiology, vol.105, pp.904-915, 2008.

M. Caws, P. M. Duy, D. Q. Tho, N. Lan, D. V. Hoa et al., Mutations Prevalent among Rifampin-and Isoniazid-Resistant Mycobacterium tuberculosis Isolates from a Hospital in Vietnam, J Clin Microbiol, vol.44, pp.2333-2340, 2006.

A. Valim, M. Rossetti, M. O. Ribeiro, and A. Zaha, Mutations in the rpoB Gene of MultidrugResistant Mycobacterium tuberculosis Isolates from Brazil, J Clin Microbiol, vol.38, pp.3119-3141, 2000.

V. Kapur, L. L. Li, S. Iordanescu, M. R. Hamrick, A. Wanger et al., Characterization by automated DNA sequencing of mutations in the gene (rpoB) encoding the RNA polymerase beta subunit in rifampin-resistant Mycobacterium tuberculosis strains from New York City and Texas, J Clin Microbiol, vol.32, pp.1095-1103, 1994.

Y. Lin, C. Tai, C. Li, C. Lin, and Z. Shi, Resistance profiles and rpoB gene mutations of Mycobacterium tuberculosis isolates in Taiwan, Immunology and Infection, vol.46, pp.266-70, 2013.

I. L. Bergval, R. Vijzelaar, D. Costa, E. R. Schuitema, A. Oskam et al., Development of Multiplex Assay for Rapid Characterization of Mycobacterium tuberculosis, Journal of Clinical Microbiology, vol.46, pp.689-99, 2008.

M. T. Mccammon, J. S. Gillette, D. P. Thomas, S. V. Ramaswamy, E. A. Graviss et al., Detection of rpoB Mutations Associated with Rifampin Resistance in Mycobacterium tuberculosis Using Denaturing Gradient Gel Electrophoresis, Antimicrob Agents Chemother, vol.49, pp.2200-2209, 2005.

Z. L. Berrada, S. Lin, T. C. Rodwell, D. Nguyen, G. F. Schecter et al., Rifabutin and Rifampin Resistance Levels and Associated rpoB Mutations in Clinical Isolates of Mycobacterium tuberculosis Complex, Diagn Microbiol Infect Dis, vol.85, pp.177-81, 2016.

A. V. Deun, L. Barrera, I. Bastian, L. Fattorini, H. Hoffmann et al., Mycobacterium tuberculosis Strains with Highly Discordant Rifampin Susceptibility Test Results, Journal of Clinical Microbiology, vol.47, pp.3501-3507, 2009.

P. J. Campbell, G. P. Morlock, R. D. Sikes, T. L. Dalton, B. Metchock et al., Molecular Detection of Mutations Associated with First-and Second-Line Drug Resistance Compared with Conventional Drug Susceptibility Testing of Mycobacterium tuberculosis, Antimicrob Agents Chemother, vol.55, pp.2032-2073, 2011.

B. Madison, B. Robinson-dunn, G. I. Gross, W. Lipman, H. Metchock et al.,

, Multicenter Evaluation of Ethambutol Susceptibility Testing of Mycobacterium tuberculosis by Agar Proportion and Radiometric Methods, J Clin Microbiol, vol.40, pp.3976-3985, 2002.

A. V. Groll, A. Martin, P. Jureen, S. Hoffner, P. Vandamme et al., Fluoroquinolone Resistance in Mycobacterium tuberculosis and Mutations in gyrA and gyrB, Antimicrobial Agents and Chemotherapy, vol.53, pp.4498-500, 2009.

C. E. Maus, B. B. Plikaytis, and T. M. Shinnick, Mutation of tlyA Confers Capreomycin Resistance in Mycobacterium tuberculosis, Antimicrob Agents Chemother, vol.49, pp.571-578, 2005.

G. P. Morlock, J. T. Crawford, W. R. Butler, S. E. Brim, D. Sikes et al., Phenotypic Characterization of pncA Mutants of Mycobacterium tuberculosis, Antimicrob Agents Chemother, vol.44, pp.2291-2296, 2000.

Q. H. Nguyen, C. Lucie, T. Hoa, N. V. Hung, N. Lan et al., Molecular analysis of pyrazinamide resistance in Mycobacterium tuberculosis in Vietnam highlights the high rate of pyrazinamide resistance-associated mutations in clinical isolates, Emerging Microbes & Infections, vol.6, p.86, 2017.

A. Sandgren, M. Strong, P. Muthukrishnan, B. K. Weiner, G. M. Church et al., Tuberculosis Drug Resistance Mutation Database, PLOS Medicine, vol.6, p.1000002, 2009.

G. B. Mackaness, The Intracellular Activation of Pyrazinamide and Nicotinamide. American Review of Tuberculosis and Pulmonary Diseases, vol.74, pp.718-746, 1956.

S. Li, B. Liu, M. Peng, M. Chen, W. Yin et al., Diagnostic accuracy of Xpert MTB/RIF for tuberculosis detection in different regions with different endemic burden: A systematic review and meta-analysis, PLOS ONE, vol.12, p.180725, 2017.

K. R. Steingart, I. Schiller, D. J. Horne, M. Pai, C. C. Boehme et al., Xpert® Mtb/Rif assay for pulmonary tuberculosis and rifampicin resistance in adults, Cochrane Database Syst Rev, pp.1-166, 2014.

M. T. Zaw, N. A. Emran, and Z. Lin, Mutations inside rifampicin-resistance determining region of rpoB gene associated with rifampicin-resistance in Mycobacterium tuberculosis, Journal of Infection and Public Health, vol.11, pp.605-615, 2018.

M. Heep, B. Brandstätter, U. Rieger, N. Lehn, E. Richter et al., Frequency of rpoB Mutations Inside and Outside the Cluster I Region in Rifampin-Resistant Clinical Mycobacterium tuberculosis Isolates, J Clin Microbiol, vol.39, pp.107-117, 2001.

. Anek-vorapong-r, C. Sinthuwattanawibool, L. J. Podewils, K. Mccarthy, K. Ngamlert et al., Validation of the GenoType® MTBDRplus assay for detection of MDR-TB in a public health laboratory in Thailand, BMC Infectious Diseases, vol.10, p.123, 2010.

Y. Bai, Y. Wang, C. Shao, Y. Hao, and Y. Jin, GenoType MTBDRplus Assay for Rapid Detection of Multidrug Resistance in Mycobacterium tuberculosis: A Meta-Analysis, PLoS One, vol.11, 2016.

Q. Liu, G. Li, C. Chen, J. Wang, L. Martinez et al., Diagnostic Performance of the GenoType MTBDRplus and MTBDRsl Assays to Identify Tuberculosis Drug Resistance in Eastern China, Chinese Medical Journal, vol.130, p.1521, 2017.

J. Zhang, S. Heng, L. Moullec, S. Refregier, G. Gicquel et al., A first assessment of the genetic diversity of Mycobacterium tuberculosis complex in Cambodia, BMC Infectious Diseases, vol.11, p.42, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-00670614

Y. Chen, C. Huang, W. Hsu, C. Cheng, H. Sun et al., Genetic diversity of the Mycobacterium tuberculosis East African-Indian family in three tropical Asian countries, Immunology and Infection, p.0, 2016.

A. S. Albanna, M. B. Reed, K. V. Kotar, A. Fallow, F. A. Mcintosh et al., Reduced Transmissibility of East African Indian Strains of Mycobacterium tuberculosis, PLOS ONE, vol.6, 2011.

Y. Pang, Y. Song, H. Xia, Y. Zhou, B. Zhao et al., Risk factors and clinical phenotypes of Beijing genotype strains in tuberculosis patients in China, BMC Infectious Diseases, vol.12, p.354, 2012.

J. R. Glynn, K. Kremer, M. W. Borgdorff, M. P. Rodriguez, D. Soolingen et al., Beijing/W genotype Mycobacterium tuberculosis and drug resistance, 2006.

O. S. Toungoussova, A. Mariandyshev, G. Bjune, P. Sandven, and D. A. Caugant, Molecular Epidemiology and Drug Resistance of Mycobacterium tuberculosis Isolates in the Archangel Prison in Russia: Predominance of the W-Beijing Clone Family, Clin Infect Dis, vol.37, pp.665-72, 2003.

T. Wada, S. Fujihara, A. Shimouchi, M. Harada, H. Ogura et al., High transmissibility of the modern Beijing Mycobacterium tuberculosis in homeless patients of Japan, Tuberculosis (Edinb), vol.89, pp.252-257, 2009.

V. Nguyen, A. Bañuls, T. Tran, K. Pham, T. S. Nguyen et al., Mycobacterium tuberculosis lineages and anti-tuberculosis drug resistance in reference hospitals across Viet Nam, BMC Microbiology, vol.16, p.167, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02003072

P. Lao, UNFPA Lao People's Democratic Republic | Results of Population and Housing Census 2015 (English Version), 2015.

F. Abebe and G. Bjune, The emergence of Beijing family genotypes of Mycobacterium tuberculosis and low-level protection by bacille Calmette-Guerin (BCG) vaccines: is there a link?, Clinical and Experimental Immunology, vol.145, pp.389-97, 2006.

T. Luo, I. Comas, D. Luo, B. Lu, J. Wu et al., Southern East Asian origin and coexpansion of Mycobacterium tuberculosis Beijing family with Han Chinese, Proceedings of the National Academy of Sciences, vol.112, pp.8136-8177, 2015.

J. R. Glynn, J. Whiteley, P. J. Bifani, K. Kremer, and D. Van-soolingen, Worldwide Occurrence of Beijing/W Strains of Mycobacterium tuberculosis: A Systematic Review, Emerg Infect Dis, vol.8, pp.843-852, 2002.

W. Mcdermott and R. Tompsett, Activation of pyrazinamide and nicotinamide in acidic environments in vitro, Am Rev Tuberc, vol.70, pp.748-54, 1954.

R. Báez-saldaña, G. Delgado-sánchez, L. García-garcía, L. P. Cruz-hervert, M. Montesinoscastillo et al., Isoniazid Mono-Resistant Tuberculosis: Impact on Treatment Outcome and Survival of Pulmonary Tuberculosis Patients in Southern Mexico, PLoS One, vol.11, 1995.

G. L. Hobby and T. F. Lenert, The Action of Rifampin Alone and in Combination with Other Antituberculous Drugs, Am Rev Respir Dis, vol.102, pp.462-467, 1970.

K. Aung, A. Van-deun, E. Declercq, M. R. Sarker, P. K. Das et al., Successful "9-month Bangladesh regimen" for multidrug-resistant tuberculosis among over 500 consecutive patients, Int J Tuberc Lung Dis, vol.18, pp.1180-1187, 2014.

M. L. Bastos, H. Hussain, K. Weyer, L. Garcia-garcia, V. Leimane et al., Treatment outcomes of patients with multidrug-resistant and extensively drug-resistant tuberculosis according to drug susceptibility testing to first-and second-line drugs: an individual patient data meta-analysis, Clin Infect Dis, vol.59, pp.1364-74, 2014.

K. Hirano, C. Abe, and M. Takahashi, Mutations in the rpoB Gene of Rifampin-Resistant Mycobacterium tuberculosis Strains Isolated Mostly in Asian Countries and Their Rapid Detection by Line Probe Assay, J Clin Microbiol, vol.37, pp.2663-2669, 1999.

A. R. Bahrmand, L. P. Titov, A. H. Tasbiti, S. Yari, and E. A. Graviss, High-Level Rifampin Resistance Correlates with Multiple Mutations in the rpoB Gene of Pulmonary Tuberculosis Isolates from the Afghanistan Border of Iran, J Clin Microbiol, vol.47, pp.2744-50, 2009.

M. Caws, P. M. Duy, D. Q. Tho, N. Lan, D. V. Hoa et al., Mutations Prevalent among Rifampin-and Isoniazid-Resistant Mycobacterium tuberculosis Isolates from a Hospital in Vietnam, J Clin Microbiol, vol.44, pp.2333-2340, 2006.

M. Seifert, D. Catanzaro, A. Catanzaro, and T. C. Rodwell, Genetic Mutations Associated with Isoniazid Resistance in Mycobacterium tuberculosis: A Systematic Review, PLoS One, vol.10, 2015.

U. Brammacharry and M. Muthaiah, Characterization of rpsL Gene Mutations in StreptomycinResistant Mycobacterium tuberculosis Isolates, American Journal of Microbiological Research, vol.2, pp.80-85, 2014.

B. Cuevas-córdoba, A. Cuellar-sánchez, A. Pasissi-crivelli, C. A. Santana-Álvarez, J. Hernández-illezcas et al., rrs and rpsL mutations in streptomycin-resistant isolates of Mycobacterium tuberculosis from Mexico, Immunology and Infection, vol.46, pp.30-34, 2013.

M. R. Farhat, B. J. Shapiro, K. J. Kieser, R. Sultana, K. R. Jacobson et al., Genomic analysis identifies targets of convergent positive selection in drug-resistant Mycobacterium tuberculosis, Nature Genetics, vol.45, pp.1183-1192, 2013.

N. S. Osorio, F. Rodrigues, S. Gagneux, and J. Pedrosa, Evidence for Diversifying Selection in a Set of Mycobacterium tuberculosis Genes in Response to Antibiotic-and Nonantibiotic-Related Pressure | Molecular Biology and Evolution | Oxford Academic, 2013.