W. Fox and D. A. Mitchison, State of the Art. Short-course chemotherapy for pulmonary tuberculosis, Am Rev Respir Dis, vol.111, pp.325-353, 1975.
URL : https://hal.archives-ouvertes.fr/hal-02625816

W. Fox, G. A. Ellard, and D. A. Mitchison, Studies on the treatment of tuberculosis undertaken by the British Medical Research Council Tuberculosis Units, 1946-1986, with relevant subsequent publications, Int J Tuberc Lung Dis, vol.3, pp.231-279, 1999.

S. Tiberi, N. Du-plessis, G. Walzl, M. J. Vjecha, M. Rao et al., Tuberculosis: progress and advances in development of new drugs, treatment regimens, and host-directed therapies, Lancet Infect Dis, vol.18, pp.183-98, 2018.

, New York: The Working Group for New TB Drugs; c2016

C. Lienhardt, A. M. Kraigsley, and C. F. Sizemore, Driving the Way to Tuberculosis Elimination: The Essential Role of Fundamental Research, Clin Infect Dis, vol.63, issue.3, pp.370-375, 2016.

C. Lienhardt and P. Nahid, Advances in clinical trial design for development of new TB treatments: A call for innovation, PLoS Med, vol.16, issue.3, p.1002769, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-02170758

, Global investments in Tuberculosis research and development: past, present, and future. Proceedings of the First WHO global ministerial conference on ending tuberculosis in the sustainable development era: a multisectoral response, pp.16-17, 2017.

R. Moscow, World Health Organization: Geneva, 2017.

, Report of the Technical Consultation on Advances in Clinical Trial Design for Development of New TB Treatments, Geneva: World Health Organization, vol.17, 2018.

D. A. Mitchison and J. Dickinson, Bactericidal mechanisms in short-course chemotherapy, Bull Int Union Tuberc, vol.53, pp.254-259, 1978.

D. A. Mitchison, Role of individual drugs in the chemotherapy of tuberculosis, Int J Tuberc Lung Dis, vol.4, issue.9, pp.796-806, 2000.

W. Mcdermott, Microbial persistence, Yale J Biol Med, vol.30, pp.257-291, 1958.

Y. Zhang, W. W. Yew, and M. R. Barer, Targeting persisters for tuberculosis control, Antimicrob Agents Chemoth, vol.5, pp.2223-2230, 2012.

C. Nathan and C. E. Barry, TB drug development: immunology at the table, Immunol Rev, vol.264, pp.308-326, 2015.

E. L. Nuermberger, T. Yoshimatsu, S. Tyagi, K. Williams, I. Rosenthal et al., Moxifloxacin-containing regimens of reduced duration produce a stable cure in murine tuberculosis, AJRCCM, vol.170, p.10, 2004.

R. Rustomjee, C. Lienhardt, T. Kanyok, G. R. Davies, and J. Levin, Mthiyane T, and the Gatifloxacin for TB (OFLOTUB) study team. A phase II study of the sterilizing activities of ofloxacin, gatifloxacin and moxifloxacin in pulmonary tuberculosis, Int J Tuberc Lung Dis, vol.12, issue.2, pp.128-138, 2008.

M. B. Conde, A. Efron, C. Loredo, D. Souza, G. Graça et al., Moxifloxacin versus ethambutol in the initial treatment of tuberculosis: a double-blind, randomised, controlled phase II trial, Lancet, vol.373, pp.1183-1189, 2009.

M. J. Imperial, P. Nahid, P. Phillips, G. R. Davies, K. Fielding et al., A Patient-Level Pooled Analysis of Treatment Shortening Regimens for Drug-Susceptible Pulmonary Tuberculosis, Nature Med, vol.24, issue.11, p.30397355, 2018.

E. I. Nielsen and L. E. Friberg, Pharmacokinetic-pharmacodynamic modeling of antibacterial drugs, Pharmacol Rev, vol.65, pp.1053-1090, 2013.

V. Dartois, The path of anti-tuberculosis drugs: from blood to lesions to mycobacterial cells, Nat Rev Microbiol, vol.12, issue.3, pp.159-67, 2014.

B. Prideaux, L. E. Via, M. D. Zimmerman, S. Eum, J. Sarathy et al., The association between sterilizing activity and drug distribution into tuberculosis lesions, Nat Med, vol.21, issue.10, pp.1223-1230, 2015.

D. A. Mitchison, Role of individual drugs in the chemotherapy of tuberculosis, Int J Tuberc Lung Dis, vol.4, issue.9, pp.796-806, 2000.

N. Strydom, S. V. Gupta, W. S. Fox, L. E. Via, H. Bang et al., Tuberculosis drugs' distribution and emergence of resistance in patient's lung lesions: A mechanistic model and tool for regimen and dose optimization, PLoS Med, vol.16, issue.4, 2019.

D. L. Clemens, B. Y. Lee, A. Silva, B. J. Dillon, S. Masle?a-gali? et al., Artificial intelligence enabled parabolic response surface platform identifies ultra-rapid near-universal TB drug treatment regimens comprising approved drugs, PLoS ONE, vol.14, issue.5, p.217670, 2019.

I. H. Bartelink, N. Zhang, R. J. Keizer, N. Strydom, P. J. Converse et al., New Paradigm for Translational Modeling to Predict Long-term Tuberculosis Treatment Response, Clin Transl Sci, vol.10, issue.5, pp.366-379, 2017.

R. S. Wallis and J. L. Johnson, The role of surrogate markers in the clinical evaluation of antituberculous chemotherapy, Curr Med Chem-Anti-Infective Agents, vol.4, issue.4, pp.1-8, 2005.

K. E. Dooley, D. Hanna, V. Mave, K. Eisenach, and R. M. Savic, Advancing the development of new tuberculosis treatment regimens: The essential role of translational and clinical pharmacology and microbiology

, PLoS Med, vol.16, issue.7, p.1002842, 2019.

R. M. Savic, M. Weiner, W. R. Mackenzie, M. Engle, W. C. Whitworth et al., Defining the optimal dose of rifapentine for pulmonary tuberculosis: Exposure-response relations from two phase II clinical trials, Clin Pharmacol Ther, vol.102, issue.2, pp.321-331, 2017.

E. M. Svensson, R. J. Svensson, T. Brake, L. Boeree, M. J. Heinrich et al., The Potential for Treatment Shortening With Higher Rifampicin Doses: Relating Drug Exposure to Treatment Response in Patients With Pulmonary Tuberculosis, Clin Infect Dis, vol.67, issue.1, pp.34-41, 2018.

A. M. Ginsberg and M. Spigelman, Challenges in tuberculosis drug research and development, Nat Med, vol.13, issue.3, pp.290-294, 2007.

P. Phillips, C. D. Mitnick, J. D. Neaton, P. Nahid, C. Lienhardt et al., Keeping phase III tuberculosis trials relevant: Adapting to a rapidly changing landscape, PLoS Med, vol.16, issue.3, p.1002767, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-02170885

P. L. Olliaro and M. Vaillant, Designing noninferiority tuberculosis treatment trials: Identifying practical advantages for drug regimens with acceptable effectiveness, PLoS Med, vol.16, issue.7, p.1002850, 2019.

K. Odem-davis and T. R. Fleming, A Simulation Study Evaluating Bio-Creep Risk in Serial Noninferiority Clinical Trials for Preservation of Effect, Stat Biopharm Res, vol.7, issue.1, pp.12-24, 2015.

A. J. Nunn, P. Phillips, S. K. Meredith, C. Y. Chiang, F. Conradie et al., A Trial of a Shorter Regimen for Rifampin-Resistant Tuberculosis, N Engl J Med, vol.380, pp.1201-1213, 2019.

P. Phillips, S. H. Gillespie, M. Boeree, N. Heinrich, R. Aarnoutse et al., Innovative trial designs are practical solutions for improving the treatment of tuberculosis, J Infect Dis, vol.205, issue.2, pp.250-257, 2012.

M. Cellamare, S. Ventz, E. Baudin, C. D. Mitnick, and L. Trippa, A Bayesian response-adaptive trial in tuberculosis: The endTB trial, Clin Trials, vol.14, pp.17-28, 2017.

N. Alipanah, L. Jarlsberg, C. Miller, N. N. Linh, D. Falzon et al., Adherence interventions and outcomes of tuberculosis treatment: A systematic review and meta-analysis of trials and observational studies, PLoS Med, vol.15, issue.7, 2018.

A. Vernon, K. Fielding, R. Savic, L. Dodd, and P. Nahid, The importance of adherence in tuberculosis treatment clinical trials and its relevance in explanatory and pragmatic trials, PLoS Med, vol.16, issue.12, p.1002884, 2019.

, Influence of Adherence to Treatment and Response of Cholesterol on Mortality in the Coronary Drug Project, N Engl J Med, vol.303, issue.18, pp.1038-1041, 1980.

I. Ford and J. Norrie, Pragmatic Trials, N Engl J Med, vol.375, pp.454-63, 2016.

J. D. Walley, A. N. Khan, J. N. Newell, and M. H. Khan, Effectiveness of the direct observation component of DOTS for tuberculosis: a randomised controlled trial in Pakistan, Lancet, vol.357, pp.664-669, 2001.

J. N. Newell, S. C. Baral, S. B. Pande, D. S. Bam, and P. Malla, Family-member DOTS and community DOTS for tuberculosis control in Nepal: cluster-randomised controlled trial, Lancet, vol.367, pp.903-909, 2006.

S. Thiam, A. M. Lefevre, F. Hane, A. Ndiaye, F. Ba et al., Improving adherence to tuberculosis treatment in a resource-poor setting: A Cluster Randomised Controlled Trial, JAMA, vol.297, issue.4, pp.380-386, 2007.

A. Gupta, M. D. Hughes, A. J. Garcia-prats, K. Mcintire, and A. C. Hesseling, Inclusion of key populations in clinical trials of new antituberculosis treatments: Current barriers and recommendations for pregnant and lactating women, children, and HIV-infected persons, PLoS Med, vol.16, issue.8, p.1002882, 2019.

L. Mckenna, M. Frick, C. Lee, D. Namutamba, L. Smit et al., A Community Perspective on the Inclusion of Pregnant Women in Tuberculosis Drug Trials, Clin Infect Dis, vol.65, issue.8, pp.1383-1387, 2017.

J. S. Mathad and A. Gupta, Tuberculosis in pregnant and postpartum women: epidemiology, management, and research gaps, Clin Infect Dis, vol.55, issue.11, pp.1532-1549, 2012.

A. Gupta, J. S. Mathad, S. M. Abdel-rahman, J. D. Albano, R. Botgros et al., Toward Earlier Inclusion of Pregnant and Postpartum Women in Tuberculosis Drug Trials: Consensus statements from an international expert panel, Clin Infect Dis, vol.62, issue.6, pp.761-769, 2016.

S. Nachman, A. Ahmed, F. Amanullah, M. C. Becerra, R. Botgros et al., Towards early inclusion of children in tuberculosis drugs trials: a consensus statement, Lancet Infect Dis, vol.15, issue.6, p.25957923, 2015.

E. D. Weld and K. E. Dooley, State-of-the-Art Review of HIV-TB Coinfection in Special Populations, Clin Pharmacol Ther, vol.104, issue.6, pp.1098-1109, 2018.

J. A. Tornheim and K. E. Dooley, Challenges of TB and HIV co-treatment: updates and insights, Curr Opin HIV AIDS, vol.13, issue.6, pp.486-491, 2018.

, New York: The Working Group for New TB Drugs; c2016, The Working Group for New TB Drugs, 2019.

, US Food and Drug Administration

C. Adebamowo, O. Bah-sow, F. Binka, R. Bruzzone, A. Caplan et al., Randomised controlled trials for Ebola: practical and ethical issues, Lancet, vol.18, issue.9952, pp.61734-61741, 2014.

C. Lienhardt, A. A. Vernon, M. Cavaleri, S. Nambiar, and P. Nahid, Development of new TB regimens: Harmonizing trial design, product registration requirements, and public health guidance, PLoS Med, vol.16, issue.9, p.1002915, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-02430787

G. Brigden, N. V. Nhung, A. Skrahina, N. Ndjeka, D. Falzon et al., Advances in clinical trial design for development of new TB treatments-Translating international tuberculosis treatment guidelines into national strategic plans: Experiences from Belarus, South Africa, and Vietnam, PLoS Med, vol.16, issue.10, p.1002896, 2019.

, End TB Strategy: global strategy and targets for tuberculosis prevention, care, and control after, 2014.

, Global Investments in Tuberculosis Research and Development: Past, Present, and Future