Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jun 9.
Published in final edited form as: Clin Chest Med. 2019 Dec;40(4):811–827. doi: 10.1016/j.ccm.2019.08.001

New Drugs for the Treatment of Tuberculosis

Elisa H Ignatius a, Kelly E Dooley b,*
PMCID: PMC9178517  NIHMSID: NIHMS1810616  PMID: 31731986

INTRODUCTION

In 2014, tuberculosis (TB) surpassed HIV as the leading infectious cause of death. According to the World Health Organization (WHO), there were 10 million incident cases of TB in 2017 with 1.3 million deaths.1 Treatment success was 82%, a number that seems to be decreasing. This is alarming given that small changes in treatment efficacy meaningfully affect population-level incidence and mortality.2 The first-line regimen, developed in the 1950s to 1970s, remains lengthy (≥6 months) and is unforgiving to minor lapses in adherence.3 Meanwhile, in 2017, 458,000 people developed multidrug-resistant (MDR) TB, Mycobacterium tuberculosis (M.tb.) resistant to isoniazid and rifampicin. Despite efforts to extend access to treatment, the prognosis for patients with MDR-TB remains poor, with only 55% treatment success.1 Treatment outcomes for patients with extensively drug-resistant (XDR) TB (MDR-TB that is also resistant to fluoroquinolones and injectable agents) are exceptionally poor, and now totally drug-resistant (TDR)-TB is documented, ushering us back to the preantibiotic era.4 In a study of patients in South Africa with “programmatically incurable tuberculosis,” many were discharged to the community once treatment options were exhausted, and community-level transmission occurred. Despite treatment failure, 90% of their sputum isolates were sensitive to newer drugs, namely linezolid, BDQ, and delamanid.5

There have been recent WHO updates to guidance for MDR-TB treatment.6 A short-duration regimen, commonly referred to as the Bangladesh regimen, yielded promising results for a 9- to 12-month course,7 and subsequent cohort studies and a phase 3 clinical trial, STREAM, confirmed these impressive results.8 This “short-course” MDR-TB regimen still requires use of 7 drugs and is offered as a complete regimen, with no allowance for substitutions or deletions. Thus, the application of this regimen in some settings may be limited by drug-resistance patterns.9

Fortunately, the second wave of antituberculosis drug development, which included the diarylquinoline, BDQ, and 2 nitroimidazoles, delamanid and pretomanid, offers the potential for simple, shorter, all-oral regimens for MDR- and XDR-TB treatment. In vitro, animal, and early clinical experience with these 3 compounds has been promising, and trials are ongoing to determine the best multidrug combinations, including among children and persons living with HIV (PLWH). Emerging results from the Otsuka phase 3 randomized controlled trial (RCT) of delamanid, the short-course MDR-TB regimen (STREAM Stage 1), and pediatric pharmacokinetic (PK) and safety studies of BDQ and delamanid, led the WHO to release a rapid communication in August 2018 with updated guidance (Table 1).10 Standard-duration treatment of MDR-TB now should include fluoroquinolone, BDQ, and linezolid plus clofazimine or cycloserine, with additional drugs such as delamanid used to comprise a 4- to 5-drug regimen.

Table 1.

Standard-duration therapy for MDR-TB recommended by the WHO

Group Drug
Group A: Include all 3 Levofloxacin (Lfx) OR moxifloxacin (Mfx, M)
Bedaquiline (BDQ, B)
Linezolid (LZD)
Group B: Add one or both medicines Clofazamine (CFZ)
Cycloserine (Cs) OR terizidone (Trd)
Group C: To complete regimen
OR Group A or B cannot be used
Ethambutol (Emb, E) Delamanid (DLM, D)
Pyrazinamide (PZA, or Z)
Imipenem-cilastatin (Ipm/Cln) OR meropenem (Mpm)a
Amikacin (Am) OR streptomycin (S)
Ethionamide (Eto) OR prothionamide (Pto)
p-Aminosalicylic acid (PAS)
a

With amoxicillin-clavulanic acid.

From World Health Organization (WHO). WHO Technical report on critical concentrations for TB drug susceptibility testing of medicines used in the treatment of drug-resistant TB. Geneva; 2019. Available at: https://apps.who.int/iris/bitstream/handle/10665/311389/9789241550529-eng.pdf?ua=1

This article outlines the history, mechanisms of action and resistance, preclinical studies, pharmacology, clinical evaluation, and treatment niche for new drugs for TB treatment that are licensed or in late clinical testing: BDQ, delamanid, and pretomanid. Also provided is a short summary of promising new chemical entities in the “third wave” of mycobacterial drug development, those that are in earlier phases of clinical testing but show great promise for shorter or less toxic regimens.

NEW DRUGS IN CLINICAL USE OR LATE-PHASE CLINICAL TESTING

BDQ

BDQ is a diarylquinoline that blocks the proton pump of adenosine triphosphate (ATP) synthase. It was granted approval for MDR-TB at 400 mg daily for 14 days followed by 200 mg thrice weekly for 22 weeks by the US Food and Drug Administration (FDA) in 2012 and by the European Medicines Agency (EMA) in 2013, making it the first drug approved for the treatment of TB in 40 years. Shortly thereafter, the WHO released interim guidance that BDQ could be used in combination with second-line drugs to treat pulmonary MDR-TB.11

Preclinical data

BDQ is bactericidal against M.tb. and bacteriostatic against other nontuberculous mycobacteria.12,13 In mice, BDQ has sterilizing activity superior to that of rifampicin and shortens treatment duration required for cure in multidrug regimens.14,15 BDQ-pyrazinamide (BZ) has superior activity to rifampin-pyrazinamide (RZ).16,17 Relapse rates after 3 months of BDQ-pyrazinamide plusrifapentine (BZP) or moxifloxacin (BZM) were similar to 6 months of standard treatment.18 Pyrazinamide-free regimens, BPM or B-pretomanid (Pa)-M (BPaM), given for 4 months showed lower relapse rate than standard 6-month treatment.16 BPaMZ cured mice in 2 months.19

Clinical pharmacology

BDQ is well absorbed, and food increases bioavailability 2-fold.20 BDQ is highly protein bound (99.9%) and has a large volume of distribution (Vd), greater than 10,000 L.21 BDQ is primarily metabolized by cytochrome P450 (CYP) 3A4, forming a metabolically active M2 metabolite. The terminal half-life of BDQ and M2 is long, ~160 days.22 BDQ’s bactericidal activity is concentration dependent.23 BDQ exposures are lower in patients of Black race or with low albumin.24,25 Rifampicin and rifapentine reduce BDQ concentrations substantially (70%–80%),26 as does EFV (50%).27 BDQ can be given with nevirapine without dose adjustment. Ritonavir-boosted lopinavir decreases BDQ clearance by 75%, so coadministration requires caution and ECG monitoring.28

Resistance

Spontaneous resistance mutations to BDQ occur at rates comparable with rifampicin with no loss of fitness.29 Given BDQ’s unique target, cross-resistance was not anticipated, but a study of clofazimine-resistant isolates found coexisting BDQ resistance via upregulation of the efflux pump MmpS5-MmpL5, owing to mutations in the transcriptional regulatory Rv0678.30 Clinical and murine studies have implicated Rv0678 and Rv2535c.31,32 Although BDQ resistance generally confers cross-resistance to clofazimine, the opposite is not universally true.33 Critical concentrations of 0.25 μg/mL on 7H11 and 1 μg/mL on MGIT have been proposed.34

Clinical data

In humans, BDQ monotherapy has delayed early bactericidal activity (EBA), likely because of its mechanism of shifting ATP use to alternative pathways such as glycolysis, leading to eventual (not immediate) cell death.35-37 In a phase 2b RCT among 160 patients with pulmonary MDR-TB receiving multidrug background regimen (MBR), time to culture conversion was 83 days with BDQ versus 125 days with placebo. Twenty-four-week culture conversion was higher (79% vs 58%), as was cure (58% vs 32%). The BDQ arm had greater QTc prolongation and mortality, although deaths were late and of variable causes.38 A subsequent single-arm trial of 233 patients confirmed the efficacy of 24 weeks of BDQ.39 Multiple trials involving BDQ as part of all-oral or treatment-shortening regimens are currently under way in both drug-sensitive and drug-resistant TB (Table 2).

Table 2.

Recent, ongoing, or upcoming trials involving BDQ, delamanid, or pretomanid

Trial Status Population Sample Description
Diarylquinoline
BDQ
 Janssen C211 (NCT02354014) Phase 1–2 Enrolling India, Philippines, Russian Federation, South Africa 60 Pediatric/adolescent PK, safety, tolerability of BDQ + background regimen
 IMPAACT P1108 (NCT02906007) Phase 1–2 Enrolling Haiti, India, South Africa 72 Pediatric PK, safety, tolerability of 24 wk BDQ + BR for MDR-TB in HIV-infected/uninfected children
 STREAM stage 2 (NCT02409290) Phase 3 Enrolling China, Ethiopia, Georgia, India, Indonesia, Republic of Moldova, Mongolia, South Africa, Uganda, Vietnam 1155 Locally used WHO approved MDR regimen OR 40 wk (Bangladesh): MXF-CFZ-EMB-PZA + INH-PTO-KAN (1st 16 wk) OR 40 wk (all-oral): CFZ-EMB-PZA-LFX-BDQ + INH-PTO (1st 16 wk) OR 28 wk CFZ-PZA-LFX-BDQ + INH-KAN (1st 8 wk)
 TRUNCATE-TB (NCT03474198) Phase 3 Enrolling Philippines, Singapore, Thailand 900 Multiarm, multistage (MAMS) trial of ultrashort regimens in drug-sensitive TB: 8 wk RHZE, then 16 wk RH OR 8–12 wk RHZE-Lzd OR 8–12 wk RHZE-CFZ OR 8–12 wk RPT-HZ-LZD-LFX OR 8–12 wk HZE-LZD-BDQ
 NExT (NCT02454205) Phase 3 Terminated South Africa 300 6–8 mo KAN-MXF-PZA-ETH-TRD, then 18 mo MXF-PZA-ETH-TRD OR 6–9 mo LZD-BDQ-LFX-PZA-TRD-ETH/INH
Nitroimidazoles
Delamanid
 Otsuka 213 (NCT01424670) Phase 3 Complete Estonia, Latvia, Lithuania, Moldova, Peru, Philippines, South Africa 511 DLM or placebo × 6 mo + BR × 18–24 mo
 Otsuka 232 (NCT01856634) Phase 1 Complete Philippines, South Africa 37 Pediatric PK, safety of DLM + BR for 10 d DLM 100 or 50 mg; pediatric formulation DLM 25, 10, or 5 mg; or optimized BR
 Otsuka 233 (NCT01859923) Phase 2 Complete Philippines, South Africa 37 Pediatric 6-mo tolerability, PK/PD, efficacy various doses DLM + BR (follow-up 232)
 MDR-END (NCT02619994) Phase 2 Enrolling Republic of Korea 238 9–12 mo DLM-LZD-LFX-PZA vs 20–24 mo MDR-TB standard of care
 IMPAACT 2005 (NCT03141060) Phase 1–2 Enrolling Botswana, India, South Africa, Tanzania 48 PK, safety, tolerability of DLM + BR for HIV-infected/noninfected children with MDR-TB 24 wk DLM + BR continued
 ACTG A5356 Phase 2a In development ACTG sites globally 120 24 wk DLM-LZD 600 mg daily + BR OR 24 wk DLM-LIN 600 + BR OR 24 wk DLM-LZD 1200 every other day
 ACTG 5300/IMPAACT 2003 (PHOENIx) Phase 3 Enrolling Treatment of adult and child contacts of MDR cases: 26 wk DLM OR 26 wk INH
Pretomanid
 NC-006 STAND (NCT02342886) Phase 3 Terminated Georgia, Kenya, Malaysia, Philippines, South Africa, Tanzania, Uganda, Zambia 1500 24 wk PaMZ (MDR or DS-TB) OR 17 wk PaMZ (DS-TB) OR 24 wk RHZE (DS-TB)
 APT (NCT02256696) Phase 2 Enrolling South Africa 183 8 wk Pa-rifabutin-INH-PZA, then 4 wk Pa-Rb-INH OR 8 wk Pa-RIF-INH-PZA, then 4 wk Pa-RIF-INH OR 8 wk Pa-Rb-INH-PZA, then 4 wk Pa-Rb-INH OR 8 wk RHZE, then 4 wk RH
Combinations of new drugs
 NixTB (NCT02333799) Phase 3 Complete South Africa 109 6–9 mo BPaLzd for XDR-TB and nonresponsive MDR-TB
 DELIBERATE (ACTG 5343) (NCT02583048) Phase 2 Complete Peru, South Africa 84 Cardiac (QT) safety of BDQ vs DLM vs BDQ plus DLM, with BR in MDR-TB × 24 wk
 NC-005 (NCT02193776) Phase 2b Complete South Africa, Tanzania, Uganda 240 Drug-sensitive: 8 wk BPaZ (with or without BDQ loading dose) OR 4HRZE/2RH Drug-resistant: 8 wk B-Pa-MXF-PZA
 TB-PRACTECAL (NCT02589782) Phase 2–3 Enrolling Belarus, South Africa, Uzbekistan 630 Short-course treatment, adaptive design 24 wk BDQ-Pa-LZD-MXF OR 24 wk BDQ-Pa-LZD-CFZ OR 24 wk BDQ-Pa-LZD OR local standard of care
 endTB (NCT02754765) Phase 3 Enrolling Georgia, Kazakhstan, Kyrgyzstan, Lesotho, Peru, South Africa 750 Adaptive design 39 wk BDQ-LZD-MXF-PZA OR 39 wk BDQ-LZD-CFZ-LFX-PZA OR 39 wk BQD-DLM-LZD-LFX-PZA OR 39 wk DLM-LZD-CFZ-LFX-PZA OR 39 wk DLM-CFZ-MXF-PZA OR 86 wk local standard of care
 ZeNiX (NCT03086486) Phase 3 Enrolling Georgia, Republic of Moldova, Russian Federation, South Africa 180 26 wk tx for XDR or nonresponsive MDR-TB, duration/dose Lzd LZD 1200 mg × 26 wk-Pa-BDQ OR LZD 1200 mg × 9 wk-Pa-BDQ OR LZD 600 mg × 26 wk-Pa-BDQ OR LZD 600 mg × 9 wk-Pa-BDQ
 SimpliciTB (NCT03338621) Phase 2–3 Enrolling Georgia 450 Drug-sensitive: 17 wk BDQ-Pa-MXF-PZA OR 26 wk standard (2HRZE/4HR) Drug-resistant: 26 wk BDQ-Pa-MXF-PZA
 IMPAACT 2020 Phase 2 In development IMPAACT international sites 148 Children with MDR-TB 8 wk LZD + 24 wk BDQ-DMN-LFX/FLO
 BEAT TB Phase 3 In development BDQ-DLM-LZD-CF-LFX for 6 mo treatment of MDR-TB

Abbreviations: BR, background regimen; CFZ, clofazimine; DLM/D, delamanid; DS, drug-sensitive; EMB, ethambutol; ETH, ethionamide; INH, isoniazid; KAN, kanamycin; LFX, levofloxacin; LZD/L, linezolid; MXF, moxifloxacin; Pa, pretomanid; PTO, prothionamide; PZA, pyrazinamide; Rb, rifabutin; RIF, rifampin; TRD, terizidone.

Reassuringly, large observational cohort studies, including a meta-analysis of 12,030 patients with MDR-TB and studies among PLWH, have shown that BDQ decreases rather than increases mortality.6,40,41 Although QT prolongation of 15 to 20 milliseconds is common, there have been no reports of clinically important cardiac toxicity, even with therapy up to 12 months.42,43 Although most data are for BDQ added to MBR, it also confers benefit when substituted for injectables.44

Delamanid

Delamanid (OPC-67683, DLM) is a nitroimidazole that inhibits synthesis of ketomycolate, a cell wall lipid that constitutes one-third of M.tb’s dry weight.45 Delamanid was approved by EMA in 2014 at a dose of 100 mg twice daily for 6 months and is recommended by WHO for MDR-TB in certain circumstances. It is approved in Europe but is not yet FDA approved.

Preclinical data

DLM has an extremely low minimal inhibitory concentration (MIC) against M.tb. (0.006–0.024 μg/mL) and displays activity against replicating and dormant bacilli.46 Animal studies are limited, although a guinea pig model of cavitary disease demonstrated activity.47 In a mouse model, DLM-RZ had similar treatment efficacy at 4 months as RZHE for 6 months.46

Clinical pharmacology

Delamanid’s absorption is enhanced by food and may be reduced by concomitant dosing with other medications.48 It displays less-than-proportional PK.49 Delamanid is catalyzed to DM-6705 by a pathway that involves albumin.50 DLM is not a substrate, inhibitor, or inducer of CYP enzymes. Whereas the half-life of the parent drug is 38 hours, DM-6705 has a terminal half-life of 121 to 322 hours.51 There were no clinically significant interactions when DLM was coadministered with efavirenz, lopinavir/ritonavir, or tenofovir.48

Resistance

Both delamanid and pretomanid (see later discussion) require activation involving coenzyme F420. Resistance mutations have been identified in fbiA, fbiB, and fbiC (involved in F420 biosynthesis) and fgd1 and ddn (prodrug activation). The F420 cofactor is not present in mammalian cells, explaining the narrow spectrum of activity.52 There are limited data on treatment-induced resistance, but case reports describe emergence of fgd1, fbiA, fbiB, and fbiC mutants.33,53 A break point of 0.016 μg/mL on 7H11 and 0.06 μg/mL on MGIT is proposed.34

Clinical data

Phase 2a EBA trials demonstrated measurable but overlapping activity of different doses of delamanid.51 In a phase 2b RCT in 481 patients with MDR-TB receiving MBR, delamanid 100 mg or 200 mg twice daily given for 8 weeks had higher culture conversion at 2 months (45% and 42%) than placebo (30%).49 Modest QT prolongation was seen. In a nonrandomized follow-on study, longer delamanid use was associated with favorable outcomes (74.5% vs 55%) and decreased mortality (1.0% vs 8.3%).54 In the phase 3 RCT of delamanid versus placebo added to MBR, overall treatment success at 30 months was high in both arms (>80%), so a statistically significant difference was not detected.55 Nonetheless, given the measurable activity of the drug in the phase 2 program coupled with the drug’s excellent safety profile, the WHO includes DLM among drugs that can be used for MDR-TB. Simplified regimens including delamanid are under study (see Table 2). Though marketed for use for 6 months, it has been well tolerated for up to 20 months.56,57

Pretomanid

Like delamanid, pretomanid (Pa) (PA-824) is a nitroimidazole that inhibits mycolic acid synthesis in actively multiplying bacilli. In addition, when it is activated by the M.tb.-specific F420-dependent nitroreductase, toxic reactive nitrogen species are released, killing nonreplicating bacilli.58,59 A new drug application to the FDA was approved in 2019.

Preclinical data

Pretomanid’s MIC against M.tb. is between 0.015 and 0.25 μg/mL for drug-sensitive strains and 0.03 to 0.53 μg/mL for drug-resistant strains.60 Pretomanid is active against “persisters”61 and under oxygen-poor conditions.60 Mouse models have revealed time above MIC to be the PK driver for nitroimidazoles.62 PaMZ in mice reduced treatment time by 1 to 2 months compared with standard therapy,63 and the addition of BDQ (BPaMZ) cured mice in 2 months.19

Clinical pharmacology

Pretomanid has a half-life of 16 to 20 hours, is 94% protein bound, and has a large Vd64,65; PK is dose proportional up to 200 mg, then less-than-dose-proportional. CYP3A is responsible for 20% of its metabolism.66 In a healthy volunteer trial, efavirenz and rifampicin decreased pretomanid trough concentrations by 46% and 85% and exposures (area under the curve) by 35% and 66%, respectively, raising concerns for coadministration.66 Lopinavir/ritonavir did not affect pretomanid exposures meaningfully.

Resistance

Like delamanid, resistance to pretomanid can be related to prodrug activation (fgd1, ddn) or the F420 biosynthetic pathway (fbiA, fbiB, fbiC).52 Resistance is present in 1 in 105 organisms pre-treatment. Mutations in Rv2983 in mouse models disrupt F420 biosynthesis, a newly identified mechanism of resistance yet to be identified in clinical samples.67

Clinical data

In the first EBA study of pretomanid, doses of 200 to 1200 mg produced similar, measurable activity.68 In a second EBA, 50 mg was less active than 100 to 200 mg. Pretomanid causes dose-dependent increases in creatinine and modest QT prolongation.69 In an EBA study of multidrug regimens, BPaZ had the highest microbiological activity, and 3 patients receiving regimens containing BPa had grade 3 or 4 transaminase elevations.70 An 8-week study of PaMZ in drug-sensitive TB showed higher 8-week culture conversion (66%–71%) than HRZE (38%).71 A phase 3 RCT (STAND) opened in 2015 to investigate PaMZ, but was put on temporary clinical hold to investigate cases of fatal hepatitis. In the mean-time, a different trial showed exceptionally high culture conversion rates in patients with MDR-TB receiving BPaMZ,72 and for this reason the SimpliciTB phase 3 RCT was launched to explore this regimen. Remarkably, in patients with XDR-TB, thought nearly untreatable, a regimen of BDQ-pretomanid-linezolid (BPaLz) tested in the NixTB trial showed favorable outcomes in ~90% of patients. Duration-dependent myelosuppression and neuropathy from linezolid are common; therefore, ZeNix was started to evaluate optimal linezolid dose and duration in BPaLZ regimens.73

PRACTICAL ASPECTS OF TREATMENT WITH NEW DRUGS

Coadministration

There is mounting evidence to support concurrent use of BDQ and a nitroimidazole, despite initial concerns for QT prolongation risk. Medecins Sans Frontières has endorsed judicious use of BDQ and delamanid since 2016 and has reported excellent outcomes (74% conversion by 6 months) and infrequent QT prolongation in a small cohort.74 Other small series have reported similar efficacy results,75-77 with episodes of QTc interval greater than 500 milliseconds generally occurring only with concurrent moxifloxacin or clofazimine.42 Interim results from DELIBERATE, an AIDS Clinical Trials Group RCT, suggest no more than additive QT prolongation when BDQ and delamanid are combined. If BDQ and delamanid will be coadministered, considerations include: (1) repletion of K+ and Mg2+ electrolytes; (2) measuring QTcF at baseline and serially; and (3) close attention to companion drugs (fluoroquinolones, clofazimine, ritonavir, methadone, and aminoglycosides all prolong QT). Pretomanid has a modest effect on the QT interval, and with moxifloxacin did not cause QTc greater than 500 milliseconds.71 QTcF was prolonged by ~20 milliseconds in patients receiving BPaZ or BPaMZ, which is only clinically significant if baseline QTcF is high.78 Although the WHO recommends BDQ in the standard (18–24 months) duration regimen, it is licensed only for 24 weeks; guidance regarding prolonged use is awaited.

Extrapulmonary Tuberculosis

There are no data for pleural or pericardial TB and only limited data for central nervous system (CNS) TB. In one patient with MDR-TB meningitis (TBM), BDQ levels in cerebrospinal fluid (CSF) were undetectable, but collection conditions were not optimized for measurement of this highly lipophilic drug.79,80 A study in rats using radiolabeled delamanid showed lasting distribution into the brain.81 Recent work in a rabbit model of TBM showed high brain but low CSF levels. Three patients with XDR-TBM, which is generally fatal, who were treated with delamanid containing regimens displayed marked clinical improvement despite very low delamanid CSF/plasma ratios.82 In rats, pretomanid seems to cross the blood-brain barrier and penetrate into the brain, although clinical correlation is needed.83

Children and Pregnant Women

Initial results from Otsuka trials 232/233 demonstrated acceptable PK and safety of delamanid in HIV-uninfected children ≥3 years old; therefore, the WHO now recommends DLM for that age group. The WHO recommends BDQ in children with MDR-TB aged 6 to 17 years.84 Trials are ongoing to optimize pediatric dosing (BDQ: Janssen C211, IMPAACT P1008; delamanid: Otsuka 233, 234, IMPAACT 2005). There is not yet a pediatric formulation of BDQ, but suspension or crushing gives similar bioavailability to whole tablets.85 Dispersible formulations of BDQ and delamanid are in development. Safety and efficacy of these drugs in children seem similar to those in adults, although data are limited.86-88 The pediatric investigation plan for pretomanid is progressing. There exist little data about use of these drugs in pregnancy89; given that these drugs are life-saving in MDR-TB, PK and safety studies in this population are needed.

THE DRUG DEVELOPMENT PIPELINE

The “third wave” of TB drug development is now here (Table 3). Some compounds are from drug classes already in clinical use for TB but have been redesigned to optimize bioavailability, potency, safety, or activity against resistant strains (fluoroquinolones, diarylquinolines, riminophenazines, carbapenems, oxazolidinones, nitroimidazoles). Many are from new classes with novel mechanisms of action (eg, inhibitors of DprE1, leucyl-tRNA synthetase, cholesterol catabolism). Many of these compounds are now in phase 1 or phase 2 trials.90-117

Table 3.

New chemical entities in development for treatment of tuberculosis

Name Developer Supporting Data Preclinical/Clinical Trial Objectives Trial Design
aEthylenediamine: targets Mmpl3 involved in mycolate transport and processing, disrupting incorporation into cell wall
SQ109 Sequella, Inc Phase 2 EBA/safety with gastrointestinal upset, no QT prolongation, significant decline in exposure when administered at 150 mg with rifampin (overcome at 300 mg SQ109)
Decreases RIF MIC; No EBA as monotherapy or additive to others; No difference in culture conversion rate
Patient with INH resistance on RHZ-SQ109 developed RIF/PZA resistance
SQ109 EBA NCT01218217 (2a, published) EBA, safety, tolerability, PK of several doses with/without RIF 14 d: SQ109 75, 150, 300 as monotherapy OR SQ109 15, 300 + RIF OR RIF monotherapy
MAMS-TB, NCT01785186 (2b, published) MAMS trial to evaluate 4 regimens 14 wk INH-RIF35-PZA-EMB OR INH-RIF10-PZA-Q300 OR INH-RIF20-PZA-Q300 OR INH-RIF20-PZA-MXF OR INH-RIF10-PZA-EMB
(2b–3, complete) Additive for MDR-TB in Russia HRZE-SQ109 OR HRZE-placebo
Imidazopyridine amide: inhibits qcrB subunit of cytochrome bc1 complex
Q203, Telacebec Qurient Co. Ltd./LLC "Infectex3 Mouse models demonstrate efficacy with once daily doses <1 mg/kg; active intracellularly and extracellularly, no CYPp450 inhibition in vitro NCT02530710 (1a, complete) Safety, tolerability, PK of single doses in healthy adults Doses of 10, 20, 50, 100, 150, 200, 400, 800 mg/kg
NCT02858973 (1b, complete) Safety, tolerability, PK of multiple doses in adults Q203 vs placebo
NCT03563599 (2, enrolling) EBA of Q203 vs RHZE Mid, high, low doses of Q203
GSK 070, GSK3036656 GlaxoSmithKline Good oral bioavailability, low protein binding, activity comparable with INH in mice; Active at lower doses and wider intervals (48 h) than linezolid NCT03557281 (2, pending) EBA, safety, tolerability in 4 sequential cohorts with DS-TB 2 wk GSK3036656 or RHZE
NCT03075410 (1, complete) Safety, tolerability, PK of single ascending and repeat doses in health adults Single dose at 5, 25, or 100 mg
Daily dosing
Oxazolidinone: protein synthesis inhibitor, binds bacterial 23s rRNA of 50S subunit to prevent formation of 70S initiation complex
Sutezolid, PNU-100480 Sequella, Inc, TB Alliance In mice, shortens standard treatment by 1 mo, activity superior to linezolid
Safety and tolerability up to 1000 mg in humans
Highly active primary metabolite (PNU-101603) at higher concentration than parent
Phase 2 data: synergy with PZA, tmax 1–2 h (fasting), 3 h (fed), half-life 3.4 h, 600 mg BID consistently higher than MIC (1200 mg was not)
No heme/neuro AE, 14% patients with ALT elevation
NCT01225640 (2, complete) EBA and whole blood activity STZ 600 mg BID, 1200 mg daily or RHZE
SUDOCU (2, proposed) Sutezolid EBA and dose finding study BDQ-DLM-MXF PLUS STZ 0 mg 600 mg daily, 1200 mg daily, 600 mg twice daily, OR 800 mg twice daily
STEP (2c, proposed) Safety, efficacy, exposure-response of rifampin doses, STZ 4 mo RhighHZE OR 4 mo RhighHZhighE OR 4–6 mo STZ-BDM OR 6 mo HRZE
NCT03237182 (4, enrolling) Gene-derived individualized drug-resistant TB regimens, including sutezolid
Delpazolid, LCB01–0371 LegoChem Biosciences, Inc Phase 1: tolerated to 1200 mg twice daily (though decline in heme values for daily doses >800 mg), rapid PO absorption, 100% oral bioavailability, tmax 0.5-1 hr, half-life 1.3–2.1 h, plasma binding 37% NCT02836483 (2, enrolling) EBA, safety, and PK 800 mg daily, 400 mg twice daily, 800 mg twice daily, LZD 600 mg twice daily
Contezolid, MRX-4 MicuRx Pharmaceuticals, Inc Broad gram-positive activity, comparable with linezolid; MRX-1 in phase 3 trials in China for skin/soft tissue, MRX-4 (prodrug of MRX-1) being studied in USA NCT03033329 (1, complete) Safety, tolerability, PK with dose escalation (IV), crossover to PO
  • Single IV dose 150–1800 mg

  • BID IV dosing for 10 d: 600, 900, 1200, 1500 mg

TBI-223 TB Alliance, Institute of Materia Medica Lower activity on mammalian mitochondrial protein synthesis; hepatocyte stability across 5 animal species, no evidence in vitro of CYP induction, half-life 3 h in mice, 8 h in rats; success in mice with BPa-TBI223 (replace linezolid); no hematologic/marrow toxicity in 14 and 28 d rat toxicity studies NCT03758612 (1, pending) Single and multiple ascending dose trial
aDprE1 inhibitors: covalent (benzothiazinone): inhibits decaprenyl-phosphoribose epimerase (DprE1) involved in cell wall arabinan biosynthesis
BTZ043 University of Munich, Hans-Knöll Institute, Jena, German Center for Infection Research (DZIF) Superior to INH at 2 mo in mice (6 mo pending)
No antagonism with existing drugs, apparent synergy in vivo with BDQ-RIF
Low-level CYP450 interaction
NCT03590600 (1, enrolling) Ascending dose study BTZ043 125, 250, 500, 1000, 2000 mg or placebo
Macozinone, MCZ, PBTZ169 iM4TB-Innovative Medicines for Tuberculosis, Bill & Melinda Gates Foundation, Nearmedic Plus LLC Highly active against replicating bacteria; no antagonism with RHZE, synergy in vitro with BDQ, CFZ, DLM, sutezolid
Prior formulation with good tolerability, bactericidal activity against DS-TB at 640 mg
NCT03776500 (1, pending) Safety, tolerability, PK at multiple ascending doses 150 mg BID OR 300 mg BID OR 600 mg daily OR 600 mg BID
aDprE1 inhibitor: non-covalent (OPC-167832: carbostyril, TBA-7371: azaindole): see above
OPC-167832 Otsuka Pharmaceutical Development & Commercialization, Inc Activity against replicating and dormant intracellular bacilli; active in acute and chronic murine models; no antagonism with other TB drugs; additive effect with DLM exceeding RHZE NCT03678688 (1–2, enrolling) Safety, tolerability, PK, efficacy of multiple oral doses in DS-TB Stage 1: 14 d of OPC-167832 10 mg OR 30 mg OR 90 mg OR 270 mg OR RHZE
Stage 2: 14 d of OPC-167832LowDLM200 OR OPC-167832HighDLM200 OR DLM200
TBA-7371 TB Alliance Efficacy in vitro and in mice Phase 1 trial complete on food effect, optimal dose, DDI, PK, PD as single dose or multiple doses NCT03199339 (1, complete) Safety, tolerability of single and multiple doses among healthy adults Single doses of 100, 250, 500 1000, 1500 mg; Multiple doses of 100, 200, 400; DDI with midazolam, bupropion
Riminophenazine: Same class as CFZ
TBI-166 Institute of Materia Medica, CAMS & PUMC Excellent in vivo, shorter half-life, less lipophilicity, less potential for skin discoloration and other toxicities of CFZ Unknown name (1, enrolling) Safety, tolerability among patients in China

Abbreviations: EBA, early bactericidal activity; RIF or R, rifampin/rifampicin; MIC, minimal inhibitory concentration; PZA or Z, pyrazinamide; PK, pharmacokinetics; INH or H, isoniazid; EMB or E, ethambutol; Q, SQ109; STZ, sutezolid; BDQ or B, bedaquiline; DLM or D, delamanid; MXF or M, moxifloxacin; LZD, linezolid; PO, per oral; Pa, pretomanid; CYP, cytochrome p450; d, day; mg, milligram; DDI, drug-drug interaction; BID, twice daily; PD, pharmacodynamic; CFZ, clofazamine.

a

Denotes novel class.

Adapted from Working Group on New TB Drugs (https://www.newtbdrugs.org/pipeline/clinical) as of January 2019.

SUMMARY

BDQ is proven to reduce mortality in MDR-TB, and it is recommended for use as a first-line agent for patients with MDR-TB. Best practices for ECG monitoring, dosing in children, and HIV cotreatment are emerging. Delamanid, despite disappointing phase 3 results, has a role as a companion drug in all-oral regimens for MDR-TB. It is well tolerated, and rational dosing recommendations and formulations for children are coming soon. Pretomanid, in combination with linezolid and BDQ, displayed exceptional efficacy among patients with XDR-TB, and it is now approved for use in combination with LZD and BDQ in patients with highly resistant TB. In addition, nitroimidazoles may have brain penetration that allows for use in CNS TB, but clinical data are lacking. Now that the WHO has listed BDQ and delamanid among the options for MDR-TB in the newest guidelines, access to these drugs will improve. Lastly, the third wave of TB drug discovery offers exciting new compounds with numerous mechanisms of action that may expand the arsenal not only for MDR-TB but for ultrashort combination regimens for drug-sensitive TB.

KEY POINTS.

  • Treatment success rates for drug-susceptible tuberculosis are too low, and prognosis for drug-resistant TB remains poor.

  • Though shorter 9- to 12-month regimens are available for the treatment of MDR-TB, therapy is complex and often toxic.

  • The diarylquinoline, bedaquiline (BDQ), and the nitroimidazoles delamanid and pretomanid, have excellent preclinical and clinical data to support their use for MDR or XDR-TB. Multiple trials are centered on use of these drugs to produce well-tolerated, all-oral, short-course regimens.

  • Further study of new drugs is urgently needed among children, pregnant women, and people living with HIV to guide clinicians.

  • The pipeline now contains numerous new chemical entities from 16 drug classes with encouraging results from late preclinical or early clinical testing, the “third wave” of TB drug development.

Disclosure Statement:

E.H. Ignatius has no conflicts of interest to report. K.E. Dooley is principal investigator or protocol chair for trials involving BDQ, delamanid, pretomanid, rifapentine, levofloxacin, high-dose isoniazid, and meropenem. Support and funding for these trials are provided by the NIH or FDA. She receives no salary support from drug companies for these studies. This work was supported by the National Institutes of Health T32-AI007291-27 to E.H. Ignatius. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. K.E. Dooley received no support for her work on this article.

REFERENCES

  • 1.World Health Organization (WHO). Global tuberculosis report 2018. WHO; 2018. Geneva (Switzerland). [Google Scholar]
  • 2.Kendall EA, Shrestha S, Cohen T, et al. Priority-setting for novel drug regimens to treat tuberculosis: an epidemiologic model. PLoS Med 2017; 14(1):e1002202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Imperial MZ, Nahid P, Phillips PPJ, et al. A patient-level pooled analysis of treatment-shortening regimens for drug-susceptible pulmonary tuberculosis. Nat Med 2018;24(11):1708–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Slomski A. South Africa warns of emergence of “totally” drug-resistant tuberculosis. JAMA 2013; 309(11):1097–8. [DOI] [PubMed] [Google Scholar]
  • 5.Dheda K, Limberis JD, Pietersen E, et al. Outcomes, infectiousness, and transmission dynamics of patients with extensively drug-resistant tuberculosis and home-discharged patients with programmatically incurable tuberculosis: a prospective cohort study. Lancet Respir Med 2017; 5(4):269–81. [DOI] [PubMed] [Google Scholar]
  • 6.Ahmad N, Ahuja SD, Akkerman OW, et al. Treatment correlates of successful outcomes in pulmonary multidrug-resistant tuberculosis: an individual patient data meta-analysis. Lancet 2018; 392(10150):821–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Van Deun A, Maug AKJ, Salim MAH, et al. Short, highly effective, and inexpensive standardized treatment of multidrug-resistant tuberculosis. Am J Respir Crit Care Med 2010;182(5):684–92. [DOI] [PubMed] [Google Scholar]
  • 8.Trébucq A, Schwoebel V, Kashongwe Z, et al. Treatment outcome with a short multidrug-resistant tuberculosis regimen in nine African countries. Int J Tuberc Lung Dis 2018;22(1):17–25. [DOI] [PubMed] [Google Scholar]
  • 9.Sotgiu G, Tiberi S, D’Ambrosio L, et al. Faster for less: the new “shorter” regimen for multidrug-resistant tuberculosis. Eur Respir J 2016;48(5): 1503–7. [DOI] [PubMed] [Google Scholar]
  • 10.World Health Organization. Rapid communication : key changes to treatment of multidrug- and rifampicin-resistant tuberculosis (MDR/RR-TB). Geneva (Switzerland): WHO; 2018. [Google Scholar]
  • 11.World Health Organization (WHO). The use of bedaquiline in the treatment of multidrug-resistant tuberculosis: interim policy guidance. Geneva (Switzerland): World Health Organization; 2013. [PubMed] [Google Scholar]
  • 12.Andries K, Verhasselt P, Guillemont J, et al. A diarylquinoline drug active on the ATP synthase of Mycobacterium tuberculosis. Science 2005; 307(5707):223–7. [DOI] [PubMed] [Google Scholar]
  • 13.Lounis N, Gevers T, Van Den Berg J, et al. ATP synthase inhibition of Mycobacterium avium is not bactericidal. Antimicrob Agents Chemother 2009; 53(11):4927–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zhang T, Li SY, Williams KN, et al. Short-course chemotherapy with TMC207 and rifapentine in a murine model of latent tuberculosis infection. Am J Respir Crit Care Med 2011;184(6):732–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Shang S, Shanley CA, Caraway ML, et al. Activities of TMC207, rifampin, and pyrazinamide against Mycobacterium tuberculosis infection in Guinea pigs. Antimicrob Agents Chemother 2011;55(1): 124–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tasneen R, Li SY, Peloquin CA, et al. Sterilizing activity of novel TMC207- and PA-824-containing regimens in a murine model of tuberculosis. Antimicrob Agents Chemother 2011;55(12):5485–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ibrahim M, Andries K, Lounis N, et al. Synergistic activity of R207910 combined with pyrazinamide against murine tuberculosis. Antimicrob Agents Chemother 2007;51(3):1011–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Andries K, Gevers T, Lounis N. Bactericidal potencies of new regimens are not predictive of their sterilizing potencies in a murine model of tuberculosis. Antimicrob Agents Chemother 2010;54(11): 4540–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Li S-Y, Tasneen R, Tyagi S, et al. Bactericidal and sterilizing activity of a novel regimen with bedaquiline, pretomanid, moxifloxacin, and pyrazinamide in a murine model of tuberculosis. Antimicrob Agents Chemother 2017;61(9):1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.FDA. SIRTURO® (bedaquiline) tablets, for oral use highlights of prescribing information 2012. Available at: https://www.sirturo.com/sites/default/files/pdf/sirturo-pi.pdf. Accessed January 13, 2019.
  • 21.van Heeswijk RPG, Dannemann B, Hoetelmans RMW. Bedaquiline: a review of human pharmacokinetics and drug-drug interactions. J Antimicrob Chemother 2014;69(9):2310–8. [DOI] [PubMed] [Google Scholar]
  • 22.Diacon AH, Donald PR, Pym A, 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. Antimicrob Agents Chemother 2012;56(6):3271–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rouan MC, Lounis N, Gevers T, et al. Pharmacokinetics and pharmacodynamics of TMC207 and its N-desmethyl metabolite in a murine model of tuberculosis. Antimicrob Agents Chemother 2012; 56(3):1444–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.McLeay SC, Vis P, Van Heeswijk RPG, et al. Population pharmacokinetics of bedaquiline (TMC207), a novel antituberculosis drug. Antimicrob Agents Chemother 2014;58(9):5315–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Svensson EM, Dosne AG, Karlsson MO. Population pharmacokinetics of bedaquiline and metabolite M2 in patients with drug-resistant tuberculosis: the effect of time-varying weight and albumin. CPT Pharmacometrics Syst Pharmacol 2016; 5(12):682–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Svensson EM, Murray S, Karlsson MO, et al. Rifampicin and rifapentine significantly reduce concentrations of bedaquiline, a new anti-TB drug. J Antimicrob Chemother 2014;70(4): 1106–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Svensson EM, Aweeka F, Park JG, et al. Model-based estimates of the effects of efavirenz on bedaquiline pharmacokinetics and suggested dose adjustments for patients coinfected with HIV and tuberculosis. Antimicrob Agents Chemother 2013; 57(6):2780–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Brill MJE, Svensson EM, Pandie M, et al. Confirming model-predicted pharmacokinetic interactions between bedaquiline and lopinavir/ritonavir or nevirapine in patients with HIV and drug-resistant tuberculosis. Int J Antimicrob Agents 2017;49(2): 212–7. [DOI] [PubMed] [Google Scholar]
  • 29.Huitric E, Verhasselt P, Koul A, et al. Rates and mechanisms of resistance development in Mycobacterium tuberculosis to a novel diarylquinoline ATP synthase inhibitor. Antimicrob Agents Chemother 2010;54(3):1022–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hartkoorn RC, Uplekar S, Cole ST. Cross-resistance between clofazimine and bedaquiline through upregulation of mmpl5 in mycobacterium tuberculosis. Antimicrob Agents Chemother 2014; 58(5):2979–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Almeida D, Ioerger T, Tyagi S, et al. Mutations in pepQ confer low-level resistance to bedaquiline and clofazimine in Mycobacterium tuberculosis. Antimicrob Agents Chemother 2016;60(8):4590–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Villellas C, Coeck N, Meehan CJ, et al. Unexpected high prevalence of resistance-associated Rv0678 variants in MDR-TB patients without documented prior use of clofazimine or bedaquiline. J Antimicrob Chemother 2017;72(3):684–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bloemberg GV, Keller PM, Stucki D, et al. Acquired resistance to bedaquiline and delamanid in therapy for tuberculosis. N Engl J Med 2015;373(20): 1986–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.World Health Organization (WHO). WHO technical report on critical concentrations for TB drug susceptibility testing of medicines used in the treatment of drug-resistant TB. Geneva (Switzerland): WHO; 2018. . Available at: http://www.who.int/tb/publications/2018/WHO_technical_report_concentrations_TB_drug_susceptibility/en/. [Google Scholar]
  • 35.Diacon AH, Dawson R, Von Groote-Bidlingmaier F, et al. 14-day bactericidal activity of PA-824, bedaquiline, pyrazinamide, and moxifloxacin combinations: a randomised trial. Lancet 2012;380(9846): 986–93. [DOI] [PubMed] [Google Scholar]
  • 36.Diacon AH, Dawson R, Von Groote-Bidlingmaier F, et al. Randomized dose-ranging study of the 14-day early bactericidal activity of bedaquiline (TMC207) in patients with sputum microscopy smear-positive pulmonary tuberculosis. Antimicrob Agents Chemother 2013;57(5):2199–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Koul A, Vranckx L, Dhar N, et al. Delayed bactericidal response of Mycobacterium tuberculosis to bedaquiline involves remodelling of bacterial metabolism. Nat Commun 2014;5:3369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Diacon AH, Pym A, Grobusch MP, et al. Multidrug-resistant tuberculosis and culture conversion with bedaquiline. N Engl J Med 2014;371(8):723–32. [DOI] [PubMed] [Google Scholar]
  • 39.Pym AS, Diacon AH, Tang SJ, et al. Bedaquiline in the treatment of multidrug and extensively drug-resistant tuberculosis. Eur Respir J 2016;47(2): 564–74. [DOI] [PubMed] [Google Scholar]
  • 40.Schnippel K, Ndjeka N, Maartens G, et al. Effect of bedaquiline on mortality in South African patients with drug-resistant tuberculosis: a retrospective cohort study. Lancet Respir Med 2018;6(9): 699–706. [DOI] [PubMed] [Google Scholar]
  • 41.Ndjeka N, Schnippel K, Master I, et al. High treatment success rate for multidrug-resistant and extensively drug-resistant tuberculosis using a bedaquiline-containing treatment regimen. Eur Respir J 2018;52(6):1801528. [DOI] [PubMed] [Google Scholar]
  • 42.Guglielmetti L, Jaspard M, Le Dû D, et al. Longterm outcome and safety of prolonged bedaquiline treatment for multidrug-resistant tuberculosis. Eur Respir J 2017;49(3). 10.1183/13993003.01799-2016. [DOI] [PubMed] [Google Scholar]
  • 43.Borisov SE, Dheda K, Enwerem M, et al. Effectiveness and safety of bedaquiline-containing regimens in the treatment of MDR- and XDR-TB: a multicentre study. Eur Respir J 2017;49(5). 10.1183/13993003.00387-2017. [DOI] [PubMed] [Google Scholar]
  • 44.Zhao Y, Fox T, Manning K, et al. Improved treatment outcomes with bedaquiline when substituted for second-line injectable agents in multidrug resistant tuberculosis: a retrospective cohort study. Clin Infect Dis 2019;68(9):1522–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Stover CK, Warrener P, VanDevanter DR, et al. A small-molecule nitroimidazopyran drug candidate for the treatment of tuberculosis. Nature 2000;405(6789):962–6. [DOI] [PubMed] [Google Scholar]
  • 46.Matsumoto M, Hashizume H, Tomishige T, et al. OPC-67683, a nitro-dihydro-imidazooxazole derivative with promising action against tuberculosis in vitro and in mice. PLoS Med 2006;3(11):2131–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chen X, Hashizume H, Tomishige T, et al. Delamanid kills dormant mycobacteria in vitro and in a Guinea pig model of tuberculosis. Antimicrob Agents Chemother 2017;61(6):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mallikaarjun S, Wells C, Petersen C, et al. Delamanid coadministered with antiretroviral drugs or anti-tuberculosis drugs shows no clinically relevant drug-drug interactions in healthy subjects. Antimicrob Agents Chemother 2016;60(10):5976–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gler MT, Skripconoka V, Sanchez-Garavito E, et al. Delamanid for multidrug-resistant pulmonary tuberculosis. N Engl J Med 2012;366(23):2151–60. [DOI] [PubMed] [Google Scholar]
  • 50.Sasahara K, Shimokawa Y, Hirao Y, et al. Pharmacokinetics and metabolism of delamanid, a novel anti-tuberculosis drug, in animals and humans: importance of albumin metabolism in vivo. Drug Metab Dispos 2015;43(8):1267–76. [DOI] [PubMed] [Google Scholar]
  • 51.Diacon AH, Dawson R, Hanekom M, et al. Early bactericidal activity of delamanid (OPC-67683) in smear-positive pulmonary tuberculosis patients. Int J Tuberc Lung Dis 2011;15(7):949–54. [DOI] [PubMed] [Google Scholar]
  • 52.Haver HL, Chua A, Ghode P, et al. Mutations in genes for the F420biosynthetic pathway and a nitroreductase enzyme are the primary resistance determinants in spontaneous in vitro-selected PA-824-resistant mutants of Mycobacterium tuberculosis. Antimicrob Agents Chemother 2015;59(9): 5316–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Pang Y, Zong Z, Huo F, et al. In vitro drug susceptibility of bedaquiline, delamanid, linezolid, clofazimine, moxifloxacin, and gatifloxacin against extensively drug-resistant tuberculosis in Beijing, China. Antimicrob Agents Chemother 2017; 61(10):1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Skripconoka V, Danilovits M, Pehme L, et al. Delamanid improves outcomes and reduces mortality in multidrug-resistant tuberculosis. Eur Respir J 2013;41(6):1393–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.von Groote-Bidlingmaier F, Patientia R, Sanchez E, et al. Efficacy and safety of delamanid in combination with an optimised background regimen for treatment of multidrug-resistant tuberculosis: a multicentre, randomised, double-blind, placebo-controlled, parallel group phase 3 trial. Lancet Respir Med 2019;2600(18):1–12. [DOI] [PubMed] [Google Scholar]
  • 56.Kuksa L, Barkane L, Hittel N, et al. Final treatment outcomes of multidrug- and extensively drug-resistant tuberculosis patients in Latvia receiving delamanid-containing regimens. Eur Respir J 2017;50(5):1701105. [DOI] [PubMed] [Google Scholar]
  • 57.Chang K-C, Leung EC-C, Law W-S, et al. Early experience with delamanid-containing regimens in the treatment of complicated multidrug-resistant tuberculosis in Hong Kong. Eur Respir J 2018;51(6):2–5. [DOI] [PubMed] [Google Scholar]
  • 58.Dogra M, Palmer BD, Bashiri G, et al. Comparative bioactivation of the novel anti-tuberculosis agent PA-824 in Mycobacteria and a subcellular fraction of human liver. Br J Pharmacol 2011;162(1): 226–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Baptista R, Fazakerley DM, Beckmann M, et al. Untargeted metabolomics reveals a new mode of action of pretomanid (PA-824). Sci Rep 2018;8(1): 5084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lenaerts AJ, Gruppo V, Marietta KS, et al. Preclinical testing of the nitroimidazopyran PA-824 for activity against Mycobacterium tuberculosis in a series of in vitro and in vivo models. Antimicrob Agents Chemother 2005;49(6): 2294–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Tyagi S, Nuermberger E, Yoshimatsu T, et al. Bactericidal activity of the nitroimidazopyran PA-824 in a murine model of tuberculosis. Antimicrob Agents Chemother 2005;49(6):2289–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lakshminarayana SB, Boshoff HIM, Cherian J, et al. Pharmacokinetics-pharmacodynamics analysis of bicyclic 4-nitroimidazole analogs in a murine model of tuberculosis. PLoS One 2014;9(8). 10.1371/journal.pone.0105222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Nuermberger E, Tyagi S, Tasneen R, et al. Powerful bactericidal and sterilizing activity of a regimen containing PA-824, moxifloxacin, and pyrazinamide in a murine model of tuberculosis. Antimicrob Agents Chemother 2008;52(4):1522–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hu Y, Coates ARM, Mitchison DA. Comparison of the sterilising activities of the nitroimidazopyran PA-824 and moxifloxacin against persisting Mycobacterium tuberculosis. Int J Tuberc Lung Dis 2008; 12(1):69–73. Available at: http://www.ncbi.nlm.nih.gov/pubmed/18173880. [PubMed] [Google Scholar]
  • 65.Ginsberg AM, Laurenzi MW, Rouse DJ, et al. Safety, tolerability, and pharmacokinetics of PA-824 in healthy subjects. Antimicrob Agents Chemother 2009;53(9):3720–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Dooley KE, Luetkemeyer AF, Park JG, et al. Phase I safety, pharmacokinetics, and pharmacogenetics study of the antituberculosis drug PA-824 with concomitant lopinavir-ritonavir, efavirenz, or rifampin. Antimicrob Agents Chemother 2014; 58(9):5245–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Rifat D, Li S-Y, Ioerger T, et al. Mutations in Rv2983 as a novel determinant of resistance to nitroimidazole drugs in Mycobacterium tuberculosis. bioRxivorg 2018;457754. Available at: https://www.biorxiv.org/content/biorxiv/early/2018/10/31/457754.full.pdf. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Diacon AH, Dawson R, Hanekom M, et al. Early bactericidal activity and pharmacokinetics of PA-824 in smear-positive tuberculosis patients. Antimicrob Agents Chemother 2010;54(8):3402–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Diacon AH, Dawson R, Du Bois J, et al. Phase II dose-ranging trial of the early bactericidal activity of PA-824. Antimicrob Agents Chemother 2012; 56(6):3027–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Diacon AH, Dawson R, Von Groote-Bidlingmaier F, et al. Bactericidal activity of pyrazinamide and clofazimine alone and in combinations with pretomanid and bedaquiline. Am J Respir Crit Care Med 2015;191(8):943–53. [DOI] [PubMed] [Google Scholar]
  • 71.Dawson R, Diacon AH, Everitt D, 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 pul. Lancet 2015;385(9979):1738–47. [DOI] [PubMed] [Google Scholar]
  • 72.Clinical trial of BPaMZ regimen will replace phase 3 STAND trial. TB Alliance. 2016. Available at: https://www.tballiance.org/news/clinical-trial-bpamz-regimen-will-replace-phase-3-stand-trial. Accessed January 13, 2019.. [Google Scholar]
  • 73.Conradie F, Diacon A, Howell P, et al. Sustained high rate of successful treatment outcomes: interim results of 75 patients in the Nix-TB clinical study of pretomanid, bedaquiline and linezolid. In: The Union 2018. The Hague. October 25, 2018. [Google Scholar]
  • 74.Ferlazzo G, Mohr E, Laxmeshwar C, et al. Early safety and efficacy of the combination of bedaquiline and delamanid for the treatment of patients with drug-resistant tuberculosis in Armenia, India, and South Africa: a retrospective cohort study. Lancet Infect Dis 2018;18(5):536–44. [DOI] [PubMed] [Google Scholar]
  • 75.Maryandyshev A, Pontali E, Tiberi S, et al. Bedaquiline and delamanid combination treatment of 5 patients with pulmonary extensively drug-resistant tuberculosis. Emerg Infect Dis 2017;23(10): 1718–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Guglielmetti L, Barkane L, Le Du D, et al. Safety and efficacy of exposure to bedaquiline-delamanid in MDR-TB: a case series from France and Latvia. Eur Respir J 2018;51(3):1702550. [DOI] [PubMed] [Google Scholar]
  • 77.Tae Kim C, Kim T-O, Shin H-J, et al. Bedaquiline and delamanid for the treatment of multidrug-resistant tuberculosis: a multi-center cohort study in Korea. Eur Respir J 2018;51(3):1702467. [DOI] [PubMed] [Google Scholar]
  • 78.Dawson R, Harris K, Conradie A, et al. Efficacy of bedaquiline, pretomanid, moxifloxacin, & PZA (BPaMZ) against DS- & MDR-TB. In: CROI Conference. Seattle, Washington, February 13-16, 2017. [Google Scholar]
  • 79.Akkerman OW, Odish OFF, Bolhuis MS, et al. Pharmacokinetics of bedaquiline in cerebrospinal fluid and serum in multidrug-resistant tuberculous meningitis. Clin Infect Dis 2015;62(4):523–4. [DOI] [PubMed] [Google Scholar]
  • 80.Verhaeghe T, Diels L, Dillen L. Quantitation of bedaquiline: points of attention. Clin Infect Dis 2016; 63(1):145–6. [DOI] [PubMed] [Google Scholar]
  • 81.Shibata M, Shimokawa Y, Sasahara K, et al. Absorption, distribution and excretion of the anti-tuberculosis drug delamanid in rats: extensive tissue distribution suggests potential therapeutic value for extrapulmonary tuberculosis. Biopharm Drug Dispos 2017;38(4):301–12. [DOI] [PubMed] [Google Scholar]
  • 82.Tucker EW, Pieterse L, Zimmerman MD, et al. Delamanid central nervous system pharmacokinetics in tuberculous meningitis in rabbits and humans. Antimicrob Agents Chemother 2019;63(10). pii: e00913–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Shobo A, Bratkowska D, Baijnath S, et al. Tissue distribution of pretomanid in rat brain via mass spectrometry imaging. Xenobiotica 2016;46(3): 247–52. [DOI] [PubMed] [Google Scholar]
  • 84.World Health Organization (WHO). WHO treatment guidelines for multidrug- and rifampin-resistant tuberculosis: 2018 update. WHO; 2018. Geneva (Switzerland). [Google Scholar]
  • 85.Svensson EM, du Bois J, Kitshoff R, et al. Relative bioavailability of bedaquiline tablets suspended in water: implications for dosing in children. Br J Clin Pharmacol 2018;84(10):2384–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Achar J, Hewison C, Cavalheiro AP, et al. Off-label use of bedaquiline in children and adolescents with multidrug-resistant tuberculosis. Emerg Infect Dis 2017;23(10):1711–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Schaaf HS, Thee S, van der Laan L, et al. Adverse effects of oral second-line antituberculosis drugs in children. Expert Opin Drug Saf 2016;15(10): 1369–81. [DOI] [PubMed] [Google Scholar]
  • 88.Esposito S, Bosis S, Tadolini M, et al. Efficacy, safety, and tolerability of a 24-month treatment regimen including delamanid in a child with extensively drug-resistant tuberculosis. Medicine (Baltimore) 2016;95(46):e5347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Jaspard M, Elefant-Amoura E, Melonio I, et al. Bedaquiline and linezolid for extensively drug-resistant tuberculosis in pregnant woman. Emerg Infect Dis 2017;23(10):153–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Clinical pipeline. Working group on new TB drugs. Available at: https://www.newtbdrugs.org/pipeline/clinical. Accessed January 15, 2019.
  • 91.Takahashi Y, Igarashi M, Miyake T, et al. Novel semisynthetic antibiotics from caprazamycins A-G: Caprazene derivatives and their antibacterial activity. J Antibiot (Tokyo) 2013;66(3):171–8. [DOI] [PubMed] [Google Scholar]
  • 92.Kling A, Lukat P, Almeida DV, et al. Antibiotics. Targeting DnaN for tuberculosis therapy using novel griselimycins. Science 2015;348(6239):1106–12. [DOI] [PubMed] [Google Scholar]
  • 93.Ahmad Z, Minkowski A, Peloquin CA, et al. Activity of the fluoroquinolone DC-159a in the initial and continuation phases of treatment of murine tuberculosis. Antimicrob Agents Chemother 2011; 55(4):1781–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zhang T, Lu X, Tu Z, et al. Compound TB47 has strong activity against Mycobacterium tuberculosis both in vitro and in vivo. In: ASM Microbe. New Orleans, June 4, 2017. [Google Scholar]
  • 95.Shoen C, Pucci M, DeStefano MC M. Efficacy of SPR720 and SPR750 Gyrase inhibitors in a mouse Mycobacterium tuberculosis infection model. In: ASM Microbe. New Orleans, June 2, 2017. [Google Scholar]
  • 96.Shionogi & Co. Ltd. Research and Development at Shionogi. Available at: http://www.shionogi.co.jp/en/ir/pdf/e_p180315.pdf.
  • 97.GSK-286. Working group on new TB drugs. Available at: https://www.newtbdrugs.org/pipeline/compound/gsk-286. Accessed January 10, 2019.
  • 98.Sutherland HS, Tong AST, Choi PJ, et al. Structure-activity relationships for analogs of the tuberculosis drug bedaquiline with the naphthalene unit replaced by bicyclic heterocycles. Bioorg Med Chem 2018;26(8):1797–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Our pipeline: TBAJ-876. TB alliance. Available at: https://www.tballiance.org/portfolio/compound/tbaj-876. Accessed January 10, 2019.
  • 100.El-Gamal MI, Brahim I, Hisham N, et al. Recent updates of carbapenem antibiotics. Eur J Med Chem 2017;131:185–95. [DOI] [PubMed] [Google Scholar]
  • 101.Mdluli K, Cooper C, Yang T, et al. TBI-223: A safer oxazolidinone in pre-clinical development for tuberculosis. In: ASM Microbe. New Orleans, June 4, 2017. Available at: http://www.abstractsonline.com/pp8/#!/4358/presentation/6174. [Google Scholar]
  • 102.Robertson GT, Scherman MS, Bruhn DF, et al. Spectinamides are effective partner agents for the treatment of tuberculosis in multiple mouse infection models. J Antimicrob Chemother 2017;72(3):770–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lechartier B, Hartkoorn RC, Cole ST. In vitro combination studies of benzothiazinone lead compound BTZ043 against Mycobacterium tuberculosis. Antimicrob Agents Chemother 2012;56(11):5790–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Palencia A, Li X, Bu W, et al. Discovery of novel oral protein synthesis inhibitors of mycobacterium tuberculosis that target leucyl-tRNA synthetase. Antimicrob Agents Chemother 2016;60(10): 6271–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Shoen C, DeStefano M, Hafkin B, et al. In vitro and in vivo activities of contezolid (MRX-I) against Mycobacterium tuberculosis. Antimicrob Agents Chemother 2018;62(8):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lupien A, Vocat A, Foo CS-Y, et al. Optimized background regimen for treatment of active tuberculosis with the next-generation benzothiazinone macozinone (PBTZ169). Antimicrob Agents Chemother 2018;62(11):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Zhang D, Liu Y, Zhang C, et al. Synthesis and biological evaluation of novel 2-methoxypyridylamino-substituted riminophenazine derivatives as antituberculosis agents. Molecules 2014;19(4):4380–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Chatterji M, Shandil R, Manjunatha MR, et al. 1,4-azaindole, A potential drug candidate for treatment of tuberculosis. Antimicrob Agents Chemother 2014;58(9):5325–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kang S, Kim RY, Seo MJ, et al. Lead optimization of a novel series of imidazo[1,2-a]pyridine amides leading to a clinical candidate (Q203) as a multi- and extensively-drug-resistant anti-tuberculosis agent. J Med Chem 2014;57(12):5293–305. [DOI] [PubMed] [Google Scholar]
  • 110.Williams KN, Brickner SJ, Stover CK, et al. Addition of PNU-100480 to first-line drugs shortens the time needed to cure murine tuberculosis. Am J Respir Crit Care Med 2009;180(4):371–6. [DOI] [PubMed] [Google Scholar]
  • 111.Wallis RS, Jakubiec W, Kumar V, et al. Biomarker-assisted dose selection for safety and efficacy in early development of PNU-100480 for tuberculosis. Antimicrob Agents Chemother 2011;55(2): 567–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Wallis RS, Dawson R, Friedrich SO, et al. Mycobactericidal activity of sutezolid (PNU-100480) in sputum (EBA) and blood (WBA) of patients with pulmonary tuberculosis. PLoS One 2014;9(4). 10.1371/journal.pone.0094462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Jeong JW, Jung SJ, Lee HH, et al. In vitro and in vivo activities of LCB01-0371, a new oxazolidinone. Antimicrob Agents Chemother 2010;54(12): 5359–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Choi Y, Lee SW, Kim A, et al. Safety, tolerability and pharmacokinetics of 21 day multiple oral administration of a new oxazolidinone antibiotic, LCB01-0371, in healthy male subjects. J Antimicrob Chemother 2018;73(1):183–90. [DOI] [PubMed] [Google Scholar]
  • 115.Tahlan K, Wilson R, Kastrinsky DB, et al. SQ109 targets MmpL3, a membrane transporter of trehalose monomycolate involved in mycolic acid donation to the cell wall core of Mycobacterium tuberculosis. Antimicrob Agents Chemother 2012;56(4): 1797–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Heinrich N, Dawson R, Bois J, et al. Early phase evaluation of SQ109 alone and in combination with rifampicin in pulmonary TB patients. J Antimicrob Chemother 2014;70(5):1558–66. [DOI] [PubMed] [Google Scholar]
  • 117.Boeree MJ, Heinrich N, Aarnoutse R, et al. High-dose rifampicin, moxifloxacin, and SQ109 for treating tuberculosis: a multi-arm, multi-stage randomised controlled trial. Lancet Infect Dis 2017; 17(1):39–49. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES