Volume 23, Issue 11 pp. 978-990
Invited Review Series: Tuberculosis Updates 2018
Free Access

New drugs and regimens for tuberculosis

Kwok-Chiu Chang

Corresponding Author

Kwok-Chiu Chang

Department of Health, Tuberculosis and Chest Service, Hong Kong, China

All authors contributed equally to this work.Correspondence: Kwok-Chiu Chang, Department of Health, Tuberculosis and Chest Service, Wanchai Chest Clinic, 1st Floor, Wanchai Polyclinic, 99, Kennedy Road, Wanchai, Hong Kong, China. Email: [email protected]Search for more papers by this author
Eric Nuermberger

Eric Nuermberger

Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA

All authors contributed equally to this work.Search for more papers by this author
Giovanni Sotgiu

Giovanni Sotgiu

Clinical Epidemiology and Medical Statistics Unit, Department of Biomedical Sciences, University of Sassari, Sassari, Italy

All authors contributed equally to this work.Search for more papers by this author
Chi-Chiu Leung

Chi-Chiu Leung

Department of Health, Tuberculosis and Chest Service, Hong Kong, China

All authors contributed equally to this work.Search for more papers by this author
First published: 19 June 2018
Citations: 24
Series Editors: Chi Chiu Leung, Cynthia Chee and Ying Zhang

ABSTRACT

Since standardized rifampin-based first-line regimens and fluoroquinolone-based second-line regimens were used to treat tuberculosis (TB), unfortunately without timely modification according to the drug resistance profile, TB and drug-resistant disease are still important public health threats worldwide. Although the last decade has witnessed advances in rapid diagnostic tools and use of repurposed and novel drugs for better managing drug-resistant TB, we need an appropriate TB control strategy and a well-functioning health infrastructure to ensure optimal operational use of rapid tests, judicious use of effective treatment regimens that can be rapidly tailored according to the drug resistance profile and timely management of risk factors and co-morbidities that promote infection and its progression to disease. We searched the published literature to discuss (i) standardized versus individualized therapies, including the choice between a single one-size-fit-all regimen versus different options with different key drugs determined mainly by rapid drug susceptibility testing, (ii) alternative regimens for managing drug-susceptible TB, (iii) evidence for using the World Health Organization (WHO) longer and shorter regimens for multidrug-resistant TB and (iv) evidence for using repurposed and novel drugs. We hope an easily applicable combination of biomarkers that accurately predict individual treatment outcome will soon be available to ultimately guide individualized therapy.

Abbreviations

  • DOTS
  • Directly Observed Treatment, Short-course
  • DST
  • drug susceptibility testing
  • FQ
  • fluoroquinolone
  • MDR-TB
  • multidrug-resistant TB
  • SLID
  • second-line injectable drug
  • TB
  • tuberculosis
  • WHO
  • World Health Organization
  • XDR-TB
  • extensively drug-resistant TB
  • INTRODUCTION

    Tuberculosis (TB) is an old disease that dates back to at least 4000 years.1 Like any other infectious disease, an epidemic curve can be drawn for TB,2 which shows that it will take about 300 years for a TB epidemic to naturally die out. Since standardized rifampin-based first-line regimens were available3 and fluoroquinolone-based second-line regimens were used, unfortunately without timely modification according to the drug resistance profile, TB and drug-resistant disease have remained important public health threats worldwide, with an estimated 10.4 million TB new cases in 2016, including 600 000 rifampin-resistant TB, of which 490 000 were multidrug-resistant TB (MDR-TB, defined as TB with bacillary resistance to at least isoniazid and rifampin).4-7 The reported treatment success rate was only 54% for MDR-TB.7 The End TB Strategy has set ambitious targets that include a 90% reduction in TB deaths and an 80% reduction in TB incidence by 2030 in comparison with 2015.7 Although the last decade has witnessed advances in rapid diagnostic tools and use of repurposed and novel drugs for better managing drug-resistant TB,8 we would probably require a new TB vaccine in order to achieve the specific targets set in the End TB strategy. Short of a major breakthrough in the development of TB vaccines,7 we need an appropriate TB control strategy and a well-functioning health infrastructure to ensure optimal operational use of rapid tests, judicious use of effective TB regimens that can be rapidly customized according to the drug resistance profile and proper management of TB risk factors and co-morbidities that promote infection and its progression to disease.

    The spread of MDR-TB in numerous low- and middle-income countries has significantly weakened already fragile healthcare systems. To cure TB disease and properly manage treatment failure and relapse, we need one or more treatment regimens for tackling both drug-susceptible and drug-resistant TB in the best possible control strategy. National TB control strategies may be broadly classified into standardized and individualized therapies.9 Standardized treatment enables simple operation and broadens access to care but at the expense of possible amplification of existing but undetected drug resistance. On the contrary, individualized treatment helps customize drug combinations to avoid amplification of drug resistance and toxicities, but it is heavily dependent on drug susceptibility testing (DST) services. While the World Health Organization (WHO) has recommended the longer MDR-TB regimens, which may be standardized or individualized, and the shorter MDR-TB regimens, which are largely standardized,9 an one-size-fit-all approach has also been advocated to develop an injection-free regimen that could be administered to any patient with active TB, with the assumptions that DST and new drug regimens are readily available, and bacillary resistance to new drugs has not developed to any significant levels.10

    Host-directed therapies are outside the scope of this review, but the important role of host immunity in TB pathogenesis and the emergence of MDR-TB have fuelled enthusiasm in exploring their use as adjunctive treatment options for either shortening treatment or improving treatment outcomes.11 By either amplifying host immunity or dampening destructive inflammation, host-directed therapies have involved use of small molecule drugs (such as metformin, nonsteroidal anti-inflammatory drugs and statins) and immunomodulatory biologics (such as vitamins A and D3, micro RNA and interleukin-2).11

    We searched the published literature to discuss (i) standardized versus individualized therapies, including the choice between a single one-size-fit-all regimen versus different options with different key drugs determined mainly by rapid DST, (ii) alternative regimens that may help facilitate management of drug-susceptible TB, (iii) evidence for using the WHO longer and shorter MDR-TB regimen and (4) evidence for using repurposed (linezolid and clofazimine) and new (bedaquiline, delamanid and pretomanid) drugs in the management of fluoroquinolone-susceptible versus fluoroquinolone-resistant MDR-TB. On the basis of these findings, we suggest the way forward for optimally controlling the TB epidemic.

    NATIONAL TB CONTROL STRATEGY: STANDARDIZED VERSUS INDIVIDUALIZED THERAPIES

    Strategic considerations

    The current TB control strategy focuses on effective treatment of infectious sources. Unfortunately, with the intrinsic biological characteristics of the pathogen, it requires long duration of treatment with often complex combination regimens to cure the disease.3 Using this method of control, a sustainable treatment programme with good coverage (>70%) and high cure rate (>85%) requires many decades to bring the disease incidence down.12 With the association between TB and poverty, there are often limited healthcare resources and poor infrastructure to support TB treatment programme in the most severely affected areas.13 The choice between standardized or an individualized regimen for treating TB is, therefore, not simply an academic issue but an ongoing strategic adjustment to the changing field circumstances and ever-evolving technologies.

    Directly observed treatment, short-course

    In the 1970s, a number of milestone studies conducted in Hong Kong, Singapore, India and Africa under the leadership of the British Medical Research Council established the efficacy of the current standard 6-month short-course regimen under direct treatment observation.3 The 6-month standard short-course regimen appeared to work equally well across a full spectrum of infectious pulmonary TB in diverse ethnic groups. With use of all four first-line drugs for 6 months, reasonable efficacy was also shown in the presence of isoniazid resistance.14 Since TB was declared a global emergency in 1993, the standardized approach facilitated its subsequent implementation in many high TB burden but resource-limited countries under the WHO Directly Observed Treatment, Short-course (DOTS) strategy, which has been reported to result in cure rates of >80% and default rates of <10%, with relapse-free cure rates of 90–95% consistently reported among patients with drug-susceptible TB treated with standard 6-month regimens in clinical trial settings.3, 15, 16

    Tailoring regimen choices according to drug resistance profiles

    Despite the rapid global expansion of DOTS coverage, the annual decline in TB incidence remained only at 2% in 2016 and resistance has also emerged literally to every drug that has been used to treat TB.7 The spread of MDR-TB, and later of extensively drug-resistant TB (XDR-TB), has necessitated the introduction of new regimens containing second-line, repurposed and/or new drugs under the guidance of genotypic and/or phenotypic DST.17 While this necessarily involves tailoring treatment choice by DST of key first- and second-line drugs, the strategic choice between standardized approach and individually tailored approach still depends critically on the local resistance profiles and available diagnostic and treatment sources. Local DST data, which may be obtained by surveillance systems or periodic surveys and done by quality-assured, reliable and efficient laboratories, should inform clinicians about the choice of the most likely effective regimen.18

    Single versus different regimens for drug-susceptible and drug-resistant TB

    With the development of new drugs that might work equally well in TB with or without bacillary resistance to existing first- and second-line drugs, attempts are being made to develop a novel TB regimen that can work for both drug-susceptible and drug-resistant TB, as shown by NC-005 trial, STAND trial and SimpliciTB trial (see Table 2), thus circumventing expensive genetic and/or phenotypic DST even in areas with high prevalence of drug resistance. While this approach might be attractive, there remained substantial concerns over the adverse effect profile of some of these newly introduced drugs, especially if they are to be regularly used in the treatment of the huge number of patients with fully drug-susceptible TB. Furthermore, like all antibiotics, new TB drugs are not exempted from the emergence of drug resistance,19, 20 thereby posing a question mark on the longer term sustainability of such an approach.

    Individually tailored regimens for drug-susceptible TB

    Increasing evidence is now available to show the substantial impact of host factors (e.g. smoking,21 diabetes mellitus,22 HIV,23 pharmacokinetics24-26 and pharmacogenetics25, 27, 28) and disease characteristics (e.g. cavity,16, 26, 29 baseline and/or 2-month smear/culture status16, 29, 30) on various aspects of treatment outcomes. For yet unclear reasons, possibly related to intermittent treatment or lower WHO-recommended dosages (especially rifampin and pyrazinamide), the standard short-course regimen also failed in a number of recent clinical trials31-33 to reproduce the very high treatment efficacy as observed in earlier trials conducted by the British Medical Research Council.3 As a high cure rate is necessary not only to interrupt the transmission chain but also to minimize the emergence of drug resistance,12 there has been recent revival of interest on the possible roles of individually tailored regimens to either shorten the duration of treatment of some forms of TB or to maximize the overall cure rate.25, 26, 30 While it may be possible to use a shorter regimen to treat some patients with smear-negative pulmonary TB,30 a previous study showed that shortening treatment from 6 to 4 months in adults with noncavitary disease and culture conversion at 2 months resulted in higher relapse rates.34 The ultimate utility of individualized regimens will depend heavily on whether we can identify an easily applicable combination of biomarkers that accurately predict individual treatment outcome.

    DRUG-SUSCEPTIBLE TB: ALTERNATIVE REGIMENS THAT MAY HELP FACILITATE ITS MANAGEMENT

    Shorter treatment would facilitate management of drug-susceptible TB by improving treatment adherence and increasing the chance of treatment completion. However, there is as yet insufficient clinical evidence for shortening the standard 6-month regimen.

    Rifampin is the key sterilizing drug in the standard short-course regimen. Although it has been in use for nearly 50 years, accumulating evidence suggests that currently recommended dosages are suboptimal and that higher doses could be used to safely shorten therapy or improve clinical outcomes. A dose-ranging trial has demonstrated that 2 weeks of rifampin dosed up to 35 mg/kg/day is safe and well tolerated, with a greater estimated fall in bacterial load in the higher dosing groups.35 A subsequent phase 2 clinical trial in a multiple-arm, multiple-stage design (NCT01785186) suggested that rifampin 35 mg/kg/day and rifampin 20 mg/kg/day with moxifloxacin may have the potential to shorten treatment. Four-month regimens with rifampin dosed to either 1200 mg daily or 1800 mg daily are being evaluated in the RIFASHORT trial (NCT02581527). High-dose rifampin has also been explored for treatment of TB meningitis.36, 37 Although the results of two randomized trials appear conflicting, they may indicate that initial intravenous administration is the preferred route for initial high-dose rifampin treatment in this lethal disease due to prominent first-pass metabolism and marked inter-individual variability in rifampin bioavailability.

    Rifapentine is another rifamycin derivative with more potent activity in vitro and a longer half-life in vivo compared with rifampin. Although rifapentine was initially approved for use in once-weekly continuation phase therapy, this regimen had inferior efficacy in patients at high risk of relapse.29, 38, 39 Daily dosing regimens that greatly multiplied rifapentine exposures were studied in murine models, where they exhibited dose-dependent treatment-shortening effects.40, 41 Although substituting rifapentine 10 mg/kg/day for rifampin was not associated with a significant change in 8-week culture conversion rates,42 a subsequent dose-ranging study demonstrated that higher rifapentine doses of 15–20 mg/kg/day significantly improve 2-month sputum culture conversion compared with standard therapy.43 Four-month regimens containing rifapentine 1200 mg daily, with or without moxifloxacin replacing ethambutol and given throughout, are being evaluated in TBTC Study 31/ACTG A5349 (NCT02410772).

    Despite encouraging findings from murine TB models and those from five randomized controlled trials and one cohort study,44-50 four randomized controlled trials failed to demonstrate the non-inferiority of 4-month regimens containing either moxifloxacin or gatifloxacin in place of ethambutol or isoniazid,31-33 or moxifloxacin and high-dose twice-weekly rifapentine in place of isoniazid and rifampin, respectively.51

    Murine model studies first suggested that a novel rifamycin-sparing combination of the investigational new nitromidazole, pretomanid, with moxifloxacin and pyrazinamide could be as effective as regimens containing rifampin, moxifloxacin and pyrazinamide and superior to the standard regimen.52 After successful phase 2 trials,53, 54 4- and 6-month treatment durations of the novel regimen were evaluated in the phase 3 STAND trial (NCT02342886). However, the trial was temporarily put on hold owing to concerns about hepatotoxicity before the sponsor discontinued enrolment in favour of a new phase 2 trial SimpliciTB (NCT03338621) that will evaluate a more promising 4-month 4-drug regimen that combines bedaquiline with pretomanid, moxifloxacin and pyrazinamide. This regimen is much more potent in mice,55 and was associated with near-100% sputum culture conversion in MDR-TB patients at 2 months of treatment using the stringent MGIT-960 liquid culture system (unpublished data of NC-005 trial presented in the 47th Union World Conference on Lung Health in 2016).

    Although it is still not possible to shorten treatment for all drug-susceptible TB patients, it may be possible to shorten treatment for the majority with more individualized treatment approaches. For example, a recent meta-analysis of the phase 3 fluoroquinolone trials indicated that the majority of patients had good outcomes with the 4-month regimen and that, if ‘hard-to-treat’ patients expected to require 6 months of treatment for cure (e.g. HIV co-infection, cavitation, 3+ or 4+ sputum smear grade, etc.) could be identified by baseline and/or on-treatment characteristics, other patients may do well with 4-month regimens in a stratified approach (unpublished data presented in the 47th Union World Conference on Lung Health in 2016).

    In addition, more efficient supervision of therapy and improved treatment adherence may be achieved using once-weekly dosing in the continuation phase. A 6-month regimen with moxifloxacin and rifapentine 1200 mg administered once-weekly in the 4-month continuation phase was found to be non-inferior to the standard 6-month regimen.51 A caveat is the importance of excluding rifampin resistance by rapid molecular DST to avoid amplification of drug resistance.

    MDR-TB: WHO LONGER AND SHORTER MDR-TB TREATMENT REGIMENS

    As available therapeutic options for the management of MDR-TB are quantitatively and qualitatively inadequate, many MDR-TB patients have been treated with poorly effective, highly toxic and expensive drugs.4-6, 56

    On the basis of the reviews of aggregated and individual patient data from published and unpublished studies carried out by an international scientific group,57, 58 WHO has revised its previous guidelines on the programmatic management of MDR-TB,6, 9 with recommendations for a shorter standardized MDR-TB regimen for carefully selected patients and a longer individualized regimen for those ineligible for the shorter one. Individual patient data meta-analysis showed that MDR-TB treatment success and survival were associated with use of later-generation fluoroquinolones, ethionamide or prothionamide, four or more likely effective drugs in the intensive phase and three or more likely effective drugs in the continuation phase.57 Treatment success was higher among MDR-TB patients without additional drug resistance or with bacillary resistance to second-line injectable drugs only, than those with fluoroquinolone resistance alone or XDR-TB.58 Among patients with XDR-TB, treatment success was highest when there were at least six drugs in the intensive phase and four in the continuation phase, with treatment success maximized by 6.6–9.0 months of treatment during the intensive phase and 20.1–25.0 months overall, and by including more drugs than those recommended for MDR-TB.58

    Shorter standardized MDR-TB treatment regimens

    On the basis of the so-called Bangladesh regimen established by a series of six prospective observational studies over a period of 12 years carried out in Bangladesh,59 and reaffirmed by studies containing the regimen or its 12-month variants,60-62 the shorter MDR-TB regimen was targeted at treatment-naïve individuals without any known bacillary resistance to second-line drugs. The shorter regimen comprises four core drugs (gatifloxacin or moxifloxacin, clofazimine, ethambutol and pyrazinamide) given throughout, supplemented by kanamycin, prothionamide and high-dose isoniazid in the initial 4–6 months. The programmatic implementation of the shorter MDR-TB regimen relies on the availability of rapid molecular DST methods for second-line drugs and a low prevalence of bacillary resistance to each key regimen component.63, 64 The shorter MDR-TB regimen has been conditionally recommended in specific situations where resistance to at least fluoroquinolones and second-line injectable is considered unlikely by DST, or by reference to drug exposure history, use of second-line medicines at country level or recent representative surveillance data.9 While there is evidence supporting the effectiveness of the standardized shorter MDR-TB regimen under the above-mentioned conditions,65 and feasibility of its use for MDR-TB with low-level fluoroquinolone resistance,66, 67 there are major concerns regarding its applicability and sustainability in endemic areas with a large number of previously treated MDR-TB patients and a substantial proportion of MDR-TB strains that are resistant to one or more of the drugs in the shorter regimen.68 Preliminary findings of the STREAM Stage 1 (see Table 2) could not confirm non-inferiority of the shorter MDR-TB regimen or the superiority of the WHO-recommended 18- to-24-month MDR-TB regimen.69 The WHO recommendation regarding use of the shorter MDR-TB regimen remains unchanged.

    Longer individualized MDR-TB treatment regimens

    On the basis of the evidence reviews for effectiveness and safety, WHO has regrouped TB drugs into six major groups (A, B, C, D1, D2 and D3) to reflect the importance of later-generation fluoroquinolones and second-line injectable agents and recognize an increasingly important role for repurposed agents (linezolid and clofazimine) and novel drugs (bedaquiline and delamanid), removed macrolides from use in MDR-TB treatment and updated recommendations for the longer individualized 20-month MDR-TB regimen.6, 9 A stepwise approach has been proposed for formulating the longer regimen with an intensive phase that contains at least five likely effective agents in the intensive phase: (i) including one Group A drug (later-generation fluoroquinolones), (ii) adding a Group B drug (second-line injectable agent), (iii) adding two or more Group C drugs (ethionamide/prothionamide, cycloserine/terizidone, linezolid and clofazimine), (iv) adding Group D1 drugs (pyrazinamide and any other first-line agent), (v) adding Group D2 drugs (bedaquiline and delamanid) and (vi) adding Group D3 drugs (para-aminosalicylic acid, carbapenem with amoxicillin-clavulanate and thiocetazone).

    In a recent meta-analysis, individualized MDR-TB treatment regimens achieved higher success rates than standardized counterparts.70 However, with the major gap in treatment of MDR-TB cases in many high-burden but resource-limited areas, the individually tailored 20-month longer regimens might be excessively long for large-scale implementation.7, 9

    MDR-TB: REPURPOSED AND NOVEL DRUGS

    The past decade has witnessed frequent use of two repurposed agents (linezolid and clofazimine) and two clinically approved novel drugs (bedaquiline and delamanid) in non-trial settings, often on a compassionate basis, owing to the poor treatment response of complicated MDR-TB to conventional fluoroquinolone-based MDR-TB treatment regimens. The initial evidence based on observational studies for using repurposed agents has initiated clinical trials for further evaluating their efficacy, safety, optimal dosages and shorter treatment durations in various combinations with novel drugs in clinical trials. Pretomanid (PA-824) is a promising novel drug still being evaluated in phase 2 and 3 clinical trials but not yet approved for clinical use.

    Table 1 briefly summarizes evidence for using repurposed agents (linezolid and clofazimine) and major novel drugs (bedaquiline, delamanid and pretomanid) in the treatment of MDR-TB.

    Table 1. Major repurposed and novel drugs that have been used or evaluated in the treatment of MDR-TB
    Drug Evidence Precautions and practical issues
    Linezolid

    (1) in vitro activity against M. tuberculosis with a minimal inhibitory concentration (inhibiting 90% of dominant strains) of 0.5 mg/L,71 and a mutant prevention concentration (inhibiting 90% of mutants) of 1.2 mg/L,72 with good pulmonary and cerebrospinal fluid penetration as well as a low propensity to acquiring bacillary drug resistance73

    (2) Off-label use in MDR-TB with promising efficacy among patients with complicated MDR-TB patients for whom it was difficult to establish an effective fluoroquinolone-based regimen74

    (3) Linezolid 600 mg once daily added to an individualized multidrug regimen may improve bacteriological conversion and treatment success only in the most complicated MDR patients75

    (4) A systematic review of observational studies and a randomized controlled trial suggest that linezolid significantly improve sputum culture conversion and treatment success among patients with fluoroquinolone-resistant MDR- or XDR-TB76, 77

    (1) Short-term use of linezolid seldom achieves stable cure,78 but dose-dependent side effects (mitochondrial protein synthesis,79 clinically manifested as anaemia, thrombocytopenia, peripheral neuropathy and optic neuropathy74) frequently makes it necessary to prematurely terminate its use

    (2) The optimal linezolid dosing schedule remains open. Reducing linezolid dosage from 600 mg bd to 600 mg once daily,75 and from 600 mg once daily to 300 mg once daily,80 significantly reduces adverse events. Prolonged use of linezolid 300 mg once daily is still associated with a substantial risk of neuropathy,77, 81 with concerns about possibly acquiring bacillary linezolid resistance77

    (3) Intermittent dosing may help prevent or alleviate neuropathy by shortening exposure at toxic levels.78, 82 Therapeutic drug monitoring may help reduce toxicity, as adverse events are reportedly associated with mean linezolid trough >2 mg/L80

    (4) No dose adjustment is needed for patients with renal disease83

    Clofazimine

    (1) Both murine models and cohort studies have suggested that clofazimine might have sterilizing activity. A murine MDR-TB model has also demonstrated that clofazimine could significantly reduce bacillary load in lungs after 2 months, attain culture negativity after 5 months and sustain cure after 8–9 months with a 7% relapse rate.84 Clofazimine has demonstrated potential for shortening first-line TB treatment in a murine TB model,85 and MDR-TB treatment as part of the shorter MDR-TB regimen

    (2) in vitro findings from animal TB models suggested that clofazimine might mutually enhance activity of pyrazinamide, fluoroquinolones, amikacin and para-aminosalicylic acid against mycobacterial persisters86

    (1) Concerns about protracted reddish-brown skin discolouration and possible stigmatization, alongside potential for developing cross-resistance with bedaquiline and adversely prolonging the QT interval through interaction with other QT-prolonging drugs, may hamper use of clofazimine,87 notwithstanding its satisfactory track record88-90
    Bedaquiline

    (1) Bedaquiline acts on both actively replicating and dormant mycobacteria by inhibiting mycobacterial ATP synthase91

    (2) Bacillary resistance to bedaquiline occurs at predictable rates that are similar to those observed for rifampin

    (3) There is no cross-resistance between bedaquiline and other anti-TB drugs, except for clofazimine, possibly via upregulation of a multisubstrate efflux pump92

    (4) Adding bedaquiline to an optimized MDR-TB background regimen resulted in faster culture conversion in early bactericidal activity and phase 2 studies, and high rates of culture conversion in XDR-TB patients93, 94

    (5) Deferring access to bedaquiline until onset of treatment failure could be detrimental to patients and public health95

    (6) Cohort-based Markov models and cohort-based decision-analytic model have demonstrated it is cost-effective to add bedaquiline to a background MDR-TB treatment regimen96-99

    (1) There are concerns about QT interval prolongation, unexplained association with death from initial findings and potential for hepatotoxicity100

    (2) Good treatment responses and safety profiles have been suggested by case series,94, 101-103 including a few on children and adolescents.104 Although data may be inadequate for any informative systematic review of bedaquiline in the treatment of MDR-TB, one suggested that bedaquiline is better tolerated than expected with prolongation of QT interval manageable in specialized centres105

    (3) Dose adjustment is not required in case of mild-to-moderate renal impairment100

    Delamanid

    (1) Being a derivative of metronidazole and a nitroimidazopyran prodrug, delamanid inhibits mycolic acid biosynthesis, with excellent activity against intracellular M. tuberculosis106

    (2) Lack of drug–drug interactions with major antiretrovirals enables its use in HIV co-infected MDR-TB patients107

    (3) An Otsuka phase II multicentre randomized placebo-controlled trial involving 481 MDR-TB patients showed that delamanid added to an optimal background regimen significantly increased 2-month sputum culture conversion from 29.6% (placebo) to 45.4% (delamanid 100 mg bd) and 41.9% (delamanid 200 mg bd).108 Its open-label extension suggested that delamanid used for more than 6 months, in comparison with its use for less than 2 months, significantly increased favourable outcomes (cure or treatment completion) from 55% to 74.5%, and significantly reduced mortality from 8.3% to 1.0%109

    (4) Otsuka Trial 213, involving use of delamanid at 100 mg bd for the first 2 months and 200 mg once daily for the next 4 months, showed small benefit from adding delamanid to an optimized background regimen, although delamanid may help prevent amplification of drug resistance110

    (1) Although delamanid is associated with QT prolongation, the effect predictably maximizes within 8 weeks of treatment, without associated cardiac clinical manifestations, and is not materially affected by concomitant use of levofloxacin and clofazimine111

    (2) Case reports have demonstrated the safety of delamanid in the treatment of complicated MDR-TB,112-115 including one showing prolonged use for 24 months in a child with XDR-TB116

    (3) Trial 213 found no significant difference regarding treatment-emergent adverse events, drug–drug interactions between delamanid and antiretroviral drugs, and prolongation of the corrected QT interval110

    (4) With a relatively high propensity to develop bacillary drug resistance,117, 118 delamanid may be better used with potent companion agents that are less prone to develop bacillary drug resistance, for example, linezolid or bedaquiline8, 112

    Pretomanid (PA-824) (1) Being a nitroimidazole that shares the same mechanism of action with delamanid, pretomanid is bactericidal against actively replicating mycobacteria (inhibiting mycolic acid biosynthesis) and non-replicating mycobacteria (generating nitric oxide inside the tubercle bacilli)117, 119, 120

    (1) Owing to similar structure, pretomanid expectedly shares cross-resistance with delamanid as well as a relatively high propensity to acquiring bacillary drug resistance

    (2) Not yet approved for use in clinical practice

    • bd, twice daily; MDR-TB, multidrug-resistant TB; TB, tuberculosis; XDR-TB, extensively drug-resistant TB.

    A regimen containing linezolid, bedaquiline and delamanid may be adequate for the most difficult-to-treat MDR-TB, but sequential acquisition of drug resistance to bedaquiline and delamanid,19 and to linezolid and delamanid,112 has been reported. This may be anticipated, as delamanid is prone to bacillary drug resistance,117, 118 and the 120-week culture conversion rates in patients with pre-XDR- and XDR-TB given an optimized background regimen plus bedaquiline in a clinical trial setting were only 70.5% and 62.2%, respectively.121 At least three case studies have reported combined use of bedaquiline with delamanid among patients with XDR-TB,122-124 but published data may be insufficient for any informative systematic review.125

    Major clinical trials of repurposed and novel drugs

    Table 2 summarizes 11 major clinical trials of MDR-TB treatment regimens that contain repurposed agents or major novel drugs.

    Table 2. Eleven major clinical trials of MDR-TB treatment regimens involving repurposed agents, bedaquiline, delamanid or pretomanid

    Trial

    (trial identifier)

    Phase Repurposed or novel drugs Use of pyrazinamide Target population Primary objectives

    Otsuka 213

    (NCT01424670)

    3 Delamanid Yes Pulmonary MDR-TB To evaluate the efficacy of delamanid administered orally as 100 mg twice daily for 2 months followed by 200 mg once daily for 4 months versus placebo with an optimized background regimen during the 6-month intensive phase of MDR-TB treatment

    STREAM Stage 1

    (ISRCTN78372190, NCT02409290)

    3 Clofazimine Yes MDR-TB, excluding those known to be FQ-resistant or SLID-resistant To assess whether treatment closely similar to the Bangladesh regimen (gatifloxacin replaced by moxifloxacin, 600 daily if weight 33–50 kg and 800 mg daily if weight >50 kg) is as good as the WHO-recommended 18- to-24-month MDR-TB regimen

    STREAM Stage 2

    (NCT02409290)

    3

    Bedaquiline

    Clofazimine

    Yes MDR-TB or rifampin-resistant TB, excluding those known to be FQ-resistant or SLID-resistant To compare two new short-course MDR-TB regimens (one 9-month injection-free, one 6-month) with the 9-month Bangladesh regimen evaluated in STREAM Stage 1. The 9-month regimen consists of bedaquiline, clofazimine, ethambutol, levofloxacin and pyrazinamide given for 40 weeks supplemented by isoniazid and prothionamide for the first 16 weeks. The 6-month regimen consists of bedaquiline, clofazimine, levofloxacin and pyrazinamide given for 28 weeks supplemented by isoniazid and kanamycin for the first 8 weeks

    NiX-TB

    (NCT02333799)

    3

    Bedaquiline

    Pretomanid

    Linezolid

    No Pulmonary XDR-TB, or MDR-TB that is treatment intolerant or non-responsive To evaluate the efficacy, safety, tolerability and pharmacokinetics of bedaquiline plus PA-824 (pretomanid) plus linezolid given for 6 months (extended to 9 months if culture positive at month 4)

    endTB

    (NCT02754765)

    3

    Bedaquiline

    Clofazimine

    Delamanid

    Linezolid

    Yes MDR-TB with no exposure within past 5 years, or bacillary resistance, to bedaquiline, delamanid, linezolid or clofazimine

    To compare the efficacy and safety of the five (see below) new, injection-free, shortened (39-week) regimens, which contain bedaquiline (Bdq) and/or delamanid (Dlm) plus up to four companion drugs (moxifloxacin, Mfx; levofloxacin, Lfx; pyrazinamide, Z; linezolid, Lzd; clofazimine, Cfz) with a control-arm regimen that may contain Bdq or Dlm with companion drugs given in line with WHO guidelines.

    (1) Bdq + Lzd + Mfx + Z

    (2) Bdq + Lzd + Cfz + Lfx + Z

    (3) Bdq + Dlm + Lzd + Lfx + Z

    (4) Dlm + Lzd + Cfz + Lfx + Z

    (5) Dlm + Cfz + Mfx + Z

    NEXT

    (NCT02454205)

    3

    Bedaquiline

    Linezolid

    Yes MDR-TB, excluding XDR-TB, and FQ-resistant or SLID-resistant MDR-TB To compare a new injection-free, 6–9 month, regimen that contains linezolid, bedaquiline, levofloxacin, pyrazinamide and ethionamide/high-dose isoniazid with the conventional longer MDR-TB regimen

    ZeNiX

    (NCT03086486)

    3

    Bedaquiline

    Linezolid

    Pretomanid

    No Pulmonary XDR-TB, pre-XDR-TB or treatment-intolerant or non-responsive MDR-TB A follow-on trial of NiX-TB to evaluate the efficacy, safety and tolerability of various doses and durations of linezolid in 6-month regimens that also contain bedaquiline and pretomanid

    TB-PRACTECAL

    (NCT02589782)

    3

    Bedaquiline

    Clofazimine

    Linezolid

    Pretomanid

    No Pulmonary MDR-TB including XDR-TB To compare the safety and efficacy of three 24-week regimens that contain bedaquiline, pretomanid and linezolid, with or without either moxifloxacin or clofazimine, with locally accepted standard of care that is consistent with WHO recommendations

    STAND

    (NCT02342886)

    3

    Pretomanid

    Yes Drug-susceptible pulmonary TB or MDR-TB (excluding those with previous exposure to PA-824, or bacillary resistance to either FQ or pyrazinamide)

    (1) Drug-susceptible pulmonary TB: To assess the efficacy, safety and tolerability of a 26-week regimen and two 17-week regimens, which contain moxifloxacin, pyrazinamide and PA-824 (100 or 200 mg daily for the 17-week regimen, and 200 mg daily for the 26-week regimen), with the standard 6-month regimen

    (2) MDR-TB: To assess the efficacy, safety and tolerability of the 26-week regimen in comparison with the same regimen used for drug-susceptible pulmonary TB

    NC-005 (NCT02193776) 2

    Bedaquiline

    Pretomanid

    Yes Drug-susceptible pulmonary TB or MDR-TB (only those with bacillary susceptibility to moxifloxacin by molecular test)

    (1) Drug-susceptible pulmonary TB: To determine the mycobactericidal activity of two 8-week regimens:

    (a) Bedaquiline (400 mg once daily for 2 weeks, then 200 mg three times per week for 6 weeks), pretomanid 200 mg once daily and pyrazinamide 1500 mg once daily

    (b) Bedaquiline 200 mg once daily, pretomanid 200 mg once daily and pyrazinamide 1500 mg once daily

    (2) MDR-TB: To determine the mycobactericidal activity of a 8-week regimen containing bedaquiline 200 mg once daily, moxifloxacin 400 mg once daily, pretomanid 200 mg once daily and pyrazinamide 1500 mg once daily

    SimpliciTB (NCT03338621) 2

    Bedaquiline

    Pretomanid

    Yes Drug-susceptible pulmonary TB or rifampin-resistant TB or MDR-TB (excluding those with bacillary resistance to FQ)

    (1) Drug-susceptible pulmonary TB: To evaluate the efficacy, safety and tolerability of a 4-month 4-drug regimen (bedaquiline, pretomanid, moxifloxacin and pyrazinamide) using the standard 6-month regimen for comparison

    (2) Rifampin-resistant TB or MDR-TB: To evaluate the efficacy, safety and tolerability of a 6-month 4-drug regimen (bedaquiline, pretomanid, moxifloxacin and pyrazinamide)

    • FQ, fluoroquinolone; MDR-TB, multidrug-resistant TB; SLID, second-line injectable drug; TB, tuberculosis; WHO, World Health Organization; XDR-TB, extensively drug-resistant TB.

    Fluoroquinolone-resistant MDR-TB or XDR-TB is the subgroup that would benefit most from use of repurposed and novel drugs. Of the 11 trials described in Table 2, only four (endTB, NiX-TB, TB-PRACTECAL and ZeNiX) are targeted at fluoroquinolone-resistant MDR-TB or XDR-TB, and evaluating combined use of bedaquiline with either delamanid (endTB) or pretomanid (Nix-TB, ZeNiX and TB-PRACTECAL), alongside concurrent use of linezolid.

    Among repurposed and novel drugs, clofazimine, bedaquiline and pretomanid may have potential for shortening MDR-TB treatment. Pyrazinamide is potentially important in shortening treatment of pyrazinamide-susceptible MDR-TB owing to its unparalleled activity against mycobacterial persisters,126-128 and likely synergistic activity with fluoroquinolone, clofazimine and novel drugs.86, 129 Of the 11 trials described in Table 2, eight explore the feasibility of using repurposed and novel drugs, with or without moxifloxacin or pyrazinamide, to shorten MDR-TB treatment to either 6 or 9 months. Three contain bedaquiline, given with pyrazinamide and either levofloxacin or moxifloxacin (STREAM Stage 2, endTB and NEXT), with clofazimine also given in STREAM Stage 2 and endTB. One contains pretomanid, given with moxifloxacin and pyrazinamide (STAND). One contains bedaquiline and pretomanid, given with moxifloxacin and pyrazinamide (SimpliciTB). Three contain bedaquiline and pretomanid, given without pyrazinamide (NiX-TB, ZeNiX and TB-PRACTECAL), with or without either clofazimine or moxifloxacin also given in TB-PRACTECAL.

    CONCLUSIONS AND THE WAY FORWARD

    To substantially reduce TB morbidity and mortality worldwide, we need implementation and scale-up of a systematic diagnostic work-up and a tailored therapeutic approach in the national TB programmes.4-6, 56 The prognosis of MDR-TB and the halt of transmission of Mycobacterium tuberculosis strains to susceptible contacts can be improved with an early diagnosis, including the detailed assessment of the drug resistance pattern, and an immediate administration of an effective therapy.6, 56

    We believe that the most rational approach to effectively treat TB and tackle drug-resistant TB is using different regimens with different key drugs (rifamycin vs fluoroquinolone vs linezolid vs novel drugs) determined mainly by rapid DST, rather than a single regimen. In the treatment of drug-susceptible TB, we advocate optimizing use of the rifamycin-based regimen, avoiding use of fluoroquinolones as far as practicable, and using fluoroquinolones only when rapid molecular DST results suggest a low likelihood of MDR-TB. Before we use shorter rifamycin-based regimens to promote the treatment success rate of drug-susceptible TB, once-daily dosing in the continuation phase using high-dose rifapentine and moxifloxacin may help promote treatment adherence, provided that rifampin resistance has been rapidly excluded. The WHO shorter MDR-TB regimen, which critically relies on bacillary susceptibility to fluoroquinolones and second-line injectable drugs, may be used when bacillary resistance to each key drug is unlikely according to rapid DST results, drug exposure history or local epidemiology. In case of fluoroquinolone resistance, linezolid should be used, with addition of bedaquiline or delamanid or both, when the number of likely effective drugs is inadequate. Reliable rapid DST methods should be developed to guide use of novel drugs, as well as pyrazinamide.127 We hope an easily applicable combination of biomarkers that accurately predict individual treatment outcome will soon be available to ultimately enable an individualized precision medicine approach for individually tailored therapy. Finally, it is important to concurrently optimize the management of common and major TB risk factors and co-morbidities (such as smoking, alcoholism, diabetes mellitus and HIV) to enhance TB treatment response and tolerance, and reduce the risk of TB relapse.

    The Authors

    K.-C.C. is a Specialist in Respiratory Medicine and a Senior Medical Officer of Tuberculosis and Chest Service, Department of Health, Hong Kong SAR China, with research interest in the clinical management of TB. E.L.N. is Professor of Medicine and International Health, Johns Hopkins University, Baltimore, MD, USA. His research is focused on drug development for TB and other mycobacterial infections. G.S. is a Specialist in Medical Statistics and Clinical Epidemiology and Infectious Diseases and a Full Professor of Medical Statistics and Clinical Epidemiology, Department of Medical, Surgical and Experimental Medicine, University of Sassari, Italy, with clinical and epidemiological interests in the management of TB and latent TB infection. C.-C.L. is Consultant Chest Physician in charge of Tuberculosis and Chest Service and Head of Public Health Service Branch of Centre for Health Protection, Department of Health, Hong Kong, China. His research interest focuses on the epidemiology, genetics, clinical management and control of TB.

      The full text of this article hosted at iucr.org is unavailable due to technical difficulties.