Scolaris Content Display Scolaris Content Display

Regímenes de tratamiento acortados versus régimen estándar para la tuberculosis pulmonar sensible a los fármacos

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Gillespie 2014 {published data only}

Gillespie SH, Crook AM, McHugh TD, Mendel CM, Meredith SK, Murray SR, et al. Four‐month moxifloxacin‐based regimens for drug‐sensitive tuberculosis. New England Journal of Medicine 2014;371(17):1577‐87. [DOI: 10.1056/NEJMoa1407426; NCT00864383]CENTRAL
NCT00864383. Controlled comparison of two moxifloxacin containing treatment shortening regimens in pulmonary tuberculosis (REMoxTB). https://clinicaltrials.gov/ct2/show/NCT00864383 (accessed 1 September 2018). CENTRAL
Phillips PJP, Mendel CM, Burger D A, Crook AM, Nunn AJ, Dawson R, et al. Limited role of culture conversion for decision‐making in individual patient care and for advancing novel regimens to confirmatory clinical trials. BMC Medicine 2016;14:19. [DOI: 10.1186/s12916‐016‐0565‐y]CENTRAL
Phillips PPJ, Mendel CM, Nunn AJ, McHugh TD, Crook AM, Hunt R, et al. A comparison of liquid and solid culture for determining relapse and durable cure in phase III TB trials for new regimens. BMC Medicine 2017;15(1):207. [DOI: 10.1186/s12916‐017‐0955‐9]CENTRAL

Jawahar 2013 {published and unpublished data}

CTRI/2012/10/003060. A study of the efficacy and tolerability of moxifloxacin and gatifloxacin containing regimens in the treatment of patients with sputum‐positive pulmonary tuberculosis. http://ctri.nic.in/Clinicaltrials/showallp.php?mid1=5124&EncHid=&userName=CTRI/2012/10/003060 (accessed 28 August 2018). [CTRI/2012/10/003060]CENTRAL
Jawahar MS, Banurekha VV, Paramasivan CN, Rahman F, Ramachandran R, Venkatesan P, et al. Randomized clinical trial of thrice‐weekly 4‐month moxifloxacin or gatifloxacin containing regimens in the treatment of new sputum positive pulmonary tuberculosis patients. PLOS One 2013;8(7):e67030. [PUBMED: 23843980]CENTRAL

Jindani 2014 {published data only (unpublished sought but not used)}

ISRCTN44153044. An international multicentre controlled clinical trial to evaluate high dose RIFApentine and a QUINolone in the treatment of pulmonary tuberculosis. https://doi.org/10.1186/ISRCTN44153044 (accessed 26 August 2018). [ISRCTN44153044]CENTRAL
Jindani A, Harrison TS, Nunn AJ, Phillips PPJ, Churchyard GJ, Charalambous S, et al. High‐dose rifapentine with moxifloxacin for pulmonary tuberculosis. New England Journal of Medicine 2014;371(17):1599‐608. CENTRAL

Merle 2014 {published data only}

Merle CS, Fielding K, Sow OB, Gninafon M, Lo MB, Mthiyane T, et al. A four‐month gatifloxacin‐containing regimen for treating tuberculosis. New England Journal of Medicine 2014;371(17):1588‐98. CENTRAL
Merle CS, Fielding K, Sow OB, Gninafon M, Lo MB, Mthiyane T, et al. Correction: A four‐month gatifloxacin‐containing regimen for treating tuberculosis. New England Journal of Medicine 2015;372(17):1677. [PUBMED: 25901444]CENTRAL
Merle CS, Sismanidis C, Sow OB, Gninafon M, Horton J, Lapujade O, et al. A pivotal registration phase III, multicenter, randomized tuberculosis controlled trial: design issues and lessons learnt from the Gatifloxacin for TB (OFLOTUB) project. Trials 2012;13:61. [DOI: 10.1186/1745‐6215‐13‐61]CENTRAL
NCT00216385. A controlled trial of a 4‐month quinolone‐containing regimen for the treatment of pulmonary tuberculosis. https://clinicaltrials.gov/ct2/show/NCT00216385 (accessed 30 June 2018). [NCT00216385]CENTRAL
Olliaro PL, Merle C, Mthiyane T, Bah B, Kassa F, Amukoye E, et al. Effects on the QT interval of a gatifloxacin‐containing regimen versus standard treatment of pulmonary tuberculosis. Antimicrobial Agents and Chemotherapy 2017;61(7):pii: e01834‐16. [DOI: 10.1128/AAC.01834‐16]CENTRAL

Velayutham 2014 {published and unpublished data}

CTRI/2008/091/000024. Randomised clinical trial to study the efficacy and tolerability of 3‐ and 4‐month regimens containing moxifloxacin in the treatment of patients with sputum smear and culture positive pulmonary tuberculosis. http://ctri.nic.in/Clinicaltrials/showallp.php?mid1=68&EncHid=&userName=CTRI/2008/091/000024 (accessed 28 August 2018). [CTRI/2008/091/000024]CENTRAL
Velayutham BV, Allaudeen IS, Sivaramakrishnan GN, Perumal V, Nair D, Chinnaiyan P, et al. Sputum culture conversion with moxifloxacin‐containing regimens in the treatment of patients with newly diagnosed sputum‐positive pulmonary tuberculosis in South India. Clinical Infectious Diseases 2014;59(10):e142‐9. [CTRI 2008/091/000024; PUBMED: 25028463]CENTRAL

Referencias de los estudios excluidos de esta revisión

Alavi 2009 {published data only}

Alavi SM. Comparative evaluation of three and six month therapeutic regimens for smear negative pulmonary tuberculosis. Jundishapur Journal of Microbiology 2009;2(1):14‐7. CENTRAL

Burman 2006 {published data only}

Burman WJ, Goldberg S, Johnson JL, Muzanye G, Engle M, Mosher AW, et al. Moxifloxacin versus ethambutol in the first 2 months of treatment for pulmonary tuberculosis. American Journal of Respiratory and Critical Care Medicine 2006;174(3):331‐8. [DOI: 10.1164/rccm.200603‐360OC]CENTRAL

Conde 2009 {published data only}

Conde MB, Efron A, Loredo C, De Souza GR, Graça NP, Cezar MC, et al. Moxifloxacin versus ethambutol in the initial treatment of tuberculosis: a double‐blind, randomised, controlled phase II trial. Lancet 2009;373(9670):1183‐9. CENTRAL

Conde 2016 {published data only}

Conde MB, Mello FC, Duarte RS, Cavalcante SC, Rolla V, et al. A phase 2 randomized trial of a rifapentine plus moxifloxacin‐based regimen for treatment of pulmonary tuberculosis. PLOS One 2016;11(5):e0154778. [DOI: 10.1371/journal.pone.0154778]CENTRAL

Dorman 2009 {published data only}

Dorman SE, Johnson JL, Goldberg S, Muzanye G, Padayatchi N, Bozeman L, et al. Substitution of moxifloxacin for isoniazid during intensive phase treatment of pulmonary tuberculosis. American Journal of Respiratory and Critical Care Medicine 2009;180(3):273‐80. [DOI: 10.1164/rccm.200901‐0078OC]CENTRAL

El‐Sadr 1998 {published data only}

El‐Sadr WM, Perlman DC, Matts JP, Nelson ET, Cohn DL, Salomon N, et al. Evaluation of an intensive intermittent‐induction regimen and duration of short‐course treatment for human immunodeficiency virus‐related pulmonary tuberculosis. Terry Beirn Community Programs for Clinical Research on AIDS (CPCRA) and the AIDS Clinical Trials Group (ACTG). Clinical Infectious Diseases 1998;26(5):1148–58. CENTRAL

Johnson 2009 {published data only}

Johnson JL, Hadad DJ, Dietze R, Maciel EL, Sewali B, Gitta P, et al. Shortening treatment in adults with noncavitary tuberculosis and 2‐month culture conversion. American Journal of Respiratory and Critical Care Medicine 2009;180(6):558‐63. [PUBMED: 19542476]CENTRAL

Kohno 1992 {published data only}

Kohno S, Koga H, Kaku M, Maesaki S, Hara K. Prospective comparative study of ofloxacin or ethambutol for the treatment of pulmonary tuberculosis. Chest 1992;102(6):1815‐8. [PUBMED: 1446494]CENTRAL

Rustomjee 2008 {published data only}

Rustomjee R, Lienhardt C, Kanyok T, Davies GR, Levin J, Mthiyane T, et al. A phase II study of the sterilising activities of ofloxacin, gatifloxacin and moxifloxacin in pulmonary tuberculosis. International Journal of Tuberculosis and Lung Disease 2008;12(2):128‐38. [PUBMED: 18230244]CENTRAL

Tuberculosis Research Centre 1986 {published data only}

Tuberculosis Research Centre, Madras, and National Tuberculosis Institute, Bangalore. A controlled clinical trial of 3‐ and 5‐month regimens in the treatment of sputum‐positive pulmonary tuberculosis in south India. American Review of Respiratory Disease 1986;134(1):27‐33. [DOI: 10.1164/arrd.1986.134.1.27]CENTRAL

Tuberculosis Research Centre 2002 {published data only}

Jawahar MS, Rahman F, Narayanan, et al. Shortening short course chemotherapy: a randomised clinical trial for treatment of smear positive pulmonary tuberculosis with regimens using ofloxacin in the intensive phase. Indian Journal of Tuberculosis 2002;49(1):27. CENTRAL

Referencias de los estudios en curso

CTRI/2008/091/000024 {published data only}

CTRI/2008/091/000024. Randomised clinical trial to study the efficacy and tolerability of 3‐ and 4‐month regimens containing moxifloxacin in the treatment of patients with sputum smear and culture positive pulmonary tuberculosis. http://ctri.nic.in/Clinicaltrials/showallp.php?mid1=68&EncHid=&userName=CTRI/2008/091/000024 (accessed 28 August 2018). [CTRI/2008/091/000024]CENTRAL

ISRCTN63579542 {published data only}

Shorter treatment for minimal TB in children (SHINE) Protocol Version 2.1. www.ctu.mrc.ac.uk/research/documents/tb_protocols/shine_protocol2.1 2015 (accessed 4 September 2018). [ ISRCTN63579542]CENTRAL
ISRCTN63579542. SHINE study. Shorter treatment for minimal TB in children. www.isrctn.com/ISRCTN63579542 (accessed 17 July 2018). CENTRAL

NCT02342886 {published data only}

NCT02342886. Shortening treatment by advancing novel drugs (STAND). https://clinicaltrials.gov/ct2/show/record/NCT02342886?term=pa‐824+moxifloxacin (accessed 28 July 2018). [NCT02342886]CENTRAL

NCT02410772 {published data only}

NCT02410772. Rifapentine‐containing tuberculosis treatment shortening regimens. https://clinicaltrials.gov/show/NCT02410772 (accessed 2 March 2018). [NCT02410772]CENTRAL

NCT02581527 {published data only}

NCT02581527. An international multicentre controlled clinical trial to evaluate 1200mg and 1800mg rifampicin daily in the reduction of treatment duration for pulmonary tuberculosis from 6 months to 4 months (RIFASHORT). https://clinicaltrials.gov/show/NCT02581527 (accessed 10 March 2018). [NCT02581527]CENTRAL

NCT02901288 {published data only}

Gao M, Gao J, Du J, Liu Y, Zhang Y, Ma L, et al. Efficacy of ultra‐short course chemotherapy for new smear positive drug susceptible pulmonary tuberculosis: study protocol of a multicenter randomized controlled clinical trial. BMC Infectious Diseases 2017;17:435. [DOI: 10.1186/s12879‐017‐2505‐7; NCT02901288]CENTRAL
NCT02901288. Shortened regimens for drug‐susceptible pulmonary tuberculosis. https://clinicaltrials.gov/ct2/show/NCT02901288 (accessed 10 July 2018). [NCT02901288]CENTRAL

Abraha 2015

Abraha I, Cherubini A, Cozzolino F, De Florio R, Luchetta ML, Rimland JM, et al. Deviation from intention to treat analysis in randomised trials and treatment effect estimates: meta‐epidemiological study. BMJ 2015;350:h2445. [DOI: 10.1136/bmj.h2445]

Agarwal 2009

Agrawal D, Udwadia ZF, Rodriguez C, Mehta A. Increasing incidence of fluoroquinolone resistant Mycobacterium tuberculosis in Mumbai, India. International Journal of Tuberculosis and Lung Disease 2009;13(1):79‐83.

Al‐Rifai 2017

Al‐Rifai RH, Pearson F, Critchley JA, Abu‐Raddad LJ. Association between diabetes mellitus and active tuberculosis: a systematic review and meta‐analysis. PLOS One 2017;12(11):e0187967. [10.1371/journal.pone.0187967]

Alipanah 2016

Alipanah N, Cattamanchi A, Menzies R, Hopewell PC, Chaisson RE, Nahid P. Treatment of non‐cavitary pulmonary tuberculosis with shortened fluoroquinolone‐based regimens: a meta‐analysis. International Journal of Tuberculosis and Lung Disease 2016;20(11):1522‐8. [DOI: 10.5588/ijtld.16.0217]

Andries 2005

Andries K, Verhasselt P, Guillemont J, Göhlmann HW, Neefs JM, Winkler H, et al. A diarylquinoline drug active on the ATP synthase of Mycobacterium tuberculosis. Science 2005;307(5707):223‐7.

Baker 2011

Baker MA, Harries AD, Jeon CY, Hart JE, Kapur A, Lönnroth K, et al. The impact of diabetes on tuberculosis treatment outcomes: a systematic review. BMC Medicine 2011;9:18. [DOI: 10.1186/1741‐7015‐9‐81]

Balshem 2011

Balshem H, Helfand M, Schunemann HJ, Oxman AD, Kunz R, Brozek J, et al. GRADE guidelines. 3. Rating the quality of evidence. Journal of Clinical Epidemiology 2011;64(4):401‐6.

Boeree 2017

Boeree MJ, Heinrich N, Aarnoutse R, Diacon AH, Dawson R, Rehal S, et al. High‐dose rifampicin, moxifloxacin, and SQ109 for treating tuberculosis: a multi‐arm, multi‐stage randomised controlled trial. Lancet Infectious Diseases 2017;17(1):39‐49. [DOI: 10.1016/S1473‐3099(16)30274‐2]

Conde 2011

Conde MB, Lapa E Silva JR. New regimens for reducing the duration of the treatment of drug‐susceptible pulmonary tuberculosis. Drug Development Research 2011;72(6):501‐8. [DOI: 10.1002/ddr.20456]

D'Ambrosio 2015

D'Ambrosio L, Centis R, Sotgiu G, Pontali E, Spanevello A, Migliori GB. New anti‐tuberculosis drugs and regimens: 2015 update. ERJ Open Research 2015;1(1):00010‐2015.

DAIDS 2009

U.S. Department of Health and Human Services, National Institutes of Health, National Institute of Allergy and Infectious Diseases, Division of AIDS (DAIDS). Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events, Version 1.0. [Updated August 2009].. Available from: https://rsc.niaid.nih.gov/sites/default/files/table‐for‐grading‐severity‐of‐adult‐pediatric‐adverse‐events.pdf (accessed 21 September 2018).

Dawson 2015

Dawson R, Diacon AH, Everitt D, van Niekerk C, Donald PR, Burger DA, et al. Efficiency and safety of the combination of moxifloxacin, pretomanid (PA‐824), and pyrazinamide during the first 8 weeks of antituberculosis treatment: a phase 2b, open‐label, partly randomised trial in patients with drug‐susceptible or drug‐resistant pulmonary tuberculosis. Lancet 2015;385(9979):1738‐47.

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG (editors). Chapter 9. Analysing data and undertaking meta‐analyses. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Devasia 2009

Devasia RA, Blackman A, Gebretsadik T, Griffin M, Shintani A, May C, et al. Fluoroquinolone resistance in Mycobacterium tuberculosis: the effect of duration and timing of fluoroquinolone exposure. American Journal of Respiratory and Critical Care Medicine 2009;180(4):365‐70. [DOI: 10.1164/rccm.200901‐0146OC]

Diacon 2012

Diacon AH, Donald PR, Pym A, Grobusch M, Patientia RF, Mahanyele R, et al. Randomized pilot trial of eight weeks of bedaquiline (TMC207) treatment for multidrug‐resistant tuberculosis: long‐term outcome, tolerability, and effect on emergence of drug resistance. Antimicrobial Agents and Chemotherapy 2012;56(6):3271‐6.

Diacon 2014

Diacon AH, Pym A, Grobusch MP, de los Rios JM, Gotuzzo E, Vasilyeva I, et al. Multidrug‐resistant tuberculosis and culture conversion with bedaquiline. New England Journal of Medicine 2014;371(8):723‐32.

Fergusson 2002

Fergusson D, Aaron SD, Guyatt G, Hebert, P. Post‐randomization exclusions: the intention to treat principle and excluding patients from analysis. BMJ 2002;325:652‐4.

Gelband 1999

Gelband H. Regimens of less than six months for treating tuberculosis. Cochrane Database of Systematic Reviews 1999, Issue 4. [DOI: 10.1002/14651858.CD001362]

Ginsberg 2010

Ginsberg AM. Tuberculosis drug development: progress, challenges, and the road ahead. Tuberculosis (Edinburgh, Scotland) 2010;90(3):162‐7.

Ginsburg 2003

Ginsburg AS, Grosset JH, Bishai WR. Fluoroquinolones, tuberculosis, and resistance. Lancet. Infectious Diseases 2003;3(7):432‐42.

Gler 2012

Gler MT, Skripconoka V, Sanchez‐Garavito E, Xiao H, Cabrera‐Rivero JL, Vargas‐Vasquez DE, et al. Delamanid for multidrug‐resistant pulmonary tuberculosis. New England Journal of Medicine 2012;366(23):2151‐60.

GRADEpro GDT [Computer program]

McMaster University (developed by Evidence Prime). GRADEpro GDT. Version accessed 12 November 2017. Hamilton (ON): McMaster University (developed by Evidence Prime), 2015.

Guyatt 2011a

Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, et al. GRADE guidelines. 1. Introduction ‐ GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 2011;64(4):383‐94. [PUBMED: 21195583]

Guyatt 2011b

Guyatt GH1, Oxman AD, Kunz R, Brozek J, Alonso‐Coello P, Rind D, et al. GRADE guidelines. 6. Rating the quality of evidence ‐ imprecision. Journal of Clinical Epidemiology 2011;64(12):1283‐93. [PUBMED: 21839614]

Guyatt 2011c

Guyatt GH, Oxman AD, Kunz R, Woodcock J, Brozek J, Helfand M, et al. GRADE guidelines. 7. Rating the quality of evidence ‐ inconsistency. Journal of Clinical Epidemiology 2011;64(12):1294‐302. [PUBMED: 21803546]

Haverkamp 2012

Haverkamp W, Kruesmann F, Fritsch A, van Veenhuyzen D, Arvis P. Update on the cardiac safety of moxifloxacin. Current Drug Safety 2012;7(2):149‐63. [PUBMED: 22873499]

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). Available from www.handbook.cochrane.org. The Cochrane Collaboration, 2011.

Hultcrantz 2017

Hultcrantz M, Rind D, Akl EA, Treweek S, Mustafa RA, Iorio A, et al. The GRADE Working Group clarifies the construct of certainty of evidence. Journal of Clinical Epidemiology 2017;87:4‐13.

Imperial 2018

Imperial MZ, Nahid P, Phillips PPJ, Davies GR, Fielding K, Hanna D, et al. A patient‐level pooled analysis of treatment‐shortening regimens for drug‐susceptible pulmonary tuberculosis. Nature Medicine 2018;24(11):1708‐15. [DOI: 10.1038/s41591‐018‐0224‐2]

Jeon 2008

Jeon CY, Murray MB. Diabetes mellitus increases the risk of active tuberculosis: a systematic review of 13 observational studies. PLOS Medicine 2008;5:e152. [PUBMED: 10.1371/journal.pmed.0050152]

Kerantzas 2017

Kerantzas CA, Jacobs WR. Origins of combination therapy for tuberculosis: lessons for future antimicrobial development and application. mBio 2017;8(2):e01586‐16.

Lee 2016

Lee HW, Lee JK, Kim E, Yim J‐J, Lee C‐H. The effectiveness and safety of fluoroquinolone‐containing regimen as a first‐line treatment for drug‐sensitive pulmonary tuberculosis: a systematic review and meta analysis. PLOS One 2015;11(7):e0159827. [10.1371/ journal.pone.015982]

Lessem 2015

Lessem E, Cox H, Daniels C, Furin J, McKenna L, Mitnick CD, et al. Access to new medications for the treatment of drug‐resistant tuberculosis: patient, provider and community perspectives. International Journal of Infectious Diseases 2015;32:56‐60.

Li 2016

Li D, Wang T, Shen S, Cheng S, Yu J, Zhang Y, et al. Effects of flouroquinolones in newly diagnosed, sputum‐positive tuberculosis therapy: a systematic review and network meta analysis. PLOS One 2015;10(12):e0145066. [10.1371/ journal.pone.0145066]

Lienhardt 2010

Lienhardt C, Vernon A, Raviglione MC. New drugs and new regimens for the treatment of tuberculosis: review of the drug development pipeline and implications for national programmes. Current Opinion in Pulmonary Medicine 2010;16(3):186‐93.

Liu 2017

Liu Q, Li W, Xue M, Chen Y, Du X, Wang C, et al. Diabetes mellitus and the risk of multidrug resistant tuberculosis: a meta‐analysis. Scientific Reports 2017;7(1):1090. [DOI: 10.1038/s41598‐017‐01213‐5]

Ma 2010

Ma Z, Lienhardt C, McIlleron H, Nunn AJ, Wang X. Global tuberculosis drug development pipeline: the need and the reality. Lancet 2010;375(9731):2100‐9.

Matsumoto 2006

Matsumoto M, Hashizume H, Tomishige T, Kawasaki M, Tsubouchi H, Sasaki H, et al. OPC‐67683, a nitro‐dihydro‐imidazooxazole derivative with promising action against tuberculosis in vitro and in mice. PLOS Medicine 2006;3(11):e466.

Mitchison 1985

Mitchison DA. The action of antituberculosis drugs in short‐course chemotherapy. Tubercle 1985;66(3):219‐25.

Moadebi 2007

Moadebi S, Harder CK, Fitzgerald MJ, Elwood KR, Marra F. Fluoroquinolones for the treatment of pulmonary tuberculosis. Drugs 2007;67(14):2077‐99.

Munsiff 2006

Munsiff SS, Kambili C, Ahuja SD. Rifapentine for the treatment of pulmonary tuberculosis. Clinical Infectious Diseases 2006;43(11):1468‐75.

Review Manager 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Rifat 2018

Rifat D, Prideaux B, Savic RM, Urbanowski ME, Parsons TL, Luna B, et al. Pharmacokinetics of rifapentine and rifampin in a rabbit model of tuberculosis and correlation with clinical trial data. Science Translational Medicine 2018;10(435):pii: eaai7786. [DOI: 10.1126/scitranslmed.aai7786]

Riza 2014

Riza AL, Pearson F, Ugarte‐Gil C, Alisjahbana B, van de Vijver S, Panduru NM, et al. Clinical management of concurrent diabetes and tuberculosis and the implications for patient services. Lancet Diabetes and Endocrinology 2014;2(9):740‐53. [PUBMED: 25194887]

Ruan 2016

Ruan Q, Liu Q, Sun F, Shao L, Jin J, Yu S, et al. Moxifloxacin and gatifloxacin for initial therapy of tuberculosis: a meta‐analysis of randomized clinical trials. Emerging Microbes and Infections 2016;5:e12. [DOI: 10.1038/emi.2016.12]

Rubinstein 2002

Rubinstein E, Camm J. Cardiotoxicity of fluoroquinolones. Journal of Antimicrobial Chemotherapy 2002;49(4):593‐6.

Sacksteder 2012

Sacksteder KA, Protopopova M, Barry CE, Andries K, Nacy CA. Discovery and development of SQ109: a new antitubercular drug with a novel mechanism of action. Future Microbiology 2012;7(7):823‐37.

Savic 2017

Savic RM, Weiner M, MacKenzie WR, Engle M, Whitworth WC, Johnson JL, et al. Defining the optimal dose of rifapentine for pulmonary tuberculosis: exposure‐response relations from two phase II clinical trials. Clinical Pharmacology and Therapeutics 2017;102(2):321‐31. [PUBMED: 28124478]

Schluger 2013

Schluger NW. Fluoroquinolones in the treatment of tuberculosis: which is best?. American Journal of Respiratory and Critical Care Medicine 2013;188(7):768‐9.

Schünemann 2011

Schünemann HJ, Oxman AD, Vist GE, Higgins JPT, Deeks JJ, Glasziou P, Guyatt GH. Chapter 12. Interpreting results and drawing conclusions. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org. The Cochrane Collaboration.

Schünemann 2013

Schünemann H, Brożek J, Guyatt G, Oxman A, editors. GRADE Handbook for Grading Quality of Evidence and Strength of Recommendations. Updated October 2013. Available from guidelinedevelopment.org/handbook. The GRADE Working Group.

Selvakumar 2015

Selvakumar N, Kumar V, Balaji S, Prabuseenivasan S, Radhakrishnan R, Sekar G, et al. High rates of ofloxacin resistance in Mycobacterium tuberculosis among both new and previously treated patients in Tamil Nadu, South India. PLOS One 2015;10(3):e0117421. [10.1371/ journal.pone.0117421]

Sotgiu 2012

Sotgiu G, Centis R, D'Ambrosio L, Alffenaar JWC, Anger HA, Caminero JA, et al. Efficacy, safety and tolerability of linezolid containing regimens in treating MDR‐TB and XDR‐TB: systematic review and meta‐analysis. European Respiratory Journal 2012;40(6):1430‐42.

Svensson 2018

Svensson EM, Svensson RJ, Te Brake LHM, Boeree MJ, Heinrich N, Konsten S, et al. The potential for treatment shortening with higher rifampicin doses: relating drug exposure to treatment response in patients with pulmonary tuberculosis. Clinical Infectious Diseases 2018;67(1):34‐41. [DOI: 10.1093/cid/ciy026]

Tegegne 2018

Tegegne BS, Mengesha MM, Teferra AA, Awoke MA, Habtewold TD. Association between diabetes mellitus and multi‐drug‐resistant tuberculosis: evidence from a systematic review and meta‐analysis. Systematic Reviews 2018;7:161. [DOI: 10.1186/s13643‐018‐0828‐0]

Temple 1999

Temple ME, Nahata MC. Rifapentine: its role in the treatment of tuberculosis. Annals of Pharmacotherapy 1999;33(11):1203‐10.

Wallis 2010

Wallis RS, Jakubiec WM, Kumar V, Silvia AM, Paige D, Dimitrova D, et al. Pharmacokinetics and whole‐blood bactericidal activity against Mycobacterium tuberculosis of single doses of PNU‐100480 in healthy volunteers. Journal of Infectious Diseases 2010;202(5):745‐51.

WHO 2010

World Health Organization. Guidelines for Treatment of Tuberculosis. 4th edition. 2010. WHO/HTM/TB/2009.420. www.who.int/tb/publications/2010/9789241547833/en/ (accessed 1 July 2017).

WHO 2015

World Health Organization. The End TB Strategy. Global Strategy and Targets for Tuberculosis Prevention, Care and Control After 2015. WHO/HTM/TB/2015.19. www.who.int/tb/post2015_strategy/en/ (accessed 1 August 2017).

WHO 2017

World Health Organization. Bending the Curve ‐ Ending TB: Annual Report 2017. apps.who.int/iris/bitstream/10665/254762/1/978929022584‐eng.pdf. WHO Regional Office for South‐East Asia, (accessed 12 July 2017).

WHO 2018

World Health Organization. Global Tuberculosis Report 2018. www.who.int/tb/publications/global_report/en/. Geneva: World Health Organization, (accessed prior to 24 October 2019). [Licence: CC BY‐NC‐SA 3.0 IGO.]

Williams 2009

Williams KN, Stover CK, Zhu T, Tasneen R, Tyagi S, Grosset JH, et al. Promising antituberculosis activity of the oxazolidinone PNU‐100480 relative to that of linezolid in a murine model. Antimicrobial Agents and Chemotherapy 2009;53(4):1314‐9.

Wood 2011

Wood R, Lawn SD, Caldwell J, Kaplan R, Middelkoop K, Bekker LG. Burden of new and recurrent tuberculosis in a major South African city stratified by age and HIV‐status. PLOS One 2011;6(10):e25098. [DOI: 10.1371/journal.pone.0025098]

Ziganshina 2013

Ziganshina LE, Titarenko AF, Davies GR. Fluoroquinolones for treating tuberculosis (presumed drug‐sensitive). Cochrane Database of Systematic Reviews 2013, Issue 6. [DOI: 10.1002/14651858.CD004795.pub4]

Zumla 2014

Zumla AI, Gillespie SH, Hoelscher M, Philips PPJ, Cole ST, Abubakar I, et al. New antituberculosis drugs, regimens, and adjunct therapies: needs, advances, and future prospects. Lancet Infectious Diseases 2014;14(4):327‐40.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Gillespie 2014

Methods

Study design: multi‐centre, randomized, parallel‐group, double‐blind (participant, care provider, investigator, outcomes assessor), 3‐armed, placebo‐controlled, non‐inferiority trial

Study period: January 2008 to February 2014

Recruitment sites: 47 sites in 9 countries

Countries where the trial was undertaken: South Africa, India, Tanzania, Kenya, Thailand, Malaysia, Zambia, China, Mexico

Length of follow‐up: 18 months after randomization (1 year after treatment completion)

Participants

No. of participants randomized: 1931

Interventions: 1291 (636 to ethambutol group; 655 to isoniazid group)

Control: 640 .

Age: > 35 years, 37% in isoniazid group, 39% in ethambutol group, and 40% in control group

Gender: male 70% in ethambutol group, 68% in isoniazid group, 70% in control group

Inclusion criteria:

  • 2 sputum specimens positive for tubercle bacilli on direct smear microscopy, of which 1 was confirmed by the REMoxTB study laboratory at the local laboratory

  • No history of previous anti‐tuberculosis chemotherapy

  • Aged 18 years and older

  • Firm home address that is readily accessible for visiting and willingness to inform the study team of any change in address and follow‐up period

  • Agreement to participate in the study and to give a sample of blood for HIV testing

  • Negative pregnancy test (women of childbearing potential)

  • Pre‐menopausal women must be using a barrier form of contraception or must be surgically sterilized or have an IUCD in place

  • Laboratory parameters performed at least 14 days before enrolment

    • Serum aspartate transaminase (AST) and alanine transaminase (ALT) activity less than 3 times upper limit of normal

    • Serum total bilirubin level less than 2.5 times upper limit of normal

    • Creatinine clearance (CrCl) level greater than 30 mL/min

    • Haemoglobin level at least 7.0 g/dL

    • Platelet count at least 50 x 10⁹ cells/L

    • Serum potassium greater than 3.5 mmol/L

Exclusion criteria:

  • Patients unable to take oral medication

  • Previously enrolled in this study

  • Receiving any investigational drug in the past 3 months or an antibiotic active against M tuberculosis

  • Pregnancy or breastfeeding

  • Any condition that may prove fatal during the first 2 months of the study period

  • Severe tuberculosis with high risk of a poor outcome (e.g. meningitis)

  • Pre‐existing condition likely to prejudice the response to, or assessment of, treatment; a condition likely to lead to uncooperative behaviour

  • Contraindication to any medications in the study regimens

  • Congenital or sporadic cardiac syndrome or taking medications that could result in QTc prolongation

  • Patients already receiving antiretroviral therapy

  • Weight less than 35 kg

  • HIV infection with CD4 count less than 250 cells/μL

  • End‐stage liver failure (class Child‐Pugh C)

  • Patients whose initial isolate was shown to be multiple drug resistant or monoresistant to rifampicin, or to any fluoroquinolone

Proportion with HIV seropositivity: 7% overall (and in intervention and control groups)

Proportion with cavitation: 71% overall (69% and 70% in intervention groups and 72% in control groups)

Baseline drug resistance: isoniazid: 7% overall (6% in control arm and 7% in each intervention arm); pyrazinamide: 2% overall (1% in each intervention arm)

Interventions

Interventions: 4‐month (17‐week) ATT regimen

Isoniazid group (moxifloxacin for 17 weeks substituting ethambutol): N = 655; 568 eligible, 514 completed (78% of those randomized; 91% of those eligible)

8 weeks of moxifloxacin, isoniazid, rifampicin, pyrazinamide, + ethambutol placebo administered daily, followed by

9 weeks of moxifloxacin, isoniazid, and rifampicin, followed by 9 weeks of isoniazid and rifampicin placebo

Ethambutol group (moxifloxacin for 17 weeks substituting isoniazid): N = 636; 551 eligible, 524 completed (82% of those randomized; 91% of those eligible)

8 weeks of moxifloxacin, ethambutol, rifampicin, pyrazinamide + isoniazid placebo administered daily, followed by

9 weeks of moxifloxacin and rifampicin + isoniazid placebo daily, followed by

9 weeks of isoniazid and rifampicin placebo

Control: 6‐month (26‐week) ATT regimen: N = 640; 555 eligible, 510 completed (80% of those randomized; 92% of those eligible)

8 weeks of isoniazid, rifampicin, ethambutol, pyrazinamide, and moxifloxacin placebo given daily, followed by

9 weeks of isoniazid, rifampicin, and moxifloxacin placebo given daily, followed by

9 weeks of isoniazid and rifampicin

Dosage: Moxifloxacin 400 mg, isoniazid 300 mg; rifampicin, ethambutol, and pyrazinamide were dosed based on weight

Outcomes

Outcomes reported and used in this review:

  • Relapse within 18 months after randomization (relapse strains were those shown to be identical on 24‐locus MIRU analysis)

  • Death from any cause

  • Rates of treatment discontinuation

  • Sputum smear/culture positivity at 8 weeks

  • Proportion with bacteriologically or clinically defined failure

  • Serious adverse events

  • Other adverse events

Outcomes sought but not reported:

  • Development of secondary drug resistance to anti‐tuberculosis drugs, identified by drug susceptibility testing

Outcomes reported but not used in this review:

  • Composite unfavourable outcome (clinical or bacteriologic failure or relapse within 18 months after randomization) (non‐inferiority was defined as a between‐group difference of less than 6 percentage points in the upper boundary of the 2‐sided 97.5% Wald confidence interval for the difference in proportion of patients with an unfavourable outcome)

  • Re‐treated for tuberculosis

  • Time to first culture‐negative sputum

  • Pharmakokinetic data (substudy reported separately)

Notes

Funding: Global Alliance for TB Drug Development (supported by multiple international donor agencies and local agencies and institutions in participating countries). Bayer Healthcare donated moxifloxacin and Sanofi donated rifampicin

Treatment supervision: treatment was given daily and was observed according to guidelines at the study site

Follow‐up method: following screening and baseline visits, there were 8 weekly visits followed by 8 visits until 18 months after randomization. Safety analysis was performed at the screening visit and thereafter at weeks 2, 8, 12, and 17

Trial registration ID: NCT00864383 (retrospectively registered: registered March 2009; study start January 2008)

Acronym: REMoxTB

Comment: data from both moxifloxacin‐containing shorter regimens were combined and compared with data from the standard treatment regimen

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from report: "Randomization was performed with the use of lists with blocks of variable sizes that were stratified according to the patient weight group and study centre"

Allocation concealment (selection bias)

Low risk

Quote from report: "During randomization, patients were assigned a unique study number selected sequentially from the appropriate randomization list that corresponded to the treatment pack allocated"

Quote from report: "Only statisticians who were responsible for preparing the reports for the independent data and safety monitoring committee and essential manufacturing and distribution staff members had access to the list of identifiers matched to the intervention"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote from study protocol: "This will be a blinded study with matching placebo for each of the study medicines except for pyrazinamide"

Quote from trial registration document: "Masking: quadruple (participant, care provider, investigator, outcomes assessor)"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote from trial registration document: "Masking: quadruple (participant, care provider, investigator, outcomes assessor)"

Incomplete outcome data (attrition bias): At the end of ATT (Treatment failure, positive sputum culture, treatment discontinuation, adverse events)

Low risk

Quote from report: "Of the 1931 patients who underwent randomization, 89% in the isoniazid group, 92% in the ethambutol group, and 89% in the control group met the requirements for treatment adherence, which was based on receipt of approximately 80% of the assigned regimen"

Comment: the modified‐intention‐to‐treat analysis used included 87% of those randomized to the combined moxifloxacin‐containing ATT regimens and 87% of those randomized to standard regimens. A sensitivity analysis included 94% of those randomized to both regimens

Incomplete outcome data (attrition bias): At the end of follow‐up (Relapse, deaths)

Low risk

Of those randomized, 91% of those allotted to the 2 moxifloxacin combination therapy arms and 92% allotted to control treatment were included in the modified intention‐to‐treat analyses. Results of the per‐protocol and modified intention‐to‐treat analyses were consistent

Selective reporting (reporting bias)

Low risk

Although the trial was retrospectively registered, all pre‐stated outcomes listed in the trial registration document and protocol were published with no evidence of selective reporting

Other bias

Low risk

Quote from report: "Bayer Healthcare donated moxifloxacin, and Sanofi donated rifampin. Neither company had any role in the study design, data accrual, data analysis, or manuscript preparation. Representatives of Bayer Healthcare reviewed the manuscript but did not suggest revisions"

Jawahar 2013

Methods

Study design: randomized, open‐label, parallel‐group, 3‐armed, active comparator, equivalence trial

Study period: started May 2004 for an anticipated duration of 5 years; terminated early (between February and October 2006) due to high recurrence rates in the shorter treatment arms

Recruitment sites: Chennai and Madurai

Country where the trial was undertaken: India

Length of follow‐up: 24 months after treatment completion

Participants

No. of participants randomized: 429 (of 1200 anticipated)

Intervention groups: 259 (gatifloxacin regimen 141; moxifloxacin regimen 118)

Control group: 170

Age: < 40 years 72% (gatifloxacin 66%; moxifloxacin 77%; control 73%)

Gender: male 74% (gatifloxacin 76%; moxifloxacin 72%; control 72%)

Inclusion criteria:

  • Adult patients 18 years or older with newly diagnosed pulmonary tuberculosis with at least 2 positive sputum cultures

  • Resident within a designated study area and permitted home visits

Exclusion criteria:

  • Those with previous treatment for tuberculosis exceeding 30 days, weighing < 30 kg, pregnant or lactating women

  • Those with concomitant diabetes mellitus, severe systemic hypertension, epilepsy, serious forms of extrapulmonary tuberculosis, or HIV infection

Proportion with HIV seropositivity: nil (excluded)

Proportion with cavitation: not reported

Baseline drug resistance: isoniazid 7% overall (gatifloxacin 4%; moxifloxacin 1.2%; control 12%); rifampicin 0.2% overall (moxifloxacin 1%); ofloxacin 1.7% overall (gatifloxacin 2%; control 3%); isoniazid and ethambutol 0.4% overall (1% in each intervention arm); isoniazid and ofloxacin 0.2% overall (control 1%)

Interventions

Interventions: 4‐month ATT regimens

Gatifloxacin regimen (gatifloxacin replacing ethambutol): N = 141; 136 eligible, 131 completed (93% of those randomized; 96% of those eligible)

2 months of gatifloxacin, isoniazid, rifampicin, and pyrazinamide thrice weekly, followed by

2 months of gatifloxacin, isoniazid, and rifampicin thrice weekly

Moxifloxacin regimen (moxifloxacin replacing ethambutol): N = 118; 115 eligible, 113 completed (96% of those randomized; 98% of those eligible)

2 months of moxifloxacin, isoniazid, rifampicin, and pyrazinamide thrice weekly, followed by

2 months of moxifloxacin, isoniazid, and rifampicin thrice weekly

Control: 6‐month ATT regimen: N = 170; 165 eligible, 159 completed (94% of those randomized; 96% of those eligible)

2 months of ethambutol, isoniazid, rifampicin, and pyrazinamide thrice weekly, followed by

4 months of isoniazid and rifampicin thrice weekly

Dosage: gatifloxacin or moxifloxacin 400 mg, rifampicin 450 or 600 mg, depending on body weight (< 60 kg or ≥ 60 kg), pyrazinamide 1500 mg, and isoniazid 600 mg

Outcomes

Outcomes reported and used in this review:

  • Recurrence of tuberculosis among those with a favourable response at the end of treatment

  • Death from any cause

  • Rates of treatment discontinuation

  • Sputum smear/culture positivity at 8 weeks

  • Proportion with bacteriologically or clinically defined failure

  • Serious adverse events

  • Other adverse events

  • Development of secondary drug resistance to anti‐tuberculosis drugs, identified by drug susceptibility testing

Notes

Funding: Tuberculosis Research Centre of the Indian Council of Medical Research

Follow‐up method: a physician examined the patient every month and recorded adherence to treatment, any adverse drug reactions, and the clinical response. Sputum specimens were examined every month by microscopy and culture: 2 (2 overnight and 1 spot) during the treatment phase, and 2 (1 overnight and 1 spot) during the follow‐up phase

Treatment supervision: directly observed, thrice‐weekly treatment in all arms

Trial registration ID: CTRI/2012/10/003060; retrospectively registered (trial commenced May 2004; trial registered 15/10/2012, after termination)

Comment: the data safety monitoring board recommended termination of both intervention arms in 2006 due to high tuberculosis recurrence rates in the 2 arms compared to the standard 6‐month regimen

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from report: "Restricted random allocation sequences were generated by a biostatistician using random number tables, separately for the two strata and sealed envelopes were used to assign regimens"

Quote from trial registration document: "Stratified block randomization"

Allocation concealment (selection bias)

High risk

Quote from report: "Patients were enrolled by the physicians, and when ready for allocation, the biostatistician drew the regimen from sealed envelopes. Allocation was stratified on sputum smear grading and extent of lesions in chest x ray"

Quote from report: "The study design envisaged enrolling 400 patients in each arm in a 1∶1:1 ratio. However, due to the non‐availability of one of the test drugs (M), patients were enrolled initially in a 1∶1 ratio in the G and control regimen arms commencing in May 2004. Subsequently, when M became available (May 2005), patients were enrolled to the G, M, and control regimen arms in a 1∶2:1 ratio to compensate for the delay in recruiting to the moxifloxacin arm at the onset"

Quotes from correspondence with study authors: "When the first patient on Moxifloxacin was allocated, there were 110 patients randomised to the Gatifloxacin regimen and 110 to the Control regimen. The last patient was allocated to the Gatifloxacin regimen on 3 February 2006"

Comment: alteration of recruitment ratios raises serious concerns that allocation concealment was compromised. Even though biostatisticians implemented allocation after clinicians confirmed eligibility, by the time the first patient was recruited to the moxifloxacin regimen in May 2005, 110 allocated to the gatifloxacin regimen (80% of 136 eligible among those finally recruited), and 110 allocated to the control regimen (67% of the 165 eligible) had already been recruited. This would have alerted investigators that most of those to be recruited over the following year would be allocated to the moxifloxacin regimen. In addition, premature termination of the trial, combined with the alteration in allocation ratios, appears to have led to imbalance in the numbers recruited to the gatifloxacin (141), moxifloxacin (118), and control (170) regimens that is not explained, given that block randomization was used. There were also baseline imbalances in proportions resistant at baseline to any of the anti‐tuberculosis drugs tested (6%, 3%, and 16% respectively)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

This was an open‐label trial; given the likelihood that allocation concealment was compromised, treating personnel may have had knowledge of allocation. However, it is unlikely that this led to performance bias

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote from report: "Sputum specimens were given identification laboratory numbers, and bacteriological investigations were carried out by technicians who were blinded to the clinical status of the patient and the regimen. ECG was done every month"

Comment: although bacteriologic outcomes were done blind to treatment allocation, clinical efficacy and safety outcomes were undertaken by study personnel. But results at the time of termination favouring the standard and not experimental interventions suggest that detection bias was unlikely

Incomplete outcome data (attrition bias): At the end of ATT (Treatment failure, positive sputum culture, treatment discontinuation, adverse events)

Low risk

In the combined fluoroquinolone arms, 228 of 259 (88%) completed treatment compared to 152 of 170 (89%) in the control arms. Study results did not differ between per‐protocol and modified intention‐to‐treat analyses (that excluded only late‐screening failures). The modified‐ITT analysis included 97%, 98%, and 97% of those in the gatifloxacin, moxifloxacin, and standard ATT arms, respectively. The results of both analyses were consistent

Incomplete outcome data (attrition bias): At the end of follow‐up (Relapse, deaths)

Low risk

In the combined fluoroquinolone arms, 230 of 259 (89%) were assessed for tuberculosis recurrence compared to 154 of 170 (91%) in the control arm. Per‐protocol and modified intention‐to‐treat analyses did not significantly alter the results. Early termination led to recruitment of only a third of the estimated 1200 participants required to prove equivalence, but although this reduces the power of the trial to detect equivalence, lack of differential attrition, with similar reasons for exclusion, is unlikely to affect the reported relative effect estimates

Selective reporting (reporting bias)

Low risk

Although the trial was retrospectively registered, stated outcomes in trial registry documents and in the online study protocol were available in the trial report and do not suggest selective reporting

Other bias

Low risk

The trial was terminated early at the recommendation of the Data Safety Monitoring Board after an interim analysis showed high recurrence rates in the fluoroquinolone arms compared to the control arm. Because this was a planned interim analysis, it is unlikely to have introduced bias, other than that discussed under allocation concealment

Jindani 2014

Methods

Study design: randomized, multi‐centre, parallel‐group, open‐label, 3‐arm, active‐controlled, equivalence trial

Study period: August 15, 2008, and August 1, 2011

Recruitment sites: Worcester, Johannesburg, Harare, Marondera, Francistown, and Macha

Countries where the trial was undertaken: Botswana, South Africa, Zambia, and Zimbabwe

Length of follow‐up: 18 months after randomization in 86%; in Botswana and South Africa, 6% of those randomized in the last 6 months of enrolment were followed up for 12 to 15 months and 8% for 15 to 18 months

Participants

No. of participants randomized: 827 (of the estimated sample of 1095)

Interventions: 275 in 4‐month regimen (277 in 6‐month regimen)

Control group: 275 in the control regimen

Age: 18 to 34 years 61% in the control regimen and 68% in the 4‐month regimen

Gender: male 64% in the control regimen, 63% in the 4‐month regimen

Inclusion criteria:

  • Newly diagnosed pulmonary tuberculosis

  • 2 sputum specimens positive for tubercle bacilli on direct smear microscopy

  • Either no previous anti‐tuberculosis chemotherapy or less than 2 weeks of previous chemotherapy at enrolment

  • Aged 18 years and older

  • Firm home address that is readily accessible for visiting and intending to remain there or within the recruitment area for the entire treatment and follow‐up period

  • Willing to agree to participate in the study and to give a sample of blood for HIV testing (and in Botswana to have HIV status disclosed to them)

  • Pre‐menopausal women must be using a barrier form of contraception or must be surgically sterilized or have an IUCD in place for the duration of the treatment phase

Exclusion criteria:

  • Any condition (except HIV infection) that may prove fatal during the study period

  • Tuberculosis meningitis

  • Pre‐existing non‐tuberculous disease likely to prejudice the response to, or assessment of, treatment (e.g. insulin‐dependent diabetes, liver or kidney disease, blood disorders, peripheral neuritis)

  • Female and known to be pregnant or breastfeeding

  • Condition likely to lead to uncooperative behaviour such as psychiatric illness or alcoholism

  • Contraindications to any medications in the study regimens

  • History of prolonged QTc syndrome or current or planned therapy with quinidine, procainamide, amiodarone, sotalol, disopyramide, ziprasidone, or terfenadine during the intensive phase of tuberculosis therapy

  • Haemoglobin < 7 g/L

  • AST or ALT > 5 times the upper range

  • Creatinine clearance < 30 mL/min

  • History of seizures

  • HIV positive with CD4 count less than 150/mm³

  • Weight < 35 kg

  • Already receiving anti‐retroviral therapy (ART)

Proportion with HIV seropositivity: 32% in the control regimen and 28% in the 4‐month regimen

Proportion with cavitation: 67% in the control regimen and 65% in the 4‐month regimen

Baseline drug resistance: excluded people resistant to isoniazid, rifampicin, or moxifloxacin

Interventions

Interventions:

4‐month (17‐week) ATT regimen: moxifloxacin replacing isoniazid throughout with twice‐weekly administration in continuation phase + rifapentine twice weekly replacing rifampicin in continuation phase: N = 275 randomized; 239 eligible, 165 completed (60% of those randomized; 69% of those eligible)

2 months (8 weeks) of ethambutol, moxifloxacin, rifampicin, and pyrazinamide administered daily, followed by

2 months (9 weeks) of moxifloxacin and rifapentine administered twice weekly

6‐month (26‐week) ATT regimen: moxifloxacin replacing isoniazid throughout with weekly rifapentine + weekly moxifloxacin in the 4‐month continuation phase

2 months (8 weeks) of ethambutol, moxifloxacin, rifampicin, and pyrazinamide administered daily, followed by

4 months (18 weeks) of rifapentine and moxifloxacin once a week

Control: 6‐month (26‐week) ATT regimen: N = 275 randomized; 240 eligible, 163 completed (59% of those randomized; 68% of those eligible)

2 months (8 weeks) of isoniazid, rifampicin, ethambutol, and pyrazinamide administered daily, followed by 18 weeks of isoniazid and rifampicin daily

Dosage: moxifloxacin 400 mg; rifapentine 900 mg in the 4‐month treatment arm (and 1200 mg in the 6‐month arm). All doses given were based on the weight of the patient

Outcomes

Outcomes reported and used in this review:

  • Relapse after treatment

  • Death from any cause

  • Failure to complete treatment

  • Treatment failure

  • Acquired drug resistance

  • Serious adverse events

Outcomes sought for this review and not reported:

  • Sputum positive smear/culture at 8 weeks (disaggregated data from the 2 moxifloxacin arms not reported)

Outcomes reported and not used in this review:

  • Culture results at end of follow‐up

  • ART start times in HIV‐infected people

  • Adherence

Notes

Funding: European and Developing Countries Clinical Trials Partnership, Wellcome Trust. Some of the trial medications were donated by Sanofi, Genus Pharmaceuticals, and Sandoz

Follow‐up method

Patients were followed‐up monthly up to 12 months after randomization and thereafter once in 3 months until 18 months. Two sputum samples were collected before treatment initiation for smear and culture, and 1 sample was collected monthly for 12 months and then again at 15 months and 18 months of follow‐up

Treatment supervision: treatment was directly observed in all participants in the intensive treatment phase. Drugs were taken under the supervision of a relative or another designated person in the 18‐week continuation phase in the control arm. Moxifloxacin and rifapentine treatment was supervised at the treatment facility twice weekly for the 9‐week continuation phase

Trial registration ID: ISRCTN44153044 (prospectively registered)

Acronym: RIFAQUIN

Comment: data from the 6‐month moxifloxacin intervention arm were not used in this study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from report: "A randomized allocation sequence was generated for each study centre with the use of blocks of varying size by an independent statistician based at the MRC CTU"

Allocation concealment (selection bias)

Low risk

Quote from updated protocol: "Sealed opaque envelopes containing the treatment allocation slips will be held by the pharmacist. When a patient is found to be eligible their details will be entered on the enrolment log by the designated member of the clinic team against the next available study number. These patient details and the study number will be entered on to the patient’s prescription. This will be taken to the pharmacy and the patient details entered onto the pharmacy register by the pharmacist against the next study number which will act as a check that the correct (next available) study number had been used. The pharmacist will then take the envelope corresponding to the study number and reveal the treatment allocation which will be written on the allocation slip. This will then be attached to the prescription and kept in the patient’s Trial folder or other appropriate place and the designated member of the clinic team made aware of the treatment allocation"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

This was an open‐label trial. The treating team was aware of allocated treatments. However, this does not seem to have influenced drug administration or use of co‐interventions

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote from report: "Apart from the statisticians reporting to the data and safety monitoring committee, the staff at St. George’s and at the MRC CTU were unaware of treatment assignment except when a lack of awareness would have been unethical (e.g., in some discussions of serious adverse events). Participating laboratories were unaware of treatment assignment throughout the study"

Comment: although treatment allocation before the start of the trial was concealed, the clinical team evaluating participants for efficacy and safety outcomes was aware of treatment allocation. However, laboratory assessments were objective and clinical outcomes were mostly based on objective assessments

Incomplete outcome data (attrition bias): At the end of ATT (Treatment failure, positive sputum culture, treatment discontinuation, adverse events)

Low risk

Although overall attrition was over 30%, there was no differential attrition in the 4‐month arm (31%) versus the control arm (32%). In the sensitivity analysis in the supplementary table, S1 attrition was 13% in each arm. We do not think this is likely to alter the estimates of relative effects

Incomplete outcome data (attrition bias): At the end of follow‐up (Relapse, deaths)

Low risk

Modifed intention‐to‐treat and per‐protocol analyses presented in Table 2 of the main report and in the sensitivity analyses in Table S1 in the online supplementary appendix do not indicate that bias due to differential attrition is likely to have affected the estimates of relative effects

Selective reporting (reporting bias)

Low risk

This trial was prospectively registered, and protocol amendments and reporting of results do not indicate selective reporting

Other bias

Low risk

Quote from report: "Some of the trial medications were donated by Sanofi, Genus Pharmaceuticals, and Sandoz, and a representative of Sanofi was a non‐voting observer at meetings of the steering committee, but none of these companies had any role in the study design, data accrual, data analysis, or manuscript preparation"

Merle 2014

Methods

Study design: randomized, multi‐centre, open‐label, parallel‐group, active‐controlled, non‐inferiority trial

Study period: June 2005 to April 2011

Recruitment sites: Conakry, Cotonou, Dakar, Durban, Nairobi

Countries where the trial was undertaken: Benin, Guinea, Kenya, Senegal, South Africa

Length of follow‐up: 24 months

Participants

No of participants randomized: 1836

Intervention: 917

Control: 919

Age: mean age intervention 30.9 years, control 30.6 years

Gender: male: Intervention 73%, control 72%

Inclusion criteria:

  • Aged 18 to 65 years (both inclusive) and weighing between 38 kg and 80 kg

  • Recently diagnosed, microscopically proven, pulmonary tuberculosis, defined as 2 consecutively positive sputum smears, of which 1 must be equal to or exceed grade 1

  • Findings in medical history and physical examination not exceeding grade 2 according to the Division of Microbiology and Infectious Disease grading system tables (DMID)

  • Voluntarily signed informed consent to participate in the study

  • Females of childbearing potential must have a confirmed negative pregnancy test at the screening visit and must employ an effective and acceptable method of birth control during treatment

  • Laboratory values that do not exceed grade 2 using the Division of Microbiology and Infectious Disease grading system (DMID) other than for glycaemia, haemoglobin, and potassium levels

Exclusion criteria:

  • Patients with a history of tuberculosis treatment within the last 3 years

  • Concomitant infection requiring additional anti‐infective treatment (especially antiretroviral medication ‐ ARV)

  • HIV‐infected patients with WHO stage 3 infection (except those presenting with only the "loss of weight > 10% body weight" criterion) and all patients at WHO stage 4 (see Appendix 5)

  • History of diabetes mellitus (DM) or non‐insulin‐dependent diabetes mellitus (NIDDM) requiring treatment or diet. Additionally, patients who have a fasting glucose level less than 70 mg/dL (3.9 mmol/L) or above 115 mg/dL (6.4 mmol/L) at screening will be excluded

  • Recreational drug abuse and alcohol abuse that, in the opinion of the investigator, could prejudice the conduct of the study in that patient

  • History of drug hypersensitivity and/or active allergic disease

  • Impaired renal, hepatic, or gastric function that may, in the opinion of the investigator, interfere with drug absorption, distribution, metabolism, or elimination

  • Any other findings in medical history and physical examination exceeding grade 2 in the DMID grading system tables

  • Patient using the following therapies:

    • Other antibiotics with known anti‐tuberculosis activity (i.e. ofloxacin, moxifloxacin, kanamycin)

    • Drugs known to prolong the QT interval (i.e. anti‐arrhythmics, psychotropics (phenothiazines, tricyclics, tetracyclics), erythromycin, pentamidine, and halofantrine)

    • Drugs known to give photosensitivity reactions

    • Receiving oral corticosteroids for longer than 2 weeks immediately before inclusion

    • Use of antacids containing aluminium or magnesium salts or sucralfate

    • Digoxin

    • Drugs that are eliminated via tubular secretion (e.g. probenecid, cimetidine, ranitidine)

  • Pregnant or lactating women

  • Patients with congenital QT interval prolongation > 480 ms

  • Patients with clinically significant bradycardia (40 beats/min)

  • Baseline laboratory values exceeding grade 2 using the Division of Microbiology and Infectious Disease grading system (DMID) except glycaemia value as previously stated, haemoglobin, and hypokalaemia for which the limit values are as follows: potassium < 3.0 mEq/L (> grade 1), haemoglobin < 6.5 g/dL

  • Any other finding considered by the investigator as compromising the participation of the patient in the trial

  • Any condition rendering the patient unable to understand the nature, scope, and possible consequences of the study and to provide consent

  • Participation in another drug trial within the 3 months before the screening visit

Proportion with HIV seropositivity: 19% in the control regimen and 18% in the 4‐month regimen

Proportion with cavitation: 50% in the control regimen and 52% in the 4‐month regimen

Baseline drug resistance: excluded people with rifampicin resistance and MDR‐TB; isoniazid resistance: (gatifloxacin 8.5%; control 6.6%)

Interventions

Intervention: 4‐month ATT regimen

Gatifloxacin: gatifloxacin replacing ethambutol in intensive and continuation phases: N = 917 randomized; 791 eligible, 651 included in per‐protocol analysis (71% of those randomized, 82% of those eligible)

2 months of gatifloxacin, isoniazid, rifampicin, and pyrazinamide given daily, followed by

2 months of gatifloxacin, isoniazid, and rifampicin

Control: 6‐month ATT regimen (N = 919 randomized; 784 eligible, 601 included in per‐protocol analysis (65% of those randomized; 77% of those eligible)

2 months of ethambutol, isoniazid, rifampicin, and pyrazinamide, followed by

4 months of isoniazid and rifampicin

Dosage:

Fixed‐dose combination tablets of isoniazid–rifampin, isoniazid–rifampin–pyrazinamide, or isoniazid–rifampin–pyrazinamide–ethambutol were used wherever needed. Gatifloxacin was given at a dose of 400 mg. Other drugs were given in weight‐based doses (< 50 kg, ≥ 50 kg)

Outcomes

Outcomes reported and used in this review:

  • Recurrence (relapse or reinfection)

  • Death from any cause

  • Treatment failure (at 4 months or 6 months)

  • Sputum positive smear/culture at 8 weeks

  • Treatment discontinuation

  • Serious adverse events

  • Other adverse events (hyperglycaemia during treatment phase, prolongation of QT interval)

Outcomes sought for this review but not reported:

  • Acquired drug resistance

Outcomes reported and not used in this review:

  • Unfavourable outcome by 24 months after treatment (composite of treatment failure, recurrence, death, or withdrawal)

  • Unfavourable outcome at 18 months after randomization

  • Time to an unfavourable outcome after treatment

  • Pharmacokinetic/pharmacodynamic data (published separately)

Notes

Funding: Institut de Recherche pour le Développement (IRD) (on behalf of the OFLOTUB Consortium), World Health Organization (WHO). Lupin Pharmaceuticals provided study medicines

Follow‐up method: trial drugs were administered orally under supervision 6 days a week during the intensive phase and were provided every 2 weeks in the continuation phase. Two sputum samples were obtained for smear examination, solid culture, and drug sensitivity tests at baseline and at all subsequent visits. Electrocardiograms (ECGs) were done at baseline, between 1 and 5 hours after drug intake, at 4 weeks, at 8 weeks, and at end of treatment

Treatment supervision: trial drugs were given orally, 6 days a week under direct observation supervision during intensive phase and were provided every 2 weeks in the continuation phase to a supervisor who ensured treatment was taken. Adherence was assessed by pill count that remained in the weekly treatment boxes

Trial Registration ID: NCT00216385 (retrospectively registered: September 2005)

Acronym: OFLOTUB/gatifloxacin

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from report: "Patients were randomly assigned, in a 1:1 ratio with stratification according to country, to either a gatifloxacin‐containing regimen (experimental group) or the 6‐month standard treatment (control group)"

Quote from protocol: "Randomization lists, stratified by study site and indicating a randomization number and which treatment is to be given, will be produced prior to the start of the trial by the medical statistician in London"

Allocation concealment (selection bias)

Low risk

Quote from protocol:"The Code for each individual will be provided in separate sealed envelopes and assigned to individuals in the order in which they are enrolled in the study"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote from protocol: "It must be noted that management of patients cannot be blinded, because of the difference in treatment length, but steps will be taken to ensure equal management and follow‐up of both treatment arms"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote from protocol: "Lastly, when patients recruited in the trial come to the clinic with a suspicion of relapse, the treatment they received will be blinded to the physician examining them"

Quote from protocol: "Laboratory technicians will be blinded to the origin of each sample, ensuring unbiased assessment of endpoints"

Incomplete outcome data (attrition bias): At the end of ATT (Treatment failure, positive sputum culture, treatment discontinuation, adverse events)

Low risk

Althought 35% of those eligible after randomization in the control arm and 29% in the intervention arm were excluded from the per‐protocol analyses, modified intention‐to‐treat analyses included 86% of those randomized to each arm. Results of modified intention‐to treat analyses and per‐protocol analyses were consistent and did not suggest that differential attrition significantly biased the relative estimates of effects

Incomplete outcome data (attrition bias): At the end of follow‐up (Relapse, deaths)

Low risk

Quote from report: "The cumulative percentage of patients retained in the experimental and control groups, respectively, was 93.5% and 93.4% by 52 weeks, 91.0% and 89.6% by 78 weeks, and 87.5% and 82.7% by 94 weeks"

Comment: there was differential attrition over 24 months, but results of the per‐protocol and intention‐to‐treat analyses were consistent

Selective reporting (reporting bias)

Low risk

The trial was retrospectively registered. However all outcomes in the registration documents and changes in the protocol were documented and reported adequately, and did not indicate selective reporting

Other bias

Low risk

Quote from report: "Lupin Pharmaceuticals had no role in the conduct of the trial, the analysis of the data, or the preparation of the manuscript"

Velayutham 2014

Methods

Study design: randomized, open‐label, parallel‐group, 5‐arm, active‐controlled trial

Study period: patient recruitment commenced in May 2007 (interim results of an ongoing trial)

Recruitment sites: Chennai, Madurai

Country where the trial was undertaken: India

Length of follow‐up: 24 months after treatment completion

Participants

No. of participants randomized: 801 (of the 1650 anticipated)

Combined intervention arms: 3 to 4 months moxifloxacin: N = 629

Control: 6 months ATT: N = 172

Age: < 35 years: moxifloxacin 52%; control 52%

Gender: male: moxifloxacin 74%; control 77%

Inclusion criteria:

  • Adult patients, 18 years or older, with newly diagnosed sputum smear positive pulmonary tuberculosis

  • Resident within a designated study area and permitted home visits.

Exclusion criteria:

  • Those with previous treatment for tuberculosis exceeding 30 days, weighing over 30 kg, pregnant or lactating women

  • Those with concomitant diabetes mellitus, severe systemic hypertension, epilepsy, serious forms of extrapulmonary tuberculosis, or HIV infection

Proportion with HIV seropositivity: nil (excluded)

Proportion with cavitation: moxifloxacin 36%; control 41%

Baseline drug resistance: isoniazid (moxifloxacin 7%; control 8%); ofloxacin (moxifloxacin 5%; control 7%); rifampicin, ethambutol, isoniazid, and ethambutol; isoniazid and ofloxacin (< 1% in both groups)

Interventions

Interventions: 3‐ and 4‐month moxifloxacin regimens: moxifloxacin added to standard ATT drugs

N = 629 randomized; 13 exclusions; 616 (98%) evaluated of those randomized

  • Rifampicin, isoniazid, pyrazinamide, ethambutol, and moxifloxacin daily for 3 months

  • Rifampicin, isoniazid, pyrazinamide, ethambutol, and moxifloxacin daily for 2 months, followed by rifampicin, isoniazid, and moxifloxacin daily for 2 months

  • Rifampicin, isoniazid, pyrazinamide, ethambutol, and moxifloxacin daily for 2 months, followed by rifampicin, isoniazid, and moxifloxacin thrice weekly for 2 months

  • Rifampicin, isoniazid, pyrazinamide, ethambutol, and moxifloxacin daily for 2 months, followed by rifampicin, isoniazid, ethambutol, and moxifloxacin thrice weekly for 2 months

Control (6‐month regimen): N = 172 randomized: 8 exclusions; 164 (95%) evaluated of those randomized

  • Rifampicin, isoniazid, pyrazinamide, and ethambutol thrice weekly for 2 months, followed by rifampicin and isoniazid thrice weekly for 4 months

Dosage: rifampicin 450 (< 60 kg) or 600 mg (> 60 kg); isoniazid 300 mg (daily) and 600 mg (thrice weekly); pyrazinamide 1500 mg; ethambutol 800 mg (daily) and 1200 mg (thrice weekly); moxifloxacin 400 mg

Outcomes

Outcomes reported and used in this review:

  • Sputum culture conversion at 2 months of treatment (assessed at 5 months)

  • Adverse reactions while on anti‐tuberculosis drugs

Outcomes sought for the review but not reported:

  • Relapse rates 24 months after treatment among those with a favourable or doubtful bacteriologic response at end of treatment (primary outcome for the ongoing trial)

  • Death from any cause

  • Failure to complete treatment

  • Treatment failure (bacteriologic response at end of treatment is an outcome in the ongoing trial)

  • Acquired drug resistance

Notes

Funding: Indian Council of Medical Research

Follow‐up method: over the 24‐month follow‐up period, participants had monthly clinical examination and adherence and adverse events recording; monthly sputum microscopy and culture (and on days 15 and 45) and monthly drug susceptibility testing for 1 positive culture; monthly ECG, haemogram, liver and kidney functions, random blood sugars, and HIV ELISA tests. Chest X‐ray after the 2‐month intensive phase. Adverse events were assessed monthly for the duration of treatment (3 to 6 months)

Treatment supervision: during the daily phase, treatment was under direct observation on 5 of 7 days of the week, whereas 2 doses were self‐administered. All thrice‐weekly phase doses were directly observed. Patients who missed treatment visits were visited at home and were motivated to attend the clinic for treatment

Trial registration ID: CTRI 2008/091/000024 (retrospectively registered on 09/05/2008)

Comment: results for sputum conversion at 2 months presented are the combined results of the 4 moxifloxacin regimens. Results for adverse events include up to the end of the 3‐ or 4‐month moxifloxacin regimens and over 6 months in the control regimen

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from report: "Restricted random allocation sequences generated using random number tables,separately for the 6 strata, were used to assign the regimens"

Allocation concealment (selection bias)

Low risk

Quote from trial registration document: "Sequentially numbered, sealed, opaque envelopes"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

This was an open‐label study but all treatment arms had supervised treatment and scheduled assessments for efficacy and safety outcomes

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The outcomes reported are sputum culture results at 2 months and drug adverse events that were assessed for all participants at specified time points. Sputum culture results and ECG reports are objective outcomes, and the likelihood of detection bias influencing the reporting of other adverse events is low

Incomplete outcome data (attrition bias): At the end of ATT (Treatment failure, positive sputum culture, treatment discontinuation, adverse events)

Low risk

590 of 616 (96%) on moxifloxacin regimens and 151 of 162 (93%) on the control regimen had sputum cultures reported at 2 months. The 3% differential attrition is unlikely to have influenced the difference in proportions with negative sputum cultures at 2 months of 14.6% (95% CI 8.8% to 21.8%)

Incomplete outcome data (attrition bias): At the end of follow‐up (Relapse, deaths)

Low risk

Not reported, as the trial is ongoing and this report includes only interim outcomes

Selective reporting (reporting bias)

Low risk

This is an interim report of an ongoing trial. The outcomes presented were pre‐stated in the trial registration document

Other bias

Low risk

No other sources of bias were detected

Abbreviations: ALT: alanine aminotransferase; ART: antiretroviral therapy; ARV: antiretroviral; AST: aspartate aminotransferase; ATT: anti‐tuberculosis treatment; CrCl: creatinine clearance; DM: diabetes mellitus; DMID: Division of Microbiology and Infectious Disease; ECG: electrocardiogram; ELISA: enzyme‐linked immunosorbent assay; IUCD: intrauterine contraceptive device; M tuberculosis: Mycobacterium tuberculosis; MDR‐TB: multi‐drug‐resistant tuberculosis; NIDDM: non‐insulin‐dependent diabetes mellitus; WHO: World Health Organization.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Alavi 2009

RCT comparing 3 months of rifampicin + isoniazid + ofloxacin versus a standard 6‐month regimen in people diagnosed with smear negative pulmonary tuberculosis

Diagnosis of pulmonary tuberculosis was not confirmed by culture or GeneXpert

Burman 2006

Factorial RCT comparing 5 days a week versus 3 days a week treatment with moxifloxacin substituting ethambutol in the intensive phase of treatment with isoniazid, rifampicin, and pyrazinamide

Not designed to compare treatments less than 6 months versus standard 6‐month regimen (phase 2b trial)

Conde 2009

RCT comparing moxifloxacin versus ethambutol in the intensive phase of treatment with rifampicin, isoniazid, and pyrazinamide

Not designed to compare treatments less than 6 months versus standard 6‐month regimen (phase 2b trial)

Conde 2016

RCT comparing rifapentine plus moxifloxacin or rifampin plus ethambutol daily for 8 weeks, along with isoniazid and pyrazinamide

Not designed to compare treatments less than 6 months versus standard 6‐month regimen (phase 2b trial)

Dorman 2009

RCT comparing moxifloxacin versus isoniazid in the intensive phase of treatment with rifampicin, ethambutol, and pyrazinamide

Not designed to compare treatments less than 6 months versus standard 6‐month regimen (phase 2b trial)

El‐Sadr 1998

RCT comparing levofloxacin added for the first 2 months to the standard 6‐month ATT regimen versus 6 to 9 months of standard ATT regimen

No comparison with a regimen shorter than 6 months

Johnson 2009

RCT in adults with newly diagnosed, sputum‐ or culture‐confirmed, non‐cavitary pulmonary tuberculosis who were culture negative after 4 months of daily treatment with 2HRZE + 2HR to stop treatment (4‐month treatment arm) or continue HR for 2 months

Participants had already taken 4 months of ATT before randomization. Only those who were sputum‐negative were randomized, After randomization, participants received either no treatment or only 2 more months of HR

Kohno 1992

Contolled trial comparing ofloxacin, rifampicin, and isoniazid with the regimen of ethambutol, rifampicin, and isoniazid given daily for 9 months

No control arm with 2HRZE + 4HR (or 4HRE) or treatment arm with shorter regimens

Rustomjee 2008

RCT (phase 2) comparing three 6‐month regimens with gatifloxacin, moxifloxacin, or ofloxacin given along with rifampicin, isoniazid, and pyrazinamide for the first 2 months followed by 4 months of isoniazid and rifampicin versus standard 2HRZE + 4HR regimens

No arm with shorter regimens

Tuberculosis Research Centre 1986

Controlled trial comparing 3 and 5 months of streptomycin, isoniazid, and pyrazinamide, with or without rifampicin

No control arm with 2HRZE + 4HR (or 4HRE)

Tuberculosis Research Centre 2002

RCT comparing 4 different regimens of ofloxacin, isoniazid, rifampicin, and pyrazinamide

No control arm with 2HRZE + 4HR (or 4HRE)

Abbreviations: ATT: anti‐tuberculosis treatment; E: ethambutol; H: isoniazid; R: rifampicin; RCT: randomized controlled trial; Z: pyrazinamide.

Characteristics of ongoing studies [ordered by study ID]

CTRI/2008/091/000024

Trial name or title

Randomized clinical trial to study the efficacy and tolerability of 3‐ and 4‐month regimens containing moxifloxacin in the treatment of patients with sputum smear and culture positive pulmonary tuberculosis

Methods

Randomized, open‐label, parallel‐group, 5‐arm, active‐controlled trial

Participants

Inclusion criteria:

  • Age 18 years to 60 years

  • Residing in or around Chennai or Madurai

  • No previous anti‐tuberculosis treatment

  • At least 2 sputum smears should be positive for tubercle bacilli by fluorescent microscopy at enrolment

  • Willing to attend the treatment centre for supervised treatment

  • Willing for home visits by staff from the centre

  • Willing to give written informed consent

Exclusion criteria:

  • Body weight less than 30 kg

  • Hepatic or renal disease as evidenced by clinical or biochemical abnormalities

  • Diabetes mellitus

  • History of seizure or loss of consciousness

  • Psychiatric illness

  • Abnormal electrocardiogram or anti‐arrhythmic medication

  • Those in a moribund state

  • Seropositive for HIV antibodies

  • Pregnancy or lactation

  • Visual disorders other than refractory error

Anticipated sample size: 1650

Interventions

Intervention(s):

Moxifloxacin arm: 4 regimens of 3 to 4 months

3 RHZEM

2 RHZEM/2 RHM

2 RHZEM/2 RHM thrice weekly

2 RHZEM/2 RHEM thrice weekly

Dose: rifampicin 450 mg; isoniazid 300 mg (daily), 600 mg (thrice weekly); pyrazinamide 1500 mg; ethambutol 800 mg (daily), 1200 mg (thrice weekly); moxifloxacin 400 mg

Control:

2 RHZE thrice weekly/4 RH thrice weekly (for 6 months)

Dose: rifampicin 450 mg; isoniazid 600 mg; pyrazinamide 1500 mg; ethambutol 1200 mg

Outcomes

Primary outcome:

  • Relapse rates 24 months after treatment among those with a favourable or doubtful bacteriologic response at end of treatment

Secondary outcomes:

  • Sputum culture conversion at 2 months of treatment

  • Bacteriologic response at end of treatment

  • Adverse reactions to anti‐tuberculosis drugs during treatment

Starting date

30 May 2007; anticipated study end date: May 2015; no results posted (last modified 06/02/2013)

Contact information

Dr MS Jawahar, Tuberculosis Research Centre, Mayor VR Ramanathan Road, Chetput, Chennai TAMIL NADU 600031 India. Tel: +91‐44‐28369500; Email: [email protected]

Notes

Study locations: Chennai and Madurai in India

Registration number:CTRI/2008/091/000024

Primary sponsor: Indican Council of Medical Research

Comment: the first author confirmed that this is completed, has been analysed, written up, submitted for publication, and will be re‐submitted after peer review

ISRCTN63579542

Trial name or title

Shorter treatment for minimal tuberculosis in children (SHINE study)

Methods

Parallel‐group, randomized, non‐inferiority, open‐label, 2‐arm, phase 3 clinical trial

Participants

Inclusion criteria:

  • Age 0 to 16 years

  • Weight > 4 kg

  • Clinician has decided to treat with standard first‐line regimen

  • Asymptomatic or symptomatic but with non‐severe tuberculosis including not previously treated for tuberculosis or successfully treated for tuberculosis over 2 years since last completed treatment

  • Known HIV status: HIV infected or HIV uninfected

  • Willing and likely to adhere to 72 weeks of follow‐up

  • Informed written consent from parent/legal caregiver

  • Home address accessible for visiting and intending to remain within recruitment area for follow‐up

Exclusion criteria:

  • Smear positive respiratory sample tuberculosis

  • Premature (< 37 weeks) and aged under 3 months

  • Miliary tuberculosis, spinal tuberculosis, tuberculosis meningitis, osteoarticular tuberculosis, abdominal tuberculosis, congenital tuberculosis

  • Pre‐existing liver or kidney disease, peripheral neuropathy, cavitation

  • Any known contraindication to taking anti‐tuberculosis drugs

  • Known contact with MDR, pre‐XDR, or XDR adult source case

  • Proven anti‐tuberculosis drug resistance in the child

  • Severely sick

  • Pregnancy

Anticipated sample size: 1200

Interventions

Intervention: 4‐month standard ATT regimen

8 weeks intensive Isoniazid (H), rifampicin (R), pyrazinamide (Z) with or without ethambutol (E) according to local practice, HRZ(E), followed by

8 weeks of continuation HR

Control: 6‐month standard ATT regimen

8 weeks intensive HRZ(E), followed by

6 weeks of continuation HR

Outcomes

Primary outcome measures:

• Efficacy: unfavourable outcome, defined by the composite endpoint of tuberculosis treatment failure, relapse (or re‐infection), or death

• Safety: grade 3/4 adverse events

Secondary outcome measures:

• Mortality

• Adverse drug reactions up to 30 days of completing treatment

• Unfavourable outcome in those with definite tuberculosis

• Suppressed HIV viral load at 24 and 48 weeks in HIV‐infected children starting ART, measured centrally on stored samples

• Adherence and acceptability

• Bacterial infection

Anciliary studies will evaluate pharmacokinetics; cost/cost‐effectiveness implications of treatment shortening, and a nested qualitative substudy will investigate the ways in which health workers manage implementation of dose and weight band recommendations, particularly in children taking anti‐tuberculosis drugs and ARVs

Starting date

April 2015; anticipated end date: April 2019 (no longer recruiting)

Contact information

SHINE Trial Management Team

MRC Clinical Trials Unit at UCL

Institute of Clinical Trials and Methodology

Aviation House, 125 Kingsway, London WC2B 6NH, UK

Ph: +44 (0) 20 7670 4700

Email: [email protected]

Notes

Study locations: South Africa, India, Uganda, Zambia

Registration number:ISRCTN63579542

Primary sponsors: University College London, Joint Global Health Trials Scheme: Department for International Development, the Wellcome Trust, the Medical Research Council, and Svizera Ltd

NCT02342886

Trial name or title

Shortening treatment by advancing novel drugs (STAND)

Methods

Phase 3 open‐label, partially randomized, controlled clinical trial

Participants

Inclusion criteria:

  • Signed written consent or witnessed oral consent in the case of illiteracy, before undertaking any trial‐related procedures

  • Male or female, aged 18 years or older

  • Body weight (in light clothing and no shoes) ≥ 30 kg

  • Sputum positive for tubercle bacilli (at least 1+ on the International Union Against Tuberculosis and Lung Disease (IUATLD) and World Health Organization (WHO) scales on smear microscopy at the trial laboratory)

  • Drug‐sensitive tuberculosis treatment arm participants should be:sensitive to rifampicin by rapid sputum‐based test (may be sensitive or resistant to isoniazid) AND either newly diagnosed with TB or with patient history of being untreated for at least 3 years after cure from a previous episode of TB. If they are entered into the trial because they are sensitive to rifampicin by rapid sputum‐based test, however, if receipt of rifampicin resistance testing via an indirect susceptibility test in liquid culture shows they are rifampicin resistant, they will be. excluded as late exclusions; possibly replaced as determined by the sponsor

  • MDR‐TB treatment arm participants should be resistant to rifampicin by rapid sputum‐based test (may be sensitive or resistant to isoniazid)

  • Chest X‐ray that in the opinion of the investigator is compatible with pulmonary TB

  • Non‐childbearing potential or using effective methods of birth control, as defined below

Non‐childbearing potential:

  • Participant not heterosexually active or practicing sexual abstinence; or

  • Female participant or male participant with female sexual partner ‐ bilateral oophorectomy, bilateral tubal ligation, and/or hysterectomy; or postmenopausal with a history of no menses for at least 12 consecutive months; or

  • Male participant or female participant with male sexual partner ‐ vasectomized or with bilateral orchidectomy minimally 3 months before screening

Effective birth control methods:

  • Double‐barrier method, which can include male condom, diaphragm, cervical cap, or female condom; or

  • Female participant: barrier method combined with hormone‐based contraceptives or an intrauterine device for the female participant

  • Male participant's female sexual partner: double‐barrier method or hormone‐based contraceptives or an intrauterine device for the female participant

Willing to continue practising birth control methods and not planning to conceive throughout treatment and for 12 weeks (male participants) or 1 week (female participants) after last dose of trial medication or discontinuation from trial medication in case of premature discontinuation

Exclusion criteria:

  • Any non‐TB‐related condition (including myasthenia gravis) where participation in the trial, as judged by the investigator, could compromise the well‐being of the participant or could prevent, limit, or confound protocol‐specified assessments

  • Being or about to be treated for malaria

  • Critically ill and, in the judgement of the investigator, with a diagnosis likely to result in death during the trial or the follow‐up period

  • TB meningitis or other forms of extrapulmonary tuberculosis with high risk of a poor outcome, or likely to require a longer course of therapy (such as TB of the bone or joint), as judged by the investigator

  • History of allergy or hypersensitivity to any of the trial IMP or related substances, including known allergy to any fluoroquinolone antibiotic, history of tendinopathy associated with quinolones, or suspected hypersensitivity to any rifampicin antibiotics

  • For HIV‐infected participants, any of the following: CD4+ count < 100 cells/µL; Karnofsky score < 60%; received intravenous antifungal medication within the last 90 days; WHO clinical stage 4 HIV disease

  • Resistant to fluoroquinolones (rapid, sputum‐based molecular screening tests). If they are entered into the trial because they are sensitive to fluoroquinolones by rapid sputum‐based test, but on receipt of the fluoroquinolone resistance test via an indirect susceptibility test in liquid culture, they show they are fluoroquinolone resistant, they will be excluded as late exclusions; possibly replaced as determined by the sponsor

  • Resistant to pyrazinamide (rapid, sputum‐based molecular screening tests). Drug‐sensitive TB treatment arm participants may be entered before receipt of the rapid, sputum‐based molecular pyrazinamide resistance screening test result. On receipt of the result, if resistant, they will be excluded as late exclusions; possibly replaced as determined by the sponsor. MDR‐TB treatment arm participants may not be entered before receipt of the rapid, sputum‐based molecular pyrazinamide resistance screening test result showing they are sensitive to pyrazinamide

  • Having participated in other clinical trials with investigational agents within 8 weeks before trial start or currently enrolled in an investigational trial

  • With any of the following at screening (per measurements and reading done by central electrocardiogram (ECG) where applicable): cardiac arrhythmia requiring medication; prolongation of QT/QTc interval with QTcF (Fridericia correction) > 450 ms; history of additional risk factors for torsade de pointes (e.g. heart failure, hypokalaemia, family history of long QT syndrome); any clinically significant ECG abnormality, in the opinion of the investigator

  • Unstable diabetes mellitus that required hospitalization for hyperglycaemia or hypoglycaemia within the past year before the start of screening. Specific treatments:

    • Previous treatment with PA‐824 as part of a clinical trial

    • For DS‐TB treatment arms: previous treatment for tuberculosis within 3 years before Day (‐9 to ‐1) (screening). Participants who have previously received isoniazid prophylactically may be included in the trial as long as that treatment is/was discontinued at least 7 days before randomization into this trial. For MDR‐TB participants: previous treatment for MDR‐TB, although may have been on MDR‐TB treatment regimen for no longer than 7 days at the start of screening. Previous treatment for TB includes, but is not limited to, gatifloxacin, amikacin, cycloserine, rifabutin, kanamycin, para‐aminosalicylic acid, rifapentine, thiacetazone, capreomycin, quinolones, thioamides, and metronidazole

    • Any disease or condition for which use of standard TB drugs or any of their components is contraindicated, including but not limited to allergy to any TB drugs, their components, or the IMP

    • Use of any drug within 30 days before randomization known to prolong QTc interval (including, but not limited to, amiodarone, amitriptyline, bepridil, chloroquine, chlorpromazine, cisapride, clarithromycin, disopyramide dofetilide, domperidone, droperidol, erythromycin, halofantrine, haloperidol, ibutilide, levomethadyl, mesoridazine, methadone, pentamidine, pimozide, procainamide, quinacrine, quinidine, sotalol, sparfloxacin, thioridazine)

    • Use of systemic glucocorticoids within 1 year of start of screening (inhaled or intranasal glucocorticoids are allowed)

    • Participants recently started or expected to need to start antiretroviral therapy (ART) within 1 month after randomization. Patients may be included who have been on ARTs for longer than 30 days before the start of screening, or who are expected to start ART more than 30 days after randomization. Laboratory abnormalities

  • Participants with the following toxicities at screening as defined by the enhanced Division of Microbiology and Infectious Disease (DMID) adult toxicity table (November 2007), where applicable:

    • Creatinine grade 2 or greater (> 1.5 times upper limit of normal (ULN))

    • Creatinine clearance (CrCl) level less than 30 mL/min according to the Cockcroft‐Gault formula

    • Haemoglobin grade 4 (< 6.5 g/dL); platelets grade 3 or greater (under 50 x 10⁹ cells/L 50,000/mm³)

    • Serum potassium less than lower limit of normal for the laboratory; this may be repeated once

    • Aspartate aminotransferase (AST) grade 3 or greater (≥ 3.0 x ULN)

    • Alanine aminotransferase (ALT) grade 3 or greater (≥ 3.0 x ULN)

    • Alkaline phosphatase (ALP): grade 4 (> 8.0 x ULN) to be excluded; grade 3 (≥ 3.0 to 8.0 x ULN) must be discussed with and approved by the sponsor medical monitor

    • Total bilirubin: 2.0 x ULN, when other liver functions are in the normal range; 1.50 x ULN when accompanied by any increase in other liver function tests among participants with total bilirubin > 1.25 x ULN and accompanied by any increase in other liver function tests must be discussed with the sponsor medical monitor before enrolment

Recruited sample size = 1500

Interventions

Interventions:

  • Moxifloxacin 400 mg + PA‐824 200 mg + pyrazinamide 1500 mg orally once daily for 26 weeks

  • Moxifloxacin 400 mg + PA‐824 200 mg + pyrazinamide 1500 mg orally once a day for 17 weeks

  • Moxifloxacin 400 mg + PA‐824 100 mg + pyrazinamide 1500 mg orally once daily for 17 weeks

Control:

  • 2HRZE/4HR (26 weeks)

(Additional MDR‐TB arm: moxifloxacin 400 mg + PA‐824 200 mg + pyrazinamide 1500 mg for 26 weeks)

Outcomes

Primary outcome:

  • Incidence of combined bacteriologic failure or relapse of clinical failure at 12 months from start of therapy

    • Bacteriologic failure: during the treatment period, failure to attain culture conversion to negative status in liquid culture

    • Bacteriologic relapse: during the follow‐up period, failure to maintain culture conversion to negative status in liquid culture, with culture conversion to positive status with a Mycobacterium tuberculosis (MTB) strain that is genetically identical to the infecting strain at baseline

    • Bacteriologic reinfection: during the follow‐up period, failure to maintain culture conversion to negative status in liquid culture, with culture conversion to positive status with MTB strain that is genetically different from the infecting strain at baseline

    • Clinical failure: change from protocol‐specified TB treatment due to treatment failure, re‐treatment for TB during follow‐up, or TB‐related death

Secondary outcomes:

  • Incidence of bacteriologic failure or relapse or clinical failure at 24 months from the start of therapy as a confirmatory analysis

  • Rate of change in time to culture positivity (TTP) over time in liquid culture MGIT in sputum, represented by the model‐fitted log(TTP) results as calculated by the regression of the observed log(TTP) results over time. [Screening; Day 1, 7; Week 2 to 7; Month 2 to 6, 9, 12, 15, 18, 24] MGIT is defined as mycobacterial growth indicator tube

  • Time in days to sputum culture conversion to negative status in liquid culture (MGIT) through the treatment period to be explored as a potential biomarker of definitive outcome. [Screening; Day 1, 7; Week 2 to 7; Month 2 to 6, 9, 12, 15, 18, 24]

  • Proportion of participants with sputum culture conversion to negative status in liquid culture (MGIT) at 4, 8, 12, and 17 weeks to be explored as a potential biomarker of definitive outcome. [Week 4, 8, 12, and 17]

  • Incidence of treatment‐emergent adverse events (TEAEs) presented by incidence and seriousness, leading to TB‐related or non‐TB‐related death. [Day 1, 7; Week 2 to 7; Month 2 to 6, 9, 12, 15, 18, 24]

  • Clinical laboratory safety measurements of haematology and chemistry, including observed and change from baseline. [Screening; Day 1; Week 1, 2, 4; Month 2, 3, 4, 6]

  • Trough plasma concentrations will be used to evaluate effects of baseline subject covariates on trial drug pharmacokinetics and associated bacteriologic endpoints. [Week 2, Month 2]

  • Minimum inhibitory concentration (MIC) against moxifloxacin and PA‐824 [Day 1; Week 17 or Week 26 ]. MIC: lowest concentration of moxifloxacin or PA‐824 that will inhibit visible growth in culture

  • Change from baseline in sperm concentration by group. [Screening; Day 1; Week 12, 13, 26, 27, 39, 40]

  • Change from baseline in male FSH by group. [Screening; Day 1; Week 12, 13, 26, 27, 39, 40] Reproductive hormones: FSH, LH, testosterone, inhibin B

  • Change from baseline in male LH by group. [Screening; Day 1; Week 12, 13, 26, 27, 39, 40]

  • Change from baseline in male testosterone by group. [Screening; Day 1; Week 12, 13, 26, 27, 39, 40]

  • Change from baseline in male inhibin B by group. [Screening; Day 1; Week 12, 13, 26, 27, 39, 40]

  • Change from baseline in proportion of total motile sperm by group. [Screening; Day 1; Week 12, 13, 26, 27, 39, 40]

  • Change from baseline in sperm morphology by group. [Screening; Day 1; Week 12, 13, 26, 27, 39, 40]

  • Change from baseline in sperm volume by group. [Screening; Day 1; Week 12, 13, 26, 27, 39, 40]

  • Change from baseline in total sperm numbers by group. [Screening; Day 1; Week 12, 13, 26, 27, 39, 40]

Starting date

February 2015; completed May 2018 (no results posted)

Contact information

Stephen H Gillespie, MD

School of Medicine, University of St. Andrews, North Haugh, St. Andrews KY16 9TF, United Kingdom

Email: shg3@st‐andrews.ac.uk

Notes

Study locations: Georgia, Kenya, Malaysia, Philippines, South Africa, Tanzania, Uganda, Zambia

Registration number:NCT02342886

Sponsors: Global Alliance for TB Drug Development

NCT02410772

Trial name or title

Rifapentine‐containing tuberculosis treatment shortening regimens

Methods

Randomized, open‐label, parallel‐assignment, controlled phase 3 clinical trial

Participants

Inclusion criteria:

  • Suspected pulmonary tuberculosis plus 1 or both of the following: (a) at least 1 sputum specimen positive for acid‐fast bacilli on smear microscopy OR (b) at least 1 sputum specimen positive for M tuberculosis by Xpert MTB/RIF testing, with semi‐quantitative result of 'medium' or 'high' and rifamycin resistance not detected

  • Age 12 years or older

  • Verifiable address or residence location that is readily accessible for visiting, and willingness to inform the study team of any change in address during treatment and follow‐up period

  • Women of childbearing potential who are not surgically sterilized must agree to practice a barrier method of contraception or must abstain from heterosexual intercourse during study drug treatment

  • Documentation of HIV infection status

  • For HIV‐positive individuals, CD4 T‐cell count greater than or equal to 100 cells/mm³ based on testing performed at or within 30 days before screening

  • Laboratory parameters done at or within 14 days before screening:

    • Serum or plasma alanine aminotransferase (ALT) less than or equal to 3 times upper limit of normal

    • Serum or plasma total bilirubin less than or equal to 2.5 times upper limit of normal

    • Serum or plasma creatinine level less than or equal to 2 times upper limit of normal

    • Serum or plasma potassium level greater than or equal to 3.5 meq/L

    • Hemoglobin level 7.0 g/dL or greater

    • Platelet count 100,000/mm³ or greater

  • For women of childbearing potential, a negative pregnancy test at or within seven (7) days before screening:

    • Karnofsky score greater than or equal to 60

    • Written informed consent

Exclusion criteria:

  • Pregnant or breastfeeding

  • Unable to take oral medications

  • Previously enrolled in this study

  • Received any investigational drug in the past 3 months

  • More than five (5) days of treatment directed against active tuberculosis within 6 months preceding initiation of study drugs

  • More than five (5) days of systemic treatment with any 1 or more of the following drugs within 30 days preceding initiation of study drugs: isoniazid, rifampin, rifabutin, rifapentine, ethambutol, pyrazinamide, kanamycin, amikacin, streptomycin, capreomycin, moxifloxacin, levofloxacin, gatifloxacin, ofloxacin, ciprofloxacin, other fluoroquinolones, ethionamide, prothionamide, cycloserine, terizidone, para‐aminosalicylic acid, linezolid, clofazimine, delamanid, or bedaquiline

  • Known history of prolonged QT syndrome

  • Suspected or documented tuberculosis involving central nervous system and/or bones and/or joints, and/or miliary tuberculosis, and/or pericardial tuberculosis

  • Current or planned use within 6 months following enrolment of 1 or more of the following medications: HIV protease inhibitors, HIV integrase inhibitors, HIV entry and fusion inhibitors, HIV non‐nucleoside reverse transcriptase inhibitors other than efavirenz, quinidine, procainamide, amiodarone, sotalol, disopyramide, ziprasidone, or terfenadine. Individuals who are currently taking efavirenz‐based antiretroviral treatment (ART) or for whom initiation of efavirenz‐based ART is planned within 17 weeks following enrolment may participate

  • Weight less than 40.0 kg

  • Known allergy or intolerance to any of the study medications

  • Individuals will be excluded from enrolment if, at the time of enrolment, their M tuberculosis isolate is already known to be resistant to any 1 or more of the following: rifampin, isoniazid, pyrazinamide, ethambutol, or fluoroquinolones

  • Other medical conditions that, in the investigator's judgment, make study participation not in the individual's best interest

  • Current or planned incarceration or other involuntary detention

Anticipated sample size: 2500

Interventions

Interventions:

Standard ATT drugs: 4‐month (17 weeks) regimen

8 weeks of daily treatment with rifapentine, isoniazid, pyrazinamide, and ethambutol, followed by

9 weeks of daily treatment with rifapentine and isoniazid

Moxifloxin combination: 4‐month (17 weeks) regimen: moxifloxacin for 4 months substituting ethambutol in intensive phase

8 weeks of daily treatment with rifapentine, isoniazid, pyrazinamide, and moxifloxacin, followed by

9 weeks of daily treatment with rifapentine, isoniazid, and moxifloxacin

Control: standard 6‐month (26‐week) ATT regimen

8 weeks of daily treatment with rifampin, isoniazid, pyrazinamide, and ethambutol, followed by

18 weeks of daily treatment with rifampin and isoniazid

Dosing:

All drugs are administered orally, 7 days/week, directly observed by a healthcare worker at least 5 of the 7 days each week. Pyridoxine (vitamin B6), 25 or 50 mg, is administered with each study dose

Study drug doses: rifampin 600 mg; isoniazid 300 mg; pyrazinamide < 55 kg 1000 mg, ≥ 55 to 75 kg 1500 mg, > 75 kg 2000 mg; ethambutol < 55 kg 800 mg, ≥ 55 to 75 kg 1200 mg, > 75 kg 1600 mg

Outcomes

Primary outcome measures:

  • TB disease‐free survival at 12 months after study treatment assignment [Time Frame: 12 months after treatment assignment]

  • Proportion of participants with grade 3 or higher adverse events during study drug treatment [Time Frame: 4 or 6 months]

Secondary outcome measures:

  • TB disease‐free survival at 18 months after study treatment assignment [Time Frame: 18 months after treatment assignment]

  • Proportion of participants who are culture negative at 8 weeks [Time Frame: 8 weeks]

Solid and liquid media considered separately:

  • Time to stable sputum culture conversion [Time Frame: 4 or 6 months] ‐ solid and liquid media considered separately

  • Speed of decline of sputum viable bacilli by automated MGIT days to detection [Time Frame: 4 or 6 months]

  • TB disease‐free survival at 12 and 18 months after study treatment assignment, assuming all losses to follow‐up and non‐tuberculosis deaths have an unfavourable outcome [Time Frame: 18 months after study treatment assignment]. Sensitivity analyses, assuming all losses to follow‐up and non‐tuberculosis deaths have an unfavourable outcome

  • TB disease‐free survival at 12 and 18 months after study treatment assignment, assuming all losses to follow‐up and non‐tuberculosis deaths have a favourable outcome [Time Frame: 18 months after study treatment assignment]. Sensitivity analyses, assuming all losses to follow‐up and non‐tuberculosis deaths have a favourable outcome

  • Discontinuation of assigned treatment for a reason other than microbiological ineligibility [Time Frame: 4 or 6 months]

  • Efavirenz maximum concentration, area under the time‐concentration curve, and half‐life [Time Frame: 4 months]

Among participants with HIV infection receiving efavirenz‐based antiretroviral therapy, these values will be used to estimate steady state efavirenz PK parameters including mid‐dosing interval concentration

Starting date

January 2016; estimated study completion date: December 2019

Contact information

Stefan Goldberg, Centers for Disease Control and Prevention. Ph: 404‐639‐5339; Email: [email protected]

Notes

Study locations: 38 including Brazil, China, India, and Malawi

Registration number:NCT02410772

Sponsor: Centers for Disease Control and Prevention

Collaborator: AIDS Clinical Trials Group

NCT02581527

Trial name or title

An international multi‐centre controlled clinical trial to evaluate 1200 mg and 1800 mg rifampicin daily in the reduction of treatment duration for pulmonary tuberculosis from 6 months to 4 months (RIFASHORT)

Methods

Study type: interventional, phase 3; study design: open‐label 3‐arm trial

Participants

Inclusion criteria:

  • Patients with:

    • Newly diagnosed, smear microscopy positive, pulmonary tuberculosis, rifampicin‐susceptible MTBC confirmed by Xpert MTB/RIF OR

    • Smear microscopy negative, suspected pulmonary tuberculosis, confirmed by Xpert MTB/RIF as sputum MTBC positive and rifampicin susceptible

  • No more than 1 week of previous treatment for tuberculosis, for active TB or confirmed or presumed latent TB infection

  • ≥ 18 years old

  • Consent to participation in the trial and to HIV testing

  • Provide informed consent

  • Stable home address within easy reach of the treatment facility and likely to remain there for the entire treatment and follow‐up period

  • Women who are pre‐menopausal or of childbearing age must be using a barrier form of contraception (condoms, diaphragms, cervical caps, or contraceptive sponges), or must be surgically sterilized or have an intrauterine coil device (IUCD) in place for the duration of the treatment phase; alternatively they should agree to abstain from sexual activity during the treatment phase.

Exclusion criteria:

  • Rifampicin resistance identified by Xpert MTB/RIF or by direct susceptibility testing (late exclusions)

  • Moribund phase

  • TB meningitis or extrapulmonary TB

  • Female and known to be pregnant or breastfeeding

  • Condition likely to lead to uncooperative behaviour such as psychiatric illness or alcoholism

  • History of seizures

  • Contraindications to any medications in the study regimens

  • HIV positive according to local testing algorithm

  • Blood disorder (including grade 4 or above thrombocytopenia)

  • Haemoglobin < 7 g/dL

  • Peripheral neuritis

  • Pre‐existing liver disease

  • ALT > 5 times upper limit of normal (ULN) for that laboratory

  • Raised bilirubin (grade 4 or above)

  • Kidney disease

  • Estimated creatinine clearance < 30 mL/min

  • Previously diagnosed diabetes mellitus (non‐insulin‐dependent or insulin‐dependent)

  • HbA1c > 48 mmol/mol

  • Weight < 35 kg

  • Taking any of the excluded medications listed in the Summary of Product Characteristics (SmPC) for any trial drugs

  • Pre‐existing non‐tuberculous disease likely to prejudice response to, or assessment of, treatment as judged by the Principal Investigator

Anticipated sample size: 654

Interventions

Interventions: 4‐month regimens

Rifampicin 1200 mg combination ATT regimen
2 months of daily ethambutol, isoniazid, rifampicin, and pyrazinamide, followed by

2 months of daily isoniazid and rifampicin

Supplement of 450 mg (weight bands 35 to 39 kg and 40 to 54 kg) or 600 mg (weight band 55 to 69 kg and 70 and more kg) of rifampicin will be given throughout the 4 months (2EHR 1200Z/2HR1200)

Rifampicin 1800 mg combination ATT regimen
2 months of daily ethambutol, isoniazid, rifampicin, and pyrazinamide, followed by

2 months of daily isoniazid and rifampicin

Supplement of 450 mg (weight bands 35 to 39 kg and 40 to 54 kg) or 600 mg (weight band 55 to 69 kg and 70 and more kg) of rifampicin will be given throughout the 4 months (2EHR1800Z/2HR1800)

Control:

Standard 6‐month ATT regimen

2 months of the standard regimen of isoniazid, pyrazinamide, and ethambutol plus 10 mg/kg rifampicin for the initial 8 weeks, followed by

4 months of isoniazid and rifampicin (at the same dose size) for an additional 4 months (2HRZE/4HR).

Outcomes

Primary outcomes:

  • Efficacy: proportion with a combined unfavourable endpoint measured 18 months from randomization; this endpoint includes failure at end of treatment, recurrence, and death. This will be measured in the modified intent‐to‐treat microscopy positive population

  • Safety: occurrence of grade 3 or 4 adverse events at any time during chemotherapy and 1 month post therapy in the intent‐to‐treat population with an MTBC positive test result Xpert MTB/RIF positive population

Secondary outcomes

  • Per‐protocol analysis of the primary efficacy outcome

  • Combined unfavourable endpoint measured 18 months from randomization in the Xpert MTB/RIF positive (i) modified intent‐to‐treat and (ii) per‐protocol populations

  • Sputum cultures positive for M tuberculosis at 8 and 12 weeks from randomization

  • Any adverse event, up to 1 month after end of treatment, graded according to DAIDS criteria

  • Time to unfavourable outcome in the modified to intent‐to‐treat and per‐protocol microscopy positive population

Starting date

March 2017; estimated study completion date: December 2020; status: recruiting

Contact information

Eduardo Rómulo Chávez Ticona. Calle Rio Huaura Nro. 319, Pueblo Libre, Lima, Peru Pueblo Libre Lima LIMA Perú. Ph: 993560268; Email: [email protected]

Notes

Study locations: Bolivia, Botswana, Peru, Uganda

Registration number:NCT02581527

Primary sponsor: St Georges University of London

NCT02901288

Trial name or title

Shortened regimens for drug‐susceptible pulmonary tuberculosis

Methods

Multi‐centre, randomized, non‐inferiority, open‐label, controlled phase 3 clinical trial

Participants

Patients with newly diagnosed drug‐susceptible pulmonary TB who fulfil the inclusion and exclusion criteria

Inclusion criteria:

  • Willing and able to give informed consent to participate in trial treatment and follow‐up

  • Aged 18 to 65 years

  • Twice positive acid‐fast bacilli (AFB) sputum smear or positive sputum culture result, along with chest X‐ray consistent with active pulmonary tuberculosis

  • Newly diagnosed cases receiving anti‐tuberculosis treatment for < 1 month

  • Urine human chorionic gonadotropin (U‐HCG) negative and must agree to use effective contraception during trial period

  • Alanine aminotransferase (ALT) and total bilirubin < 2 times upper limit of normal

  • Creatinine clearance rate > 30 mL/min

  • Haemoglobin > 7.0 g/dL

  • Platelets > 50 x 10⁹/L

Exclusion criteria:

  • Concomitant severe cardiovascular, liver, kidney, nervous system, haematopoietic system, and other disease, or concomitant neoplastic disease. Or extensive lesion with respiratory insufficiency

  • Uncontrolled diabetes mellitus

  • Concomitant mental disorder

  • HIV‐positive individuals

  • Critically ill patients

  • Pregnant or breastfeeding mothers

  • Unable or unwilling to comply with treatment, assessment, or follow‐up schedule

  • Taking any medications contraindicated with medicines in any trial regimen of the study

  • Known allergy to any drug of treatment regimens

  • Currently taking part in another trial

  • QTc interval > 480 ms

Anticipated sample size: 3900 (1300 in each group)

Interventions

Interventions:

Levofloxacin + ethambutol 4.5‐month combination ATT regimen

4.5 months of isoniazid, rifampin, pyrazinamide, ethambutol, and levofloxacin

Dosage: isoniazid 300 mg (given once daily), rifampin 450 mg (less than 50 kg given once daily) or 600 mg (more than 50 kg given once daily), pyrazinamide 1500 mg ((less than 50 kg given once daily) or 30 mg/kg (more than 50 kg once daily), ethambutol 750 mg (less than 50 kg once daily) or 1000 mg (more than 50 kg once daily), levofloxacin 600 mg (less than 50 kg given once daily) or 800 mg (more than 50 kg once daily)

Ethambutol 4.5‐month combination ATT regimen

4.5 months of isoniazid, rifampin, pyrazinamide, and ethambutol. Dosage of isoniazid, rifampin, pyrazinamide, and ethambutol is same as that of control regimen

Control:

Standard 6‐month ATT regimen

2 months of isoniazid, rifampin, pyrazinamide, and ethambutol, followed by

4 months of isoniazid and rifampin

Dosage: isoniazid 300 mg (given once daily), rifampin 450 mg (less than 50 kg given once daily) or 600 mg (more than 50 kg given once daily), pyrazinamide 1500 mg (less than 50 kg given once daily) or 30 mg/kg (more than 50 kg once daily), ethambutol 750 mg (less than 50 kg once daily), or 1000 mg (more than 50 kg, once daily)

Outcomes

Primary outcome measures:

  • Percentage of participants with TB recurrence/relapse by 24 months after end of treatment

  • Percentage of participants with treatment failure at 4.5 months or 6 months after randomization

Secondary outcome measures:

  • Treatment adverse reactions occurring

  • Time to sputum smear or culture conversion within intensive phase

  • Sputum smear or culture conversion proportion at treatment completion

  • Radiological manifestation change in TB lesion or cavity

  • Patient adherence rate

Starting date

August 2016; estimated study completion date: December 2018

Contact information

Shenjie Tang, MD. Beijing Chest Hospital

Email: [email protected]

Notes

Study location: China

Registration number:NCT02901288

Primary sponsors: Beijing Chest Hospital, Hubei Provincial Center for Disease Control and Prevention

Abbreviations: AFB: acid‐fast bacilli; ALP: alkaline phosphatase; ALT: alanine aminotransferase; ART: antiretroviral therapy; ARV: antiretroviral; AST: aspartate aminotransferase; ATT: anti‐tuberculosis treatment; CrCl: creatinine clearance; E: ethambutol; ECG: electrocardiogram; FSH: follicle‐stimulating hormone; H: isoniazid; HbA1c: glycosylated haemoglobin; IUCD: intrauterine coil device; LH: luteinizing hormone; M: moxifloxacin; MDR‐TB: multi‐drug‐resistant tuberculosis; MGIT: mycobacterial growth indicator tube; MIC: minimum inhibitory concentration; MTB: Mycobacterium tuberculosis; PK: pharmacokinetics; R: rifampicin; TB: tuberculosis; TEAE: treatment‐emergent adverse event; TTP: time to positivity; U‐HCG: urine human chorionic gonadotropin; ULN: upper limit of normal; WHO: World Health Organization; XDR: extensively drug‐resistant; Z: pyrazinamide.

Data and analyses

Open in table viewer
Comparison 1. Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse Show forest plot

3

2265

Risk Ratio (M‐H, Fixed, 95% CI)

3.56 [2.37, 5.37]

Analysis 1.1

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 1 Relapse.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 1 Relapse.

2 Relapse: subgroup analysis Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 2 Relapse: subgroup analysis.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 2 Relapse: subgroup analysis.

2.1 Moxifloxacin replacing ethambutol

2

1386

Risk Ratio (M‐H, Fixed, 95% CI)

2.74 [1.69, 4.43]

2.2 Moxifloxacin replacing isoniazid

2

1424

Risk Ratio (M‐H, Fixed, 95% CI)

4.89 [3.02, 7.92]

3 Relapse: sensitivity analysis accounting for missing data Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 3 Relapse: sensitivity analysis accounting for missing data.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 3 Relapse: sensitivity analysis accounting for missing data.

3.1 Modified‐ITT analysis

3

2265

Risk Ratio (M‐H, Fixed, 95% CI)

3.56 [2.37, 5.37]

3.2 Per‐protocol analysis

3

2135

Risk Ratio (M‐H, Fixed, 95% CI)

3.79 [2.48, 5.78]

3.3 Imputing missing data

3

2524

Risk Ratio (M‐H, Fixed, 95% CI)

3.83 [2.58, 5.70]

4 Death from any cause Show forest plot

3

2760

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.65, 1.75]

Analysis 1.4

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 4 Death from any cause.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 4 Death from any cause.

5 Treatment discontinuation Show forest plot

3

2335

Risk Ratio (M‐H, Fixed, 95% CI)

1.12 [0.78, 1.61]

Analysis 1.5

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 5 Treatment discontinuation.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 5 Treatment discontinuation.

6 Positive sputum culture/smear at 8 weeks Show forest plot

3

2828

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.22, 1.13]

Analysis 1.6

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 6 Positive sputum culture/smear at 8 weeks.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 6 Positive sputum culture/smear at 8 weeks.

6.1 Moxifloxacin replacing isoniazid or ethambutol in 4‐month ATT regimen

2

2087

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.45, 1.20]

6.2 Moxifloxacin augmenting standard 6‐month ATT regimen

1

741

Risk Ratio (M‐H, Random, 95% CI)

0.24 [0.15, 0.39]

7 Treatment failure Show forest plot

3

2282

Risk Ratio (M‐H, Fixed, 95% CI)

0.71 [0.33, 1.52]

Analysis 1.7

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 7 Treatment failure.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 7 Treatment failure.

8 Acquired drug resistance Show forest plot

3

2282

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.08, 1.31]

Analysis 1.8

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 8 Acquired drug resistance.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 8 Acquired drug resistance.

9 Serious adverse events Show forest plot

4

3548

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.74, 1.27]

Analysis 1.9

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 9 Serious adverse events.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 9 Serious adverse events.

9.1 Moxifloxacin replacing standard drugs in 4‐month ATT regimens

3

2760

Risk Ratio (M‐H, Fixed, 95% CI)

0.96 [0.72, 1.26]

9.2 Moxifloxacin augmenting standard 6‐month ATT regimens

1

788

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.45, 3.06]

Open in table viewer
Comparison 2. Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse Show forest plot

2

1633

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.56, 2.84]

Analysis 2.1

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 1 Relapse.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 1 Relapse.

2 Relapse: sensitivity analysis accounting for missing data Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.2

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 2 Relapse: sensitivity analysis accounting for missing data.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 2 Relapse: sensitivity analysis accounting for missing data.

2.1 Modified‐ITT analysis

2

1633

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.56, 2.84]

2.2 Per‐protocol analysis

2

1525

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.55, 2.87]

2.3 Modified‐ITT analysis (all eligible participants ‐ imputing missing data)

2

1851

Risk Ratio (M‐H, Fixed, 95% CI)

2.01 [1.53, 2.63]

3 Death from any cause Show forest plot

2

1886

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.53, 1.53]

Analysis 2.3

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 3 Death from any cause.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 3 Death from any cause.

4 Treatment discontinuation Show forest plot

2

1657

Risk Ratio (M‐H, Fixed, 95% CI)

0.70 [0.46, 1.08]

Analysis 2.4

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 4 Treatment discontinuation.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 4 Treatment discontinuation.

5 Positive sputum culture at 8 weeks Show forest plot

2

1818

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.80, 1.23]

Analysis 2.5

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 5 Positive sputum culture at 8 weeks.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 5 Positive sputum culture at 8 weeks.

6 Treatment failure Show forest plot

2

1657

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.51, 1.70]

Analysis 2.6

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 6 Treatment failure.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 6 Treatment failure.

7 Acquired drug resistance Show forest plot

1

301

Risk Ratio (M‐H, Fixed, 95% CI)

0.24 [0.01, 5.01]

Analysis 2.7

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 7 Acquired drug resistance.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 7 Acquired drug resistance.

8 Serious adverse events Show forest plot

2

1993

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.58, 1.77]

Analysis 2.8

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 8 Serious adverse events.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 8 Serious adverse events.

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included trial.
Figuras y tablas -
Figure 2

Risk of bias summary: review authors' judgements about each risk of bias item for each included trial.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 3

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Forest plot of comparison: 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, outcome: 1.3 Relapse: sensitivity analysis accounting for missing data.
Figuras y tablas -
Figure 4

Forest plot of comparison: 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, outcome: 1.3 Relapse: sensitivity analysis accounting for missing data.

Forest plot of comparison: 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, outcome: 2.2 Relapse: sensitivity analysis accounting for missing data.
Figuras y tablas -
Figure 5

Forest plot of comparison: 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, outcome: 2.2 Relapse: sensitivity analysis accounting for missing data.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 1 Relapse.
Figuras y tablas -
Analysis 1.1

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 1 Relapse.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 2 Relapse: subgroup analysis.
Figuras y tablas -
Analysis 1.2

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 2 Relapse: subgroup analysis.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 3 Relapse: sensitivity analysis accounting for missing data.
Figuras y tablas -
Analysis 1.3

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 3 Relapse: sensitivity analysis accounting for missing data.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 4 Death from any cause.
Figuras y tablas -
Analysis 1.4

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 4 Death from any cause.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 5 Treatment discontinuation.
Figuras y tablas -
Analysis 1.5

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 5 Treatment discontinuation.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 6 Positive sputum culture/smear at 8 weeks.
Figuras y tablas -
Analysis 1.6

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 6 Positive sputum culture/smear at 8 weeks.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 7 Treatment failure.
Figuras y tablas -
Analysis 1.7

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 7 Treatment failure.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 8 Acquired drug resistance.
Figuras y tablas -
Analysis 1.8

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 8 Acquired drug resistance.

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 9 Serious adverse events.
Figuras y tablas -
Analysis 1.9

Comparison 1 Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 9 Serious adverse events.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 1 Relapse.
Figuras y tablas -
Analysis 2.1

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 1 Relapse.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 2 Relapse: sensitivity analysis accounting for missing data.
Figuras y tablas -
Analysis 2.2

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 2 Relapse: sensitivity analysis accounting for missing data.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 3 Death from any cause.
Figuras y tablas -
Analysis 2.3

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 3 Death from any cause.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 4 Treatment discontinuation.
Figuras y tablas -
Analysis 2.4

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 4 Treatment discontinuation.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 5 Positive sputum culture at 8 weeks.
Figuras y tablas -
Analysis 2.5

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 5 Positive sputum culture at 8 weeks.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 6 Treatment failure.
Figuras y tablas -
Analysis 2.6

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 6 Treatment failure.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 7 Acquired drug resistance.
Figuras y tablas -
Analysis 2.7

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 7 Acquired drug resistance.

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 8 Serious adverse events.
Figuras y tablas -
Analysis 2.8

Comparison 2 Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens, Outcome 8 Serious adverse events.

Summary of findings for the main comparison. Moxifloxacin‐containing 4‐month ATT regimens versus standard 6‐month ATT regimen for drug‐sensitive pulmonary tuberculosis

Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimen for drug‐sensitive pulmonary tuberculosis

Patient or population: adults with drug‐sensitive pulmonary tuberculosis
Setting: low‐ and middle‐income countries in Africa, Asia, and Latin America
Intervention: moxifloxacin‐containing 4‐month ATT
Comparison: standard 6‐month ATT

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect

(95% CI)

Number of participants (studies)

Certainty of the evidence (GRADE)

Comments

Risk with 6‐month standard ATT

Risk with 4‐month moxifloxacin‐containing ATT

Relapse
Follow‐up: range 12 to 24 months

32 per 1000

82 more relapses per 1000
(44 more to 140 more)

RR 3.56
(2.37 to 5.37)

2265
(3 RCTs)

⊕⊕⊕⊝
MODERATEa,b,c

Due to indirectness

The 4‐month regimen probably increases relapse compared to the 6‐month regimen

Death from any cause

Follow‐up: range 18 to 24 months

21 per 1000

2 more deaths per 1000
(7 fewer to 16 more)

RR 1.06
(0.65 to 1.75)

2760
(3 RCTs)

⊕⊕⊕⊝
MODERATEa,c,d

Due to indirectness

The 4‐month regimen probably makes little or no difference in death from any cause compared to the 6‐month regimen

Treatment failure

16 per 1000

5 fewer treatment failures per 1000
(11 fewer to 8 more)

RR 0.71
(0.33 to 1.52)

2282
(3 RCTs)

⊕⊕⊕⊝
MODERATEa,c,d

Due to indirectness

The 4‐month regimen probably results in little or no difference in treatment failure compared to the 6‐month regimen

Acquired drug resistance

7 per 1000

5 fewer with acquired drug resistance per 1000

(6 fewer to 2 more)

RR 0.33
(0.08 to 1.31)

2282

(3 RCTs)e

⊕⊕⊝⊝
LOWc,f,g

Due to indirectness and imprecision

The 4‐month regimen may be little or no different than the 6‐month regimen in the incidence of acquired drug resistance

Serious adverse events

Follow‐up: range 18 to 24 months

62 per 1000

2 fewer with serious adverse events per 1000
(16 fewer to 16 more)

RR 0.97
(0.74 to 1.27)

3548
(4 RCTs)g

⊕⊕⊕⊝
MODERATEa,c,d,h

Due to indirectness

The 4‐month regimen probably results in little or no difference in serious adverse events compared to the 6‐month regimen

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
ATT: anti‐tuberculosis treatment; CI: confidence interval; RCT: randomized controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

aNo serious risk of bias: although Jawahar 2013 was at high risk of allocation bias, exclusion of this trial from the sensitivity analysis did not change the direction of effect. Not downgraded.
bNo serious inconsistency: although trial results indicated a moderate degree of heterogeneity (I² = 58%), the differences were between small and large effects favouring 6‐month ATT. Changing the model from fixed effect to random effects did not alter the direction of effect. Not downgraded.
cDowngraded one level for serious indirectness: trials excluded children and adolescents, people with diabetes, and other physical comorbid conditions.
dNo serious imprecision: the 95% CI for the risk ratio was wide but event rates were low and the sample size was large; the risk ratio and the 95% CI around absolute estimates did not indicate clinically appreciable differences with either regimen. Not downgraded.
eNo serious imprecision: the 95% CI for the risk ratio was wide but event rates were low and the sample size was large; the 95% CI for the risk ratio (RR 0.5% fewer with the 4‐month regimen, 95% CI 1.1% fewer to 0.8% more) did not indicate that there were clinically important differences in proportions with treatment failure. Not downgraded.
fDrug resistance was assessed using LJ solid media in one trial, MGIT liquid media in another trial, and either or both in the third trial.
gSerious imprecision: events were few and the 95% CI for the pooled estimate was wide. In the largest study that also reported the most events, results were equivocal for acquired resistance and only possible resistance was reported. Downgraded one level.
hThree trials provided data for all outcomes in this summary table (Gillespie 2014; Jawahar 2013; Jindani 2014); Velayutham 2014 provided data only for serious adverse events.

Figuras y tablas -
Summary of findings for the main comparison. Moxifloxacin‐containing 4‐month ATT regimens versus standard 6‐month ATT regimen for drug‐sensitive pulmonary tuberculosis
Summary of findings 2. Gatifloxacin‐containing 4‐month ATT regimens compared to standard 6‐month ATT regimens for drug‐sensitive pulmonary tuberculosis

Gatifloxacin‐containing 4‐month ATT regimens compared to standard 6‐month ATT regimens for drug‐sensitive pulmonary tuberculosis

Patient or population: adults with drug‐sensitive pulmonary tuberculosis
Setting: low‐ and middle‐income countries in sub‐Saharan Africa and India
Intervention: gatifloxacin‐containing 4‐month ATT regimen
Comparison: standard 6‐month treatment regimen

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with 6‐month standard ATT

Risk with gatifloxacin‐containing
4‐month ATT

Relapse
Follow‐up: 24 months

70 per 1000

77 more relapses per 1000
(32 more to 128 more)

RR 2.11
(1.56 to 2.84)

1633
(2 RCTs)

⊕⊕⊕⊝
MODERATEa,b

Due to indirectness

The 4‐month regimen probably increases relapse compared to the 6‐month regimen

Death from any cause
Follow‐up: 24 months

29 per 1000

3 fewer deaths per 1000
(14 fewer to 16 more)

RR 0.90
(0.53 to 1.53)

1886
(2 RCTs)

⊕⊕⊕⊝
MODERATEa,b,c

Due to indirectness

The 4‐month regimen probably makes little or no difference in death compared to the 6‐month regimen

Treatment failure

25 per 1000

1 less treatment failure per 1000
(12 fewer to 18 more)

RR 0.93
(0.51 to 1.70)

1657
(2 RCTs)

⊕⊕⊝⊝
MODERATEa,b,c

Due to indirectness

The 4‐month regimen probably makes little or no difference in treatment failure compared to the 6‐month regimen

Acquired drug resistance

12 per 1000

9 fewer with acquired drug resistance per 1000

(12 fewer to 49 more)

RR 0.24

(0.01 to 5.01)

301

(1 RCT)d

⊕⊝⊝⊝
VERY LOWb,e,f

Due to indirectness, risk of bias, and imprecision

We do not know if acquired drug resistance is any different in the 4‐month and the 6‐month regimens

Serious adverse events

24 per 1000

0 fewer serious adverse events per 1000
(10 fewer to 18 more)

RR 1.02
(0.58 to 1.77)

1993
(2 RCTs)

⊕⊕⊕⊝
MODERATEa,b,c

Due to indirectness

The 4‐month regimen probably results in little or no difference in serious adverse events compared to the 6‐month regimen

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
ATT: anti‐tuberculosis treatment; CI: confidence interval; RCT: randomized controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.

High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

aNo serious risk of bias: although Jawahar 2013 was assigned high risk of bias for allocation concealment, removal of this trial from the sensitivity analysis did not significantly alter the direction, magnitude, or precision of the effect estimate. Not downgraded.
bDowngraded one level for serious indirectness: trials excluded children and adolescents and people with diabetes mellitus and other comorbid physical conditions and those with alcohol abuse.
cNo serious imprecision: the 95% CI of the risk ratio was wide, but events were few and the sample size was reasonably large; the 95% CI for the absolute estimates did not indicate clinically appreciable benefits for either regimen. Not downgraded.
dOne trial provided data on acquired drug resistance (Jawahar 2013). Merle 2014 reported only drug susceptibility at baseline.
eDowngraded one level for serious risk of bias: allocation concealment was compromised and there were baseline imbalances in proportions with drug resistance at baseline in the sole trial for this outcome (Jawahar 2013).
fDowngraded two levels for very serious imprecision: the data for acquired resistance come from only one trial with 301 participants, and this trial did not evaluate resistance to gatifloxacin.

Figuras y tablas -
Summary of findings 2. Gatifloxacin‐containing 4‐month ATT regimens compared to standard 6‐month ATT regimens for drug‐sensitive pulmonary tuberculosis
Table 1. Summary of outcomes in included studies

Study ID

(Acronym)

Gillespie 2014

(REMoxTB)

Jawahar 2013

Jindani 2014

(RIFAQUIN)

Merle 2014

(OFLOTUB)

Velayutham 2014

Setting

Multiple sites in Africa (Kenya, South Africa, Tanzania, Zambia), Asia (China, India, Malaysia Thailand), Latin America (Mexico)

6 sites in 2 cities in India

6 cities in 4 countries in Africa (Botswana, South Africa, Zambia, Zimbabwe)

5 countries in Africa (Benin, Guinea, Kenya, Senegal, South Africa)

2 cities in India

Participants

Number randomized

1931

429

827

1836

801

Age

Adults (> 18 years)

Adults (> 18 years)

Adults (> 18 years)

Adults (18 to 65 years)

Adults (> 18 years)

HIV infection

Included (if CD4 count > 250 cells/μL and not on ART); 110 (7%)

Excluded

Included (if CD4 count > 150/mm³ and not on ART; 158 (27%)

Included if not stage 3 or 4 disease and not on ART; 304 (17%)

Excluded

Diagnosis of TB

Positive sputum smears on 2 occasions

Culture‐confirmed susceptibility to rifampicin, isoniazid, pyrazinamide, and moxifloxacin

Newly diagnosed pulmonary TB with at least 2 positive sputum cultures. Confirmed by culture and MDR‐TB excluded, susceptibility to ofloxacin (as proxy for moxifloxacin)

2 sputum samples that were positive for tubercle bacilli on direct smear microscopy

No resistance to isoniazid, rifampicin, or moxifloxacin

Acid‐fast bacilli in 2 consecutive sputum smears; confirmed by culture (solid medium) and drug sensitivity tests to rifampicin, isoniazid, ethambutol, streptomycin, and gatifloxacin

2 positive sputum smear smears for tuberculosis. Culture‐confirmed and MDR‐TB ruled out; susceptible to ofloxacin (as proxy for moxifloxacin)

Intervention(s) and comparator

Duration of ATT

4 monthsa

6 months

4 monthsb

6 months

4 months

6 monthsc

4 months

6 months

4 monthsa

6 months

Regimens

2HRZM/2HRM

+

2MRZE/2MR

2HRZE/4HR

2(HRZG)₃/

2 (HRG)₃

2(HRZM)₃/2(HRM)₃

2(HRZE)₃ /4(HR)₃

2MRZE/

2P₂M₂

2HRZE/

4HR

2HRZG/

2HRG

2HRZE/4HR

3HRZM

+

2HRZM/

2RHM

+

2HRZM/

2(RHM)₃

+

2HRZM/

2(RHEM)₃

2(HRZE)₃/

4(HR)₃

Number allocated

655 + 636 = 1291

640

141

118

170

275

275

917

919

629

172

Late screening failures excluded after allocation

38 + 32

= 70

40

5

3

5

36

35

62

51

13

8

Number eligible

1231

600

136

115

165

239

240

852

868

616

164

Number analysed in m‐ITT analysis (% of those allocated)

568 + 551 = 1119

(87)

555

(87)

136

(97)

115

(98)

165

(97)

193

(70)

188

(68)

791

(86)

794

(86)

590

(94)

151

(88)

Number analysed in per‐protocol analysis (% of those allocated)

514 + 524

=

1038

(80)

510

(80)

131

(93)

113

(96)

159

(94)

165

(60)

163

(59)

651

(71)

601

(65)

As above

Number analysed in ancillary analysis (ITT) (% of those allocated)

617 + 604

= 1221 (94)

600

(94)

Not done

239

(87)

240 (87)

Not reported

Not reported

Outcomes reported

Relapse

Relapse within 18 months after randomization in those with negative culture with treatment. Relapse strains were those shown to be identical on 24‐locus MIRU analysis

LJ solid media and MGIT liquid media used for culture

Recurrence of TB over 24 months after treatment in those with a favourable response with treatment: either bacteriologic recurrence (LJ solid media) or clinical/radiologic recurrence
Relapse not differentiated from re‐infection but majority occurred within 6 months after treatment

Relapse within 12 to 18 months after treatment. Two positive cultures within a period of 4 months without an intervening negative culture). Re‐infections differentiated from relapse through genotyping (MIRU‐VNTRs)

LJ solid media used for culture in some centres, MGIT liquid media in others, and both in some centres

Recurrence of TB over 24 months after treatment proven bacteriologically (2 consecutive positive sputum samples a day apart) or clinically

Genotyping (MIRU‐VNTRs) results available for only 70/140 (55%) of those with culture confirmed recurrence. Most were relapses

Not reported

Deaths

All deaths

TB deaths

Reported (only non‐TB deaths occurred)

All deaths

TB deaths

Death during treatment

Death after treatment

Not reported

Treatment discontinuation

Includes those who did not complete treatment, relocated, or withdrew consent

Includes those who did not complete treatment and those lost to follow‐up

Includes change in treatment due to adverse events, loss to follow‐up, and other treatment changes

Includes those who withdrew consent during treatment and dropouts

Reported but disaggregated data for each group not available

Positive smear/ sputum culture at 2 months

Reported using LJ solid media (used in this review) and MGIT liquid media for all randomized participants excluding late screening failures

Reported using LJ solid media for all randomized participants excluding late screening failures

Reported but disaggregated data for moxifloxacin 4‐month and 6‐month treatment groups not available

Data also not available for all participants from LJ media

Reported for 752 in the 4‐month and 759 in the 6‐month regimens (88% and 87% of those eligible, respectively)

Culture using LJ solid media

Reported for 590

(94%) in the 4‐month and 151

(88%) in the 6‐month regimens

Acquired drug resistance

Reported

Reported

Reported

Not reported

Not reported

Treatment failure

Includes culture confirmed and not confirmed

Includes culture confirmed and unconfirmed

Culture confirmed

Includes culture confirmed failure

Not reported

Serious adverse events

Reported for all randomized participants excluding late screening failures. Grade 3 and 4 severity (DAIDS 2009)

Deduced from adverse events reported for all randomized participants excluding late screening failures. Not graded

Reported for all participants randomized who took 1 dose and assessed as severe or life‐threatening during and 2 weeks after treatment. grade 3 and 4 severity (DAIDS 2009)

Reported for 1692 (92%) of all randomized participants.

grade 3 and 4 severity (DAIDS 2009)

Deduced from adverse events reported. Not graded

Other adverse events

Not reported

Reported

Not reported

QT prolongation

Hyperglycaemic episodes

Reported

Abbreviations: ART: anti‐retroviral treatment; ATT: anti‐tuberculosis treatment; E: ethambutol; G: gatifloxacin; H: isoniazid; ITT: intention‐to‐treat; LJ: Löwenstein‐Jensen; M: moxifloxacin; MGIT: mycobacterial growth indicator tube; MIRU‐VNTRs: mycobacterial interspersed repetitive unit–variable number tandem repeats; m‐ITT: modified intention‐to‐treat; P: rifapentine; R: rifampicin; Z: pyrazinamide.

Leading numbers in regimens indicate duration in months. Drugs were administered daily, except when given thrice weekly as indicated by subscripts.

aData from moxifloxacin‐containing shortened regimens combined for data synthesis.
bData from the 2 shortened regimens compared separately with standard 6‐month regimens.
cData from an additional arm evaluating moxifloxacin‐containing 6‐month regimen not included.

Figuras y tablas -
Table 1. Summary of outcomes in included studies
Table 2. Sensitivity analysis: moxifloxacin‐based 4‐month versus standard 6‐month ATT regimens

Primary outcome: relapse

Trial ID

Gillespie 2014

Jawahar 2013

Jindani 2014

Regimens

4 months

6 months

4 months

6 months

4 months

6 months

aModified‐ITT analysis (primary analysis)

110/1119

(9.8%)

13/555

(2.3%)

11/108

(10.1%)

10/155

(6.5%)

27/165

(16.4%)

6/163

(3.7%)

aPer‐protocol analysis

110/1038

(10.6%)

12/510

(2.4%)

11/107

(10.1%)

10/152

(6.6%)

26/165

(15.8%)

5/163

(3.1%)

bSensitivity analysis imputing missing data

126/1184

(10.7%)

14/577

(2.4%)

11/114

(9.7%)

10/159

(6.3%)

36/225

(16.0%)

71/232

(2.6%%)

Abbreviations: ATT: anti‐tuberculosis treatment; ITT: intention‐to‐treat.

aAs reported in trial reports.
bIncludes in the denominators for each trial arm all randomized participants minus those excluded post randomization due to ineligibility (not confirmed TB, or drug resistant), those who died, and those who experienced treatment failure. The difference in this denominator and the denominator in per‐protocol analyses are missing data. Relapse rates for missing people were imputed from rates in the per‐protocol analysis for each trial arm.

Figuras y tablas -
Table 2. Sensitivity analysis: moxifloxacin‐based 4‐month versus standard 6‐month ATT regimens
Table 3. Sensitivity analysis: gatifloxacin‐based 4‐month versus standard 6‐month ATT regimens

Primary outcome: relapse

Trial ID

Jawahar 2013

Merle 2014

Regimen

4 months

6 months

4 months

6 months

aModified‐ITT analysis (primary analysis)

19/122

(15.6%)

10/155

(6.5%)

101/694

(14.6%)

47/662

(7.1%)

aPer‐protocol analysis

19/121

(15.7%)

10/152

(6.6%)

98/651

(15.1%)

44/601

(7,3%)

bSensitivity analysis imputing missing data

19/132

(14.4%)

10/159

(6.3%)

122/786

(15.5%)

61/774

(7,9%)

Abbreviations: ATT: anti‐tuberculosis treatment; ITT: intention‐to‐treat.

aAs reported in trial reports.
bIncludes in the denominators for each trial arm all randomized participants minus those excluded post randomization due to ineligibility (not confirmed TB, or drug resistant), those who died, and those who experienced treatment failure. The difference in this denominator and the denominator in per‐protocol analyses are missing data. Relapse rates for missing people were imputed from rates in the per‐protocol analysis for each trial arm.

Figuras y tablas -
Table 3. Sensitivity analysis: gatifloxacin‐based 4‐month versus standard 6‐month ATT regimens
Comparison 1. Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse Show forest plot

3

2265

Risk Ratio (M‐H, Fixed, 95% CI)

3.56 [2.37, 5.37]

2 Relapse: subgroup analysis Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2.1 Moxifloxacin replacing ethambutol

2

1386

Risk Ratio (M‐H, Fixed, 95% CI)

2.74 [1.69, 4.43]

2.2 Moxifloxacin replacing isoniazid

2

1424

Risk Ratio (M‐H, Fixed, 95% CI)

4.89 [3.02, 7.92]

3 Relapse: sensitivity analysis accounting for missing data Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Modified‐ITT analysis

3

2265

Risk Ratio (M‐H, Fixed, 95% CI)

3.56 [2.37, 5.37]

3.2 Per‐protocol analysis

3

2135

Risk Ratio (M‐H, Fixed, 95% CI)

3.79 [2.48, 5.78]

3.3 Imputing missing data

3

2524

Risk Ratio (M‐H, Fixed, 95% CI)

3.83 [2.58, 5.70]

4 Death from any cause Show forest plot

3

2760

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.65, 1.75]

5 Treatment discontinuation Show forest plot

3

2335

Risk Ratio (M‐H, Fixed, 95% CI)

1.12 [0.78, 1.61]

6 Positive sputum culture/smear at 8 weeks Show forest plot

3

2828

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.22, 1.13]

6.1 Moxifloxacin replacing isoniazid or ethambutol in 4‐month ATT regimen

2

2087

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.45, 1.20]

6.2 Moxifloxacin augmenting standard 6‐month ATT regimen

1

741

Risk Ratio (M‐H, Random, 95% CI)

0.24 [0.15, 0.39]

7 Treatment failure Show forest plot

3

2282

Risk Ratio (M‐H, Fixed, 95% CI)

0.71 [0.33, 1.52]

8 Acquired drug resistance Show forest plot

3

2282

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.08, 1.31]

9 Serious adverse events Show forest plot

4

3548

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.74, 1.27]

9.1 Moxifloxacin replacing standard drugs in 4‐month ATT regimens

3

2760

Risk Ratio (M‐H, Fixed, 95% CI)

0.96 [0.72, 1.26]

9.2 Moxifloxacin augmenting standard 6‐month ATT regimens

1

788

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.45, 3.06]

Figuras y tablas -
Comparison 1. Moxifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens
Comparison 2. Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse Show forest plot

2

1633

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.56, 2.84]

2 Relapse: sensitivity analysis accounting for missing data Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2.1 Modified‐ITT analysis

2

1633

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.56, 2.84]

2.2 Per‐protocol analysis

2

1525

Risk Ratio (M‐H, Fixed, 95% CI)

2.11 [1.55, 2.87]

2.3 Modified‐ITT analysis (all eligible participants ‐ imputing missing data)

2

1851

Risk Ratio (M‐H, Fixed, 95% CI)

2.01 [1.53, 2.63]

3 Death from any cause Show forest plot

2

1886

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.53, 1.53]

4 Treatment discontinuation Show forest plot

2

1657

Risk Ratio (M‐H, Fixed, 95% CI)

0.70 [0.46, 1.08]

5 Positive sputum culture at 8 weeks Show forest plot

2

1818

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.80, 1.23]

6 Treatment failure Show forest plot

2

1657

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.51, 1.70]

7 Acquired drug resistance Show forest plot

1

301

Risk Ratio (M‐H, Fixed, 95% CI)

0.24 [0.01, 5.01]

8 Serious adverse events Show forest plot

2

1993

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.58, 1.77]

Figuras y tablas -
Comparison 2. Gatifloxacin‐containing 4‐month ATT versus standard 6‐month ATT regimens