You are on page 1of 35

As a library, NLM provides access to scientific literature.

Inclusion in an NLM
database does not imply endorsement of, or agreement with, the contents by NLM
or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

RESPIRATION Karger Publishers

Respiration. 2023 Feb; 102(2): 83–100. PMCID: PMC9932851


Published online 2022 Dec 14. doi: 10.1159/000528274 PMID: 36516792

New and Repurposed Drugs for the Treatment of Active Tuberculosis: An


Update for Clinicians
Jessica M Aguilar Diaz, a Ahmed A Abulfathi, b , c , d Lindsey HM te Brake, e Jakko van Ingen, f
Saskia Kuipers, f Cecile Magis-Escurra, a Jelmer Raaijmakers, f Elin M Svensson, e , g and
Martin J Boeree a ,*

Abstract

Although tuberculosis (TB) is preventable and curable, the lengthy treatment (generally 6
months), poor patient adherence, high inter-individual variability in pharmacokinetics (PK),
emergence of drug resistance, presence of comorbidities, and adverse drug reactions
complicate TB therapy and drive the need for new drugs and/or regimens. Hence, new
compounds are being developed, available drugs are repurposed, and the dosing of existing
drugs is optimized, resulting in the largest drug development portfolio in TB history. This
review highlights a selection of clinically available drug candidates that could be part of
future TB regimens, including bedaquiline, delamanid, pretomanid, linezolid, clofazimine,
optimized (high dose) rifampicin, rifapentine, and para-aminosalicylic acid. The review
covers drug development history, preclinical data, PK, and current clinical development.

Keywords: Drugs, Treatment, Tuberculosis

Introduction
In the last century, the incidence of tuberculosis (TB) has declined considerably with a
setback in the past few years due to the COVID-19 pandemic. The decline in TB incidence
was accelerated by the introduction of anti-TB treatment after the Second World War, which
stopped in the 1980s among other factors due to the emergence of HIV and drug-resistant TB
(DR-TB). At that moment, new drugs were not specifically developed for TB. However in the
last 2 decades, a new impulse in drug treatment was given through the development of global
coordination in the fight against the poverty-related diseases HIV/acquired
immunodeficiency syndrome, malaria, and TB. The efforts resulted in the formation of
several research consortia sometimes in collaboration with the pharmaceutical industry,
leading to an enhanced effort to develop new compounds, repurpose already available drugs,
or optimize doses of first-line TB drugs: Pan-African Consortium for the Evaluation of
Antituberculosis Antibiotics (PanACEA), International Consortium for Trials of
Chemotherapeutic Agents in Tuberculosis (INTERTB), Tuberculosis Trials Consortium
(TBTC), Acquired Immunodeficiency Syndrome Clinical Trials Group (ACTG), Project to
Accelerate New Treatments for Tuberculosis (PAN-TB), and most recently the UNITE4TB
consortium. Currently, there is a portfolio of novel/repurposed/redeveloped TB drugs in
preclinical and clinical development, representing the largest pipeline in history of TB drug
development. This review will highlight a selection of the clinically available drug candidates
that could be part of future TB regimens. We selected registered and clinically available
drugs, which were selected because they are new (bedaquiline, delamanid, and pretomanid),
repurposed (linezolid), revived (clofazimine), or optimized (rifampicin, rifapentine, and para-
aminosalicylic acid [PAS]). The selected drugs will potentially be suitable for an optimized
or shortened regimen for drug-sensitive TB (DS-TB) or DR-TB in the near future. The
review describes drug development history, preclinical data, pharmacokinetics (PK), and
current clinical development.

New Drugs

Bedaquiline

The antimycobacterial properties of diarylquinolines were patented in 2004. The first


scientific report on the activity of bedaquiline (formerly known as TMC207 and R207910)
was published in Science in 2005 [1]. The compound was developed by Johnson & Johnson,
which obtained conditional market approval for the treatment of DR-TB from the US Food
and Drug Administration (FDA) in 2012. Bedaquiline was then the first anti-TB drug with a
novel mechanism of action to reach TB patients in more than 40 years (Table 1).
Table 1

Characteristics of each drug in the treatment of TB

Drug Molecule name Class Mechanism of Clinical Dose adults


action indication

Bedaquiline C32H31 (Diaryl) Inhibits ATP DR-TB, 400 mg once-


BrN202 quinoline synthesis by investigated daily orally for 2
targeting in DS-TB weeks and 200
mycobacterial mg 3 times a
ATP synthase weekafterward
for 22 weeks [20]

Delamanid C25H25F3N406 Nitro-dihydro- Inhibits mycolic DR-TB 100 mg twice a


imidazooxazole acid synthesis and day for 24 weeks
cellular [37]
respiration

PA C14H12F3N305 Nitro-dihydro- Inhibits mycolic DR-TB, 200 mg once


imidazooxazole acid synthesis investigated daily for 26
in DS-TB weeks [49]

Linezolid C16H20FN3O4 Oxazolidinone Inhibits protein DR-TB 600 mg once or


synthesis, binds twice daily [20];
to the 23S RNA 6 months in
peptidyl newest BPaLM
transferase center regimen; higher
(PTC) of the once-daily doses
prokaryotic are under
ribosomal 50S investigation for
subunit TB meningitis

Clofazimine C27H22CI2N4 Phenazine Incompletely DR-TB 100 mg once


understood, daily [20] at least
includes 9 months;

Bedaquiline targets mycobacterial ATP synthases and thereby the energy metabolism of the
bacteria (shown in Fig. 1 [2]) [3]. The minimum inhibitory concentration (MIC) of
bedaquiline is typically 0.012–1.0 mg/L when tested by broth microdilution with the
Mycobacterium Growth Indicator Tube (MGIT) system, and a clinical cut-off at 1 mg/L has
been suggested [4]. A dose-fractionation study in mice identified total exposure (AUC) as the
main driver of bactericidal effect [5]. In a murine model of latent TB, bedaquiline
demonstrated at least as good sterilizing activity as rifampicin [6].

Fig. 1

Bacterial representation of M. tuberculosis with mechanism of action for each drug, adapted with
permission from Hoelscher [2].

Bedaquiline is primarily metabolized through n-demethylation by hepatic isoenzyme


CYP3A4 to the approximately 5 times less active M2 metabolite [7]. The terminal half-life
(T1/2) of bedaquiline is extremely long (>5 months), and the approved dosing regimen
therefore includes a loading-phase (2 weeks of 400 mg once daily) and a maintenance phase
(200 mg 3 times per week) (Table 1) [7]. The plasma protein binding of bedaquiline is
>99.9% [7]. Bedaquiline exposures are affected by drugs interacting with CYP3A4, and
important examples are rifampicin and rifapentine (decreasing bedaquiline exposures almost
4- to 5-fold) and antiretroviral drugs such as efavirenz (decreasing bedaquiline exposures by
half) and lopinavir/ritonavir (increasing bedaquiline exposures by 2- to 3-fold increase) [8, 9,
10, 11].

Bedaquiline has limited early bactericidal activity (EBA) on its own, and the effect can only
be seen after about 1 week of treatment [12]. In phase 2 trials including multi-DR-TB (MDR-
TB) patients, it was shown that the addition of bedaquiline to an optimized background
regimen shortened the time-to-culture conversion and increased the proportion of favorable
outcomes at follow-up [13]. Model-based analyses found that the weekly average bedaquiline
concentration is associated with the rate of decline in bacterial load and suggest that the
currently used dosing regimen is not achieving maximal possible efficacy [14]. The main side
effect of bedaquiline is QT prolongation, driven primarily by the exposure to the M2
metabolite [15]. This raised concerns regarding combining bedaquiline with delamanid, a
drug that also causes QT prolongation, but recent data indicate that the combined effect of
the two is clinically modest and no more than additive [16]. Bedaquiline for treatment of
MDR-TB is currently being tested in a phase 3 setting through the STREAM trial (stage 2),
with results expected in 2022 [17]. Bedaquiline in combination with pretomanid and
linezolid has demonstrated 90% (95% CI: 83–95%) favorable outcomes in extensively DR-TB
(XDR-TB) and treatment-intolerant MDR-TB patients, enabling the recent approval of this
specific regimen (bedaquiline, pretomanid, and linezolid [BPaL]) [18]. Furthermore, in May
2022, the WHO published a rapid communication endorsing the 6-month bedaquiline,
pretomanid, linezolid, and moxifloxacin (BPaLM) regimen based on data of the ZeNix-TB
study and TB-PRACTECAL, which are described in further sections of this review [19]. If
there is resistance to fluoroquinolones, moxifloxacin should not be given [19].

In the latest update of WHO guidance on treatment of DR-TB, bedaquiline was categorized
as a group A drug that is considered highly effective and strongly recommended for inclusion
in all regimens unless contraindicated [20]. Very limited data on the use of bedaquiline in
children are available [21]. A child-friendly, 20 mg scored, dispersible tablet has been
developed. Pediatric PK and safety studies are still ongoing, but FDA and European
Medicines Agency (EMA) already recommend bedaquiline for use in children 5 years and
older, weighing at least 15 kg. The WHO published consolidated guidelines in 2022, where it
conditionally recommends bedaquiline use in all children [22]. Experience with bedaquiline
for DR-TB in programmatic use is growing, and reports are mainly positive with relatively
high rates of culture conversion (80–95%) and few serious adverse events [23, 24, 25, 26, 27,
28, 29]. Bedaquiline can be used during pregnancy and in a South African cohort including
108 pregnant women where about half were treated with bedaquiline, and 88% of babies
exposed to bedaquiline in utero were thriving and developing normally at 12 months
compared to 82% of the babies not exposed [30]. It is unclear how well bedaquiline
penetrates into the central nervous system, enabling effective treatment of TB meningitis.
One case report noted that concentrations of bedaquiline in cerebrospinal fluid were
undetectable [31], but a recent study reported bedaquiline levels in cerebrospinal fluid in the
same order of magnitude as unbound plasma concentrations [32].

Few concerns regarding emergence of resistance to bedaquiline and MIC-increasing


mutations in the atpE, mmpR (Rv0678), and pepQ (Rv2535c) genes have been described
[33]. Notably, cross-resistance with clofazimine has also been detected [34, 35].

Delamanid

Delamanid (Deltyba®), a nitro-dihydro-imidazooxazole derivative, formerly known as OPC-


67683 [36], was patented in the 1990s. In 2014, shortly following the approval of
bedaquiline, delamanid obtained EMA's approval for early release to the market as the
second new drug for DR-TB treatment in over 40 years (Table 1).

Delamanid's exact target is yet to be determined. However, delamanid is known to inhibit


mycolic acid synthesis and cellular respiration (shown in Fig. 1 [2], Table 1). The inhibitory
effect probably involves the release of reactive radicals such as nitric oxide, which are crucial
in the mammalian defense mechanism against mycobacterial infections [37].

The drug has a low MIC90: 0.006–0.024 μg/mL and is found to be active against DS and DR
M. tuberculosis isolates [38]. It inhibits replicating and dormant bacilli, both extracellularly
and intracellularly. In murine experiments, the combination of delamanid with rifampicin and
pyrazinamide resulted in more rapid sterilization of lung tissue than the standard regimen of
first-line anti-TB drugs. Its activity against intracellular M. tuberculosis was equivalent to
that of rifampicin at a concentration of 1–3 μg/mL [37].

Delamanid has a time to peak concentration (Tmax) of 4 h, with the peak concentration
(Cmax) varying from 175 to 286 ng/mL in the dose range of 100–400 mg [39]. The area under
the plasma concentration-time curve from 0 to 24 h (AUC0–24) ranged from 2,500 to 5,483
h*ng/mL at the dose range of 100–400 mg, indicating that the increase in exposure is not
dose-proportional, and reaches a plateau at 300 mg dose [39]. When taken with food,
delamanid bioavailability increases 2- to 4-fold. The apparent T1/2 is 30–38 h, while the T1/2
of its main metabolite is approximately 150–600 h. Delamanid is metabolized to M1, a
unique metabolite formed by plasma albumin. Nonhepatic formation of M1 and multiple
separate pathways for metabolism of M1, including CYP3A4, suggest that clinically
significant drug-drug interactions with delamanid and M1 are limited, although concurrent
administration with medicines known to induce/inhibit CYP3A4 may modestly alter
delamanid exposures [40]. Delamanid and its major metabolites do not inhibit or induce CYP
enzyme activity and thus have little potential for interaction with antiretroviral drugs [40,
41]. Its high plasma protein binding (≥99.5%), especially to albumin, alters its volume of
distribution [42].

The 14-day EBA in a phase 1 trial with a 200 mg daily dose was 0.052 log10 cfu/mL sputum
per day supporting the bactericidal potential of this drug. The EBA of delamanid was
monophasic and not significantly different between dosages, in line with the overlapping
exposure profiles. Adverse events in this 14-day trial were of either mild or moderate
severity. No serious adverse events occurred, and QT intervals were not prolonged [39]. The
first randomized placebo-controlled-phase 2 trial (2012) showed an increase in 2-month
culture conversion rates in patients with DR-TB treated with delamanid in combination with
a backbone regimen compared to placebo [43]. However, a randomized, double-blind,
placebo-controlled phase 3 multicenter trial, published in 2019, could not show any
difference in efficacy between the two groups at 6 months of evaluation [44]. A multicenter
observational study (n = 53) showed that 67.6% of a difficult-to-treat cohort of DR-TB
patients successfully culture converted by 6 months of treatment with delamanid. A total of
31 serious adverse events were reported in 14 patients (26.4%); most common were
hepatotoxicity (5), electrolyte imbalance (5), and QT prolongation (3) [45].

A recent meta-analysis of seven studies that used a bedaquiline and delamanid combination
to treat 87 cases of DR-TB showed promising outcomes. Most M/XDR-TB patients were
concomitantly treated with bedaquiline and delamanid, and in most cases for a duration >6
months. The sputum culture conversion rate after 6 months of treatment was considerably
higher (81.4%) than in historical M/XDR-TB patient cohorts. Out of 87 patients, 23 (26.4%)
had slight increases in QTc. However, only 2.3% of treatments were interrupted because of
life-threatening cardiac adverse events [46]. In children (aged 3 years and above), delamanid
shows an excellent safety and side effect profile [47].

Currently, there are several ongoing studies but because of paucity and weaknesses of the
existing evidence the efficacy of delamanid is still challenging to define [48]. Therefore,
pivotal clinical trials are needed to clarify its clinical value.

Pretomanid

Pretomanid (Dovprela®) (PA), another nitro-dihydro-imidazooxazole derivative, formerly


known as PA-824, was approved in 2019 by the FDA for XDR-TB or treatment-intolerant or
nonresponsive MDR-TB in the BPaL regimen, and in 2020 it received conditional approval
by the EMA (Table 1) [49, 50, 51]. PA was first identified in 1995 and is co-licensed by the
TB Alliance and Mylan with affordable access agreements for low- and middle-income
countries.

PA inhibits mycolic acid synthesis, a necessary step in cell wall formation (shown in Fig. 1
[2]) [52, 53]. PA is metabolized to reactive nitrogen species, which permits its activity
against nonreplicating bacilli. In vitro activity is shown with an MIC of <1 μg/mL and a
MIC90 of 0.063 μg/mL [54]. PA was found to be highly active in mice, especially in
combination with bedaquiline and linezolid [55]. Five genes are associated with the
emergence of resistance (ddn, fgd1, fbiA, fbiB, and fbiC) [56, 57]. Cross-resistance with
delamanid has been observed.

PA is administered orally as a tablet once daily. Following administration of a single dose of


200 mg with food, the mean Cmax is 2.0 μg/mL, the median Tmax is 5 h, and the mean AUC∞
is 51.6 μg*h/mL (Table 1) [49]. It is 86% protein-bound, and steady state is achieved after 4–
6 days. PA is metabolized via several pathways, 20% of which is via CYP3A4. PA is cleared
in urine and feces mainly as metabolites. PA's co-administration with rifampicin,
lopinavir/ritonavir, or efavirenz reduces its exposures, in the case of rifampicin by 53–85%
[58]. PA inhibits OAT3 transporter, suggesting that it may increase exposures of OAT3
substrates such as methotrexate.

PA has been investigated in several phase 2 trials for DS-TB. A phase 2A 14-day study with
PA evaluating various combinations bedaquiline, pretomanid, and pyrazinamide showed
equal EBA compared to the standard control [59]. In a phase 2B study (NC002) for 8 weeks,
a regimen combining moxifloxacin, pretomanid 200 mg, and pyrazinamide showed that there
was a significant increase of sputum culture conversion compared to the control [60]. In a
later phase 2B trial (NC005), adding bedaquiline to the regimen of moxifloxacin, pretomanid,
and pyrazinamide (BPaMZ) increased the bactericidal activity over 2 months considerably, in
both DS- and DR-TB [61].
Recently, two phase 3 trials for PA were performed with a focus on DR-TB. The BPaL
regimen was successfully investigated in the Nix-TB trial and was licensed for XDR-TB,
treatment-intolerant or nonresponsive MDR pulmonary TB [18]. TB-PRACTECAL, a phase
2/3 clinical trial, investigated the 6-month BPaLM regimen, against the locally accepted
standard of care. It found that 89% of patients in the group of patients receiving a 6-month
regimen of BPaLM were cured versus 52% of patients who had been prescribed the standard
regimen of up to 20 months of treatment, but this has not been published yet [62]. The
BPaLM regimen has now been included in the recent WHO rapid communication [19].

For both DS- as DR-TB, the TB Alliance has initiated the pivotal SimpliciTB trial. The trial
is almost finished, and the results will be presented at the Union World Conference on Lung
Health in November 2022. SimpliciTB is the continuation of the STAND trial that was
changed from PaMZ into BPaMZ when the results came available of the NC005 trial.
Further trials with PA are the phase 3 ZeNix trial (aimed to reduce linezolid toxicity) and the
phase 2B APT trial evaluating PA added to a first-line regimen with either rifampicin or
rifabutin, isoniazid, and pyrazinamide [63].

The most common adverse events of PA are gastrointestinal symptoms and vomiting and
suggested to be dose related [64], and the following described symptoms are not dose related:
transaminase increase, hepatotoxicity, and headache. The presence of hepatoxicity is
unfortunate as it reduces the value of a regimen to be used in low resources settings.

Repurposed Drugs

Linezolid

In 1987, when the antimicrobial activity of oxazolidinones against plant pathogens was
discovered [65], one of these agents was developed into linezolid (DuP721, PNU-100766, S-
N-3-3-fluoro-4-[4-morpholinyl]phenyl-2-oxo-5-oxazolidinylmethyl-acetamide). Linezolid
was marketed for treating gram-positive bacterial infections of the lungs, skin, and soft
tissues in 2000 and considered for the treatment of MDR-TB as a group 5 agent in 2006 by
the WHO [66].

Oxazolidinones inhibit protein synthesis by decreasing mRNA translation through binding to


the 23S RNA peptidyl transferase center of the prokaryotic ribosomal 50S subunit (shown in
Fig. 1 [2], Table 1) [67]. The binding of linezolid to human mitochondria can cause
myelosuppression, peripheral and optic neuropathy, and hyperlactatemia [68, 69].

The critical linezolid concentration for M. tuberculosis is 1 mg/L in 7H10 and MGIT
medium [70, 71]. Median linezolid MIC of 4,470 strains tested in M7H9 medium is 0.5 mg/L
(range 0.06–16 mg/L) [72].
In mice, 25–260 mg/kg of linezolid showed anti-tuberculous activity against replicating and
nonreplicating bacilli [73, 74]. A once daily linezolid hollow fiber model demonstrated
substantial activity against M. tuberculosis in acid-phase and nonreplicating persister states
[75].

Linezolid has an excellent oral bioavailability approaching 100%, low plasma protein binding
of 31%, and good tissue penetration including cerebrospinal fluid [76], alveolar macrophages
[77], and bone [78]. Plasma Cmax is 15–27 mg/L, and Tmax is 0.5–2 h [79]. When dosed 600
mg every 12 h, linezolid has an elimination half-life of 4.8–5.5 h (Table 1). Children under
12 years have a faster clearance and a shorter elimination half-life than adults [80].

Sixty-five percent of linezolid is oxidized into two inactive metabolites. About 30% of
linezolid and 50% of the metabolites are excreted via urine [81]. Although metabolism via
CYP3A4 plays a minor role in linezolid elimination, rifampicin co-administration reduces
linezolid exposure. Concomitant use of monoamine oxidase inhibitors with oxazolidinones is
contraindicated because of the risk of serotonin syndrome. Linezolid trough plasma
concentrations and AUC0-24 are linearly related, and trough concentrations are predictive of
linezolid exposure [82, 83]. Linezolid's EBA is modest in HIV-positive TB patients [84].

Currently, the WHO categorizes linezolid as a group A drug for rifampicin-resistant or


MDR-TB [47]. Several studies provide evidence for this choice. The addition of linezolid to
the ongoing regimen in 41 XDR-TB patients without response to earlier treatment showed
sputum culture conversion after 4 months in 15 out of 19 patients (79%) in the group that
started immediately and a negative sputum culture after 6 months of treatment in 34 out of
39 patients (87%) [85]. Improved 24-month treatment outcome, based on the WHO
definition, was found for an oral 6-month regimen including linezolid, levofloxacin, and
bedaquiline compared to the conventional empiric injection-based regimen, although it was
stopped early due to a new standard of care [86]. In the Nix-TB study, where 1,200 mg
linezolid daily was given with PA and bedaquiline anemia developed in 37% of patients, and
peripheral neuropathy in 81%, mainly attributed to linezolid toxicity. Only 34% completed
treatment, warranting careful monitoring of side effects of linezolid [18]. The phase 3 ZeNix
trial investigates the efficacy and toxicity of 600 mg versus 1,200 mg linezolid daily in
combination with PA and bedaquiline, and aims to reduce linezolid toxicity. Presented
results, which are not published yet, showed that the lower doses and/or shorter duration
treatment arms of linezolid had similar success rates and less toxicity in comparison with the
treatment arm including 1,200 mg of linezolid for 6 months [87]. As described in the PA
section, the BPaLM regimen was found to be efficacious in the TB-PRACTECAL trial [62,
88] and is included in the newest WHO rapid communication [19]. Another study including
linezolid is the MDR-END study, which compares the effectiveness and safety of oral
linezolid, delamanid, levofloxacin, and pyrazinamide for 9–12 months with the WHO 2016
MDR-TB regimen for 20–24 months [89]. [62].
Thus, a major concern and drawback in long-term linezolid use are its toxicity, which was a
reason to conduct the ZeNix trial, described above. A meta-analysis of twelve studies of
linezolid-containing TB regimens showed adverse events in 58.9% (63/107) of patients.
Anemia was found in 38.1% (32/84) of patients, peripheral neuropathy in 47.1% (40/85),
gastrointestinal disorders in 16.7% (14/84), optic neuritis in 13.2% (10/76), and
thrombocytopenia in 11.8% (10/85) of patients, especially with doses above 600 mg [90].
Monitoring hemoglobin levels and neuropathy may guide linezolid dosing optimization [91].
Therapeutic drug monitoring of linezolid is helpful when the daily dose is below 300 mg
[92]. A trough concentration of 2.5 mg/L should be evaluated as a target for therapeutic drug
monitoring [93].

Phenotypic linezolid resistance in M. tuberculosis, defined by an MIC >1 mg/L, emerged in


2007 and is mainly linked to mutations in or close to the PTC-binding site like the ribosomal
protein L3 rplC gene and the 23S rRNA rrl gene [33, 94]. In a South African study of MDR-
TB patients, linezolid resistance developed after 7–32 months of therapy. Sixteen
phenotypically resistant strains most frequently showed the T460C substitution in rplC, and
G2814T, G2270 C/T, and A2810C mutations in the rrl gene. No mutations were detected in
isolates with MICs at or below the critical concentration [95]. Mutations in rrl led to growth
impairment and decreased fitness which could limit spread in clinical settings [96]. Other
resistance mechanisms need further elucidation.

Revived Drugs

Clofazimine

Clofazimine was first synthesized and reported from Trinity College Dublin in 1957; it is the
prototype riminophenazine antibiotic [97]. While initially developed for TB treatment, its
preclinical development stalled as its in vivo activity proved inconsistent in particular animal
models, i.e., guinea pig and primates. This lack of in vivo activity was later linked to poor
absorption, low plasma concentrations, and/or high burden of extracellular mycobacteria.
Thus, for decades, its use remained limited to leprosy treatment [98].

Its exact mechanism of action remains incompletely understood but includes inhibition of
mycobacterial respiration via intracellular redox cycling as well as membrane disruption [99,
100]. Other proposed mechanisms of action include generation of reactive oxygen species,
inhibition of mycobacterial phospholipase A2, microbial K+ transport inhibition, and efflux
pump inhibition (shown in Fig. 1; Table 1) [97, 98, 99, 101, 102]. Recently, studies have
shown that upregulation of the MmpS5/MmpL5 efflux system after mutations in its repressor
gene (Rv0678) leads to increased MICs against clofazimine as well as cross-resistance with
bedaquiline [33]. In addition, mutations in the pepQ (Rv2535c) and Rv1979c genes,
encoding an aminopeptidase and a permease transporter, increase clofazimine MICs by
unknown mechanisms [33, 103, 104].

In vitro, clofazimine is highly active against M. tuberculosis complex isolates with a reported
MIC90 of 0.25 mg/L in wild-type isolates using broth microdilution [98, 105]. It took mouse
models with long treatment durations to demonstrate that clofazimine has potent, albeit
delayed and dose-independent, anti-TB activity (across the range of doses investigated) and
that clofazimine adds significant activity to first- and second-line TB treatment regimens, at
serum concentrations above the MIC [106, 107]. Clofazimine also exhibits a sustained
antimycobacterial activity after treatment cessation, yet again most prominent if serum
concentrations during treatment were ≥0.25 mg/L (the MIC90), suggesting a potential for
treatment shortening with clofazimine [108]. This treatment shortening effect was further
investigated in first-line regimens where rifampicin was replaced with rifapentine and
clofazimine was added; in two distinct mouse models of more acute (Balb/c) and caseous-
necrotic disease (C3HeB/FeJ), adding clofazimine and rifapentine increased the bactericidal
and sterilizing activity of the regimens, more than either drug alone [109]. This confirmed
the potential for treatment shortening by clofazimine-containing regimens [109].

Compared to administration during fasting, the oral bioavailability of clofazimine is


increased by ingestion with a fatty-meal, whereas aluminum/magnesium antacid has the
opposite effect [110]. Clofazimine is highly lipophilic and has peculiar PK including long
time-to-steady state, low serum concentrations (0.4–1 mg/L at 100 mg once-daily dose), high
protein binding, accumulation in skin, fatty tissues, reticuloendothelial organs and
macrophages, and a very long half-life (up to 70 days with long-term repeating dosing)
(Table 1) [99, 110]. Although clofazimine is a weak inducer of CYP3A4 itself [111], its PK
is not affected by concomitant rifamycin administration [112]. Despite its long use, the
optimal dose remains undetermined (Table 1). A 100 mg once-daily dose is standard, but a
recent modeling study showed that 200 mg once-daily loading doses in the first 2–4 weeks
shorten the time to effective steady-state concentrations by a month [113].

Clofazimine as a monotherapy showed no detectable 14-day EBA, nor did it add to EBA of
regimens with other drugs [59]; subsequent modeling revealed that it did have concentration-
dependent effects, primarily on the persister subpopulation, and therefore contributes
sterilizing activity to regimens [114]. In a randomized controlled trial, adding clofazimine to
personalized MDR-TB treatment regimens led to accelerated time-to-culture conversion and
higher treatment success rates (74% vs. 54%, p = 0.035) [115]. Subsequent meta-analyses of
individual patient data have presented conflicting conclusions on the efficacy of clofazimine
as part of MDR-TB treatment, with 2017 meta-analyses reporting no beneficial effect of
clofazimine [116]. However, a follow-up meta-analysis in 2018 reported greater treatment
success with the use of clofazimine than without it, with an adjusted risk difference of 0.06
(95% CI: 0.01–0.10) [117]. Very recent results from the TB-PRACTECAL study showed that
the bedaquiline, pretomanid, linezolid, and clofazimine regimen was effective and safe, but it
was not the most effective and safe regimen within the trial, but this has not been published
yet [88, 118]. Based on its performance in clinical trials and meta-analyses, clofazimine was
categorized as a group B drug in the 2019 WHO consolidated guidelines on MDR-TB
treatment [20]. With only three drugs in group A, it implicitly recommends adding
clofazimine to all MDR-TB regimens, where it is available. Yet, clofazimine availability is
limited and its cost further impairs access to the drug for MDR-TB treatment [119].

Clofazimine is well tolerated; in a meta-analysis, 1.6% (95% CI 0.5–5.3%) of MDR-TB


patients interrupt or stop therapy because of adverse events [120]. Adverse events associated
with clofazimine use include skin discoloration (reported in frequencies ranging from 3% to
94% [99, 115, 116, 120]). Informing patients about the possibility of clofazimine causing
reddish-black discoloration of skin and body secretions could mitigate against unnecessary
anxiety and nonadherence. Other adverse events include gastrointestinal discomfort, nausea,
vomiting, and QTc interval prolongation [99, 115, 121]. QTc interval prolongation is
important particularly when clofazimine is combined with other QTc interval prolonging
drugs, e.g., bedaquiline, fluoroquinolones, delamanid, and oxazolidinones (linezolid,
sutezolid) [99].

Optimized Drugs

Optimized Dose (High Dose) Rifampicin

Rifampicin is considered to be the cornerstone in the treatment of TB. A review concluded


that the current dose (10 mg/kg) for the short-course treatment regimen was chosen because
the drug was expensive at the time due to fear of adverse events/toxicity and because serum
concentrations were above the minimal inhibitory concentration of M. tuberculosis. Twenty
years ago, Dennis Mitchison already suggested that high dose of rifampicin should be
investigated because of an increased sterilizing effect [122].

According to a murine aerosol infection model and a hollow fiber model, rifampicin shows
concentration-dependent killing and the most adequate parameter for the killing of bacteria is
AUC/MIC [123, 124]. Also in mice, it was shown that the maximum tolerated dose was 160
mg/kg daily and a dose of 80 mg/kg per day reduced treatment duration to 9 weeks, without
adverse effects [125]. Other murine studies showed that a higher dose led to faster culture
conversion, a shortened treatment period, a decreased relapse rate, and an increase in
bactericidal and sterilizing activity [123, 126, 127].

The concentration of rifampicin increases more than proportional with dose, up to 7-fold
[128, 129, 130, 131]. Modeling and simulations showed that higher exposures of rifampicin
led to greater EBA [132]. Rifampicin induces CYP450 enzymes which leads to decreased
concentrations of many drugs, but not the other standard drugs [133, 134, 135]. There is
probably a ceiling in the maximal inductive capacity of rifampicin at relatively low doses of
rifampicin [133]. Currently, the PHENORIF study is looking at the interaction potential of
high-dose rifampicin.

A systematic review found an advantage in terms of likelihood of culture conversion in


patients that received at least 900 mg rifampicin based on historical clinical trials [136]. In
two phase 2B clinical trials, it was found that 20 mg/kg rifampicin for 2 months was safe and
tolerated by patients [137, 138]. Findings of a dose ranging trial showed that 40 mg/kg
rifampicin was safe and tolerated by patients for 2 weeks and that 50 mg/kg was poorly
tolerated [139]. In another phase 2B trial, it was found that a regimen containing 35 mg/kg
rifampicin for 12 weeks was safe and reduced time to stable culture conversion in liquid
media [134]. Increasing rifampicin doses (up to 30 mg/kg orally) and exposures resulted in a
better survival of patients with TB meningitis [130, 140], although this was not confirmed in
a larger trial with an increase of 15 mg/kg orally (Table 1) [141].

Based on a change in international guidelines in 2010, rifampicin is administered at a higher


dose (15 mg/kg) [142] in children than in adults because 10 mg/kg in children leads to lower
serum concentrations than in adults [142, 143, 144, 145, 146, 147]. The results of the SHINE
trial showed that with the revised WHO doses of all drugs a 4-month treatment regimen was
noninferior to a 6-month treatment regimen in children with regard to efficacy [148], which
has led to the recommendation of a 4-month regimen by the WHO [149]. Furthermore, high
doses of rifampicin up to 60 mg/kg in children for a period of 2 weeks were found to be safe
based on the Opti-Rif trial [150].

Suboptimal concentrations of rifampicin could induce resistance at standard doses, explained


by poor penetration into cavities [131, 151]. In one study, it was shown that rifampicin
reaches adequate concentrations in critical lesions such as necrotic caseum after multiple
doses [152]. Other drugs do not achieve adequate concentrations; thus, it was suggested that
there can be monotherapy at specific time and locations, which could lead to the emergence
of resistance [152].

Optimized doses of rifampicin have not been implemented in guidelines, and the WHO is
awaiting additional evidence, which includes phase 3 clinical trials, and a systematic review
on all available literature. A phase 2C trial is planned within the PanACEA consortium to
investigate 3- and 4-month combination regimens with isoniazid, pyrazinamide in a standard
dose and an optimized dose of rifampicin, pyrazinamide, and moxifloxacin [153].
Furthermore, a pragmatic randomized trial will be conducted in Europe to study the safety of
an optimized dose of rifampicin in an operational setting. There is an increasing amount of
evidence to suggest that the time has come to reconsider the guidelines in special situations
(meningitis, severe TB, children, diabetes mellitus, HIV, etc.).

Rifapentine
Rifapentine is a cyclopentyl-substituted semi-synthetic derivative of rifampicin. Like other
rifamycins, rifapentine selectively binds to bacterial DNA-dependent RNA polymerase, and
inhibition will occur already after short periods of drug exposure (shown in Fig. 1; Table 1).
Specific mutations in the β subunit of the RNA polymerase gene (rpoB) lead to the
development of cross-resistance to all rifamycins, including rifapentine and rifampicin [154].

The MIC against M. tuberculosis of rifapentine is approximately 0.02 μg/mL [155]. MIC and
the minimum bactericidal concentration of rifapentine for intracellular bacteria are 2- to 4-
fold lower than those of rifampicin; for extracellular bacteria, this difference was
approximately 2-fold [156]. The prolonged effect of rifapentine and its potential for
intermittent use found in this study may be associated with high intracellular accumulation,
which were 4- to 5-fold higher than those found for rifampicin [156].

PK of rifapentine has been investigated at various doses, frequencies, regimens, and


populations [157, 158, 159, 160, 161]. In general, time to Cmax is observed in 4–6 h and the
half-life is relatively long with about 12–15 h [154]. This is significantly longer than for
rifampicin (1.5–5.0 h) and supports a more intermittent use, e.g., for latent TB treatment.
Rifapentine peak exposures have been found to be dose linear [161] or less than dose linear
[159]. AUCs are less than dose-proportional [162]. The latter is in contrast to the greater than
dose-proportional PK observed for rifampicin. The rifapentine Cmax (15 μg/mL) at a dose of
600 mg sufficiently exceeds the MIC [154, 162]. However, rifapentine is about 98% plasma
protein-bound [154], which is 4- to 10-fold higher than for rifampicin (about 8–25% protein-
unbound [163]), compensating for the lower MIC of rifapentine and possibly explaining the
suboptimal efficacy of historically recommended doses of rifapentine.

Recently, a systematic review was published describing rifapentine population PK in a large


cohort of nine clinical studies [164]. The analysis showed that rifapentine bioavailability was
clinically relevant and was affected by HIV status, food, and dose. With intermittent dosing,
autoinduction of clearance was minimal to moderate. With daily dosing, maximum induction
was achieved with doses of 300 mg or more. The maximum effect was a 72% increase in
clearance after 21 days [164]. Month 2 culture conversion was found to be less likely in
individuals with low weight (<50 kg) and in those with low rifapentine exposure. Body
weight was not a clinically relevant predictor of clearance, suggesting weight-band dosing of
rifapentine should be removed from latent TB infection (LTBI) dosing guidelines [164]. Of
note, very limited PK data are available for LTBI: only one study is available that collected
PK data in LTBI in 77 participants [160].

Similar to rifampicin, daily/intermittent rifapentine administration significantly reduces


exposures of co-administered drugs [154], such as midazolam/bedaquiline (CYP3A4
substrate) [11, 165] and moxifloxacin (UGT1A1) [166]. While in vitro rifampicin seemed to
have a superior drug interaction potential [167], oral midazolam clearance was reduced to
significantly greater extent (74 vs. 93%) by rifapentine than rifampicin in vivo [159].
Induction of enzyme activities by rifapentine occurs within 4 days after the first dose,
returning to baseline levels 14 days after discontinuing rifapentine [154]. In general, less
drug-drug interactions are anticipated for once weekly dosing of rifapentine. For example,
moxifloxacin exposures remained almost unaffected when rifapentine was dosed once weekly
[168]. Rifapentine given once weekly also did not affect concentrations of efavirenz, an HIV
drug that is a CYP2B6 substrate [169].

Several rifapentine-based short courses of LTBI treatment have been evaluated to date. A 1-
month regimen of rifapentine plus isoniazid was noninferior to 9 months of isoniazid alone
for preventing TB in HIV-infected patients [170]. However, the rifapentine-based LTBI
regimen with the most extensive track record at present is 12 weeks weekly
rifapentine/isoniazid (3HP). Multiple large-scale clinical trials and analyses have shown that
3HP is noninferior to standard INH and has significantly higher rates of treatment completion
and lower rates of adverse events [171, 172, 173, 174]. Completion of 3HP in routine
healthcare settings seems even greater than rates reported from clinical trials, and greater
than historically observed using other regimens among reportedly nonadherent populations
[175].

For pulmonary TB treatment, more frequent rifapentine administrations seem desirable, as


once weekly or less frequent use of rifapentine increases the risk of bacteriological relapse
compared to intermittent use of rifampicin [176]. In phase 2 clinical trials, no obvious safety
concerns have been noted with the use of daily rifapentine during the first 8 weeks of
combination therapy for pulmonary TB [177], and increasing the PK exposure to rifapentine
was associated with improved sterilization at the end of the intensive phase as compared to
standard dose rifampicin in mice and humans [177, 178]. In a pivotal, ground-breaking
recent phase III trial the threshold of shortening to 4 months was finally reached: the efficacy
of this 4-month rifapentine-based regimen containing moxifloxacin was noninferior to the
standard 6-month regimen in the treatment of TB, and it was similar in regard to safety [179].
This was the first study supporting the treatment shortening potential of a rifamycin when
combined with moxifloxacin. The WHO now conditionally recommends the use of a 4-
month regimen consisting of rifapentine, isoniazid, pyrazinamide, and moxifloxacin in
patients with drug-susceptible TB that are 12 years or older (Table 1) [149].

PAS

PAS, first discovered in 1902 by Seidel and Bittner, was rediscovered in 1943 by Jorgen
Lehmann, a Swedish clinician-scientist [180, 181]. PAS is arguably the first synthetic drug
used in the clinic to successfully treat TB patients in 1944 [182, 183, 184].

The precise mechanism of action of PAS remains elusive, and PAS is considered an
antimetabolite, which is structurally similar to para-aminobenzoic acid, a substrate of
dihydropteroate synthase [185, 186]. In turn, the generation of hydroxyl dihydrofolate
antimetabolite in turn inhibits dihydrofolate reductase [186]. An additional potential PAS
mechanism of action is its inhibition of mycobactin, a component of the mycobacterial cell
wall resulting in reduced iron uptake by M. tuberculosis (Fig. 1; Table 1) [187]. A PAS MIC
≤1 mg/L defines susceptibility in clinical isolates [188, 189]. Concentration-dependent
activity of PAS was demonstrated in an in vitro study, in which PAS concentration of 100
mg/L was decidedly better than 10 mg/L in suppressing resistance emergence in the
companion drugs streptomycin and isoniazid [182, 190]. Animal models of TB show the
dose and concentration-dependent bacteriostatic effects of PAS [182, 191].

The PK of PAS varies with the formulation. PAS salts are readily absorbed when
administered orally and achieve higher Cmax in plasma than with PAS acid or the widely
available granular slow-release PAS formulation (PASER) [188, 192, 193, 194]. Under the
influence of N-acetyltransferase 1, PAS undergoes first-pass metabolism to acetyl-PAS in the
gut and liver, which might explain acetyl-PAS appearance in blood earlier than PAS [181,
195]. PAS is metabolized to glycine-PAS in the liver [195]. N-acetyltransferase 1 and N-
acetyltransferase 2 genetic polymorphisms have been recognized to influence PAS clearance
[182, 196]. Approximately 80% of the administered dose is excreted in urine as PAS and its
two major metabolites, acetyl-PAS and glycine-PAS [181, 195]. PAS has an elimination half-
life of 0.5–2.5 h and a modest plasma protein binding [181, 187, 193, 197].

Oral administration of PASER with food increases the oral bioavailability of PAS, Cmax, and
overall systemic exposure, while reducing intolerance particularly when taken with acidic
beverages [182, 197]. The gastrointestinal intolerance to PAS is primarily because of its
direct irritant effect while in the gastrointestinal tract and PAS decarboxylation to meta-
aminophenol toxin [182, 187].

The early appreciation that gastrointestinal intolerance to PAS could be formulation related
led to the development of many formulations [181, 182, 198]. With improving intolerance in
mind, PASER is enteric-coated and designed to slowly release PAS in the intestine rather
than in the stomach [187]. Furthermore, the gastrointestinal intolerance is neither related to
plasma concentrations nor to the conjugated metabolites [182, 194, 196, 199, 200].

Clinical trials conducted in the 1950s by the British Medical Research Council established
the role of PAS in suppressing resistance emergence in companion drugs, allowing the
construction of an efficacious anti-TB regimen for the next three decades [201, 202, 203,
204]. This ability to suppress resistance emergence is clearly dose-dependent, with 20 g/day
sodium PAS outperforming 10 g/day and 5 g/day doses administered in four divided doses
[203]. A subsequent study found that a sodium PAS dose of 20 g/day has no advantage over
10 g/day in the ability of PAS to suppress resistance emergence to isoniazid [204]. It is
however important to note that the individual PAS dosage administered in the study was 5 g
because 20 g/day and 10 g/day were administered in 4 and 2 divided doses, respectively.
These findings and preclinical evidence of the PAS concentration-dependent effect could
indicate that Cmax and, by extension, Cmax/MIC are the parameters linked to the prevention of
resistance in companion drugs [190, 202, 203, 204].

In addition, an EBA study showed PAS might have a bactericidal effect if used at a high
enough dose and, consequently, a high Cmax could be reached with a single-daily dose [205].
In this study, 15 g once-daily PAS resulted in a 2-day EBA similar to that of once-daily 10
mg/kg rifampicin [205]. Furthermore, two clinical studies conducted in the early days of PAS
use provided an early indication of the potential of PAS to be a bactericidal agent [206, 207].

The Cmax/MIC ratio has been suggested to be linked to PAS' ability to prevent resistance
emergence in companion drugs; if that hypothesis is correct, the plasma PAS concentrations
reached with PASER are low [182, 188, 196, 208] and maybe inadequate at the current
dosing regimen of 12 g/day in 2–3 divided doses. Therefore, there remains an urgent need for
a prospective EBA study with a PK component to evaluate different once daily PASER
regimens. This prospective study should clarify the most appropriate PK/pharmacodynamics
determinant of PAS efficacy.

In its 2019 MDR-TB treatment guideline, the WHO downgraded the use of PAS (group C) to
only when there is resistance or toxicity to the more preferred anti-TB agents (group A and
B) [20]. A meta-analysis of observational studies at least in part was responsible for the
WHO recommendation regarding PAS use [117]. The limitations inherent in observational
studies strengthen the need for prospective studies to clearly outline the use of PAS, as we are
likely not using PASER optimally. Thus, to win the fight against the DR-TB epidemic, all
drugs including PAS in a regimen must be optimally utilized, especially in the face of
resistance emergence in newly approved drugs such as bedaquiline and delamanid.

Conclusion

We have arrived at a promising stage in the fight against TB, with several academic,
governmental, and nongovernmental organizations facilitating and accelerating TB drug
development. There have been recommendations and communication of new treatment
regimens, with progress been made in treatment shortening for DS- and DR-TB. Moreover,
currently new drugs with new targets and existing or repurposed drugs are investigated in
clinical trials, with another series of new and repurposed drugs in earlier stages of
development. This review highlighted a selection of clinically available drug candidates for
current and future TB regimens. Nevertheless, studies are still ongoing to further investigate
these drugs and their optimal application. The most recent developments are a shortened
treatment regimen for DS-TB in guidelines and recent guidelines and a rapid communication
for DR-TB, including new definitions of multi- and extensive drug resistance. For DS-TB, a
short (4 months) rifapentine and moxifloxacin-based regimen has now been adopted in
guidelines for people of 12 years or older [149]. Optimized (high dose) dose rifampicin is
already implemented in severe cases of TB in some clinical settings and is currently being
investigated in a study with a large sample size. Furthermore, for children with nonsevere TB
a short (4 months) treatment regimen has been recommended [149]. The newest WHO rapid
communication on DR-TB now stresses the importance of bedaquiline, pretomanid,
linezolid, and, to a lesser extent, clofazimine [19]; for rifampicin- and MDR-TB, the (6
months) BPaLM regimen is endorsed, and if there is resistance to fluoroquinolones it can be
used without moxifloxacin [19].

Newer oxazolidinones with a lower toxicity profile than linezolid are being studied.
Moreover, a universal regimen for both DS- and DR-TB is also currently being investigated,
the BPaMZ regimen was meant to become such a regimen, although this could be questioned
considering the rapid emergence of resistance to bedaquiline and the current resistance to
quinolones and pyrazinamide.

In this review, we have outlined various anti-TB drugs that to a greater or lesser extent could
potentially contribute to improved treatment regimens. Some drugs are new, some are
repurposed, and some are optimized with increased dosages. In order to achieve the targets of
the End TB strategy [209], increasing efforts still need to be made of which the development
of new treatment regimens for active (and latent) TB is essential.

Conflict of Interest Statement

The authors have no conflicts of interest to declare.

Funding Sources

This research did not receive grants from any funding agency in the public, commercial, or
not-for-profit sectors.

Author Contributions

Jessica Maria Aguilar Diaz: abstract, introduction, section about an optimized dose of
rifampicin, conclusion, table, adaptation figure, lay-out article, general comments, references,
and end-editing. Ahmed Aliyu Abulfathi: section about para-aminosalicylic acid, English
edits, general comments. Lindsey Hendrika Maria te Brake: abstract, introduction, section
about rifapentine, conclusion, and general comments. Jakko van Ingen: section about
clofazimine, general comments, and conclusion. Saskia Kuipers: section about linezolid and
general comments. Cecile Magis-Escurra: section about delamanid and general comments.
Jelmer Raaijmakers: references and general comments. Elin Margareta Svensson: section
about bedaquiline and general comments. Martin Johan Boeree: abstract, introduction,
section about pretomanid, conclusion, and end-editing.
Funding Statement

This research did not receive grants from any funding agency in the public, commercial, or
not-for-profit sectors.

References

1. Andries K, Verhasselt P, Guillemont J, Göhlmann HWH, Neefs JM, Winkler H, et al. A diarylquinoline drug
active on the ATP synthase of Mycobacterium tuberculosis. Science. 2005;307((5707)):223–227. [PubMed]
[Google Scholar]

2. Hoelscher M. Netherlands: Nijmegen; 2022. 24 March, Presentation TB treatment-new drugs, ESCMID


Postgraduate Course, Diagnosis and treatment of mycobacterial infections; p. 2022. [Google Scholar]

3. Koul A, Dendouga N, Vergauwen K, Molenberghs B, Vranckx L, Willebrords R, et al. Diarylquinolines target


subunit c of mycobacterial ATP synthase. Nat Chem Biol. 2007;3((6)):323–324. [PubMed] [Google Scholar]

4. World Health Organization Technical Report on critical concentrations for drug susceptibility testing of
medicines used in the treatment of drug-resistant tuberculosis [WHO/CDS/TB/2018.5]
http://apps.who.int/iris/bitstream/10665/260470/1/WHO-CDS-TB-2018.5-eng.pdf [Accessed 2 July 20202018]

5. Rouan MC, Lounis N, Gevers T, Dillen L, Gilissen R, Raoof A, et al. Pharmacokinetics and pharmacodynamics
of TMC207 and its N-desmethyl metabolite in a murine model of tuberculosis. Antimicrob Agents Chemother.
2012;56((3)):1444–1451. [PMC free article] [PubMed] [Google Scholar]

6. Zhang T, Li SY, Williams KN, Andries K, Nuermberger EL. 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–
737. [PMC free article] [PubMed] [Google Scholar]

7. van Heeswijk RPG, Dannemann B, Hoetelmans RMW. Bedaquiline a review of human pharmacokinetics and
drug–drug interactions. J Antimicrob Chemother. 2014;69((9)):2310–2318. [PubMed] [Google Scholar]

8. Brill MJ, Svensson EM, Pandie M, Maartens G, Karlsson MO. 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 Feb;49((2)):212–217. [PubMed] [Google Scholar]

9. Svensson EM, Aweeka F, Park J-G, Marzan F, Dooley KE, Karlsson MO. 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)):2780–2787. [PMC free article] [PubMed] [Google
Scholar]

10. Svensson EM, Dooley KE, Karlsson MO. Impact of lopinavir/ritonavir or nevirapine on bedaquiline exposures
potential implications for patients with TB/HIV co-infection. Antimicrob Agents Chemother. 2014;58((11)):6406–
6412. [PMC free article] [PubMed] [Google Scholar]
11. Svensson EM, Murray S, Karlsson MO, Dooley KE. Rifampicin and rifapentine significantly reduce
concentrations of bedaquiline a new anti-TB drug. J Antimicrob Chemother. 2015;70((4)):1106–1114. [PMC free
article] [PubMed] [Google Scholar]

12. Diacon AH, Dawson R, von Groote-Bidlingmaier F Symons G, Venter A, Donald PR, 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)):2199–2203.
[PMC free article] [PubMed] [Google Scholar]

13. Diacon AH, Pym A, Grobusch MP, De los Rios JM Gotuzzo E, Vasilyeva I, et al. Multidrug-resistant
tuberculosis and culture conversion with bedaquiline. N Engl J Med. 2014;371((8)):723–732. [PubMed] [Google
Scholar]

14. Svensson EM, Karlsson MO. Modelling of mycobacterial load reveals bedaquiline's exposure-response
relationship in patients with drug-resistant, TB. J Antimicrob Chemother. 2017;72((12)):3398–3405. [PMC free
article] [PubMed] [Google Scholar]

15. Tanneau L, Svensson EM, Rossenu S, Karlsson MO. Exposure-safety analysis of QTc interval and
transaminase levels following bedaquiline administration in patients with drug-resistant tuberculosis. CPT
Pharmacometrics Syst Pharmacol. 2021;10((12)):1538–1549. [PMC free article] [PubMed] [Google Scholar]

16. Dooley KE, Rosenkranz SL, Conradie F, Moran L, Hafner R, von Groote-Bidlingmaier F, et al. QT effects of
bedaquiline or both in patients with rifampicin-resistant tuberculosis a phase 2, open-label, randomised, controlled
trial. Lancet Infect Dis. 2021;21((7)):975–983. [PMC free article] [PubMed] [Google Scholar]

17. Nunn AJ, Rusen ID, van Deun A Torrea G, Phillips PPJ, Chiang CY, et al. Evaluation of a standardized
treatment regimen of anti-tuberculosis drugs for patients with multi-drug-resistant tuberculosis (STREAM) study
protocol for a randomized controlled trial. Trials. 2014;15:353. [PMC free article] [PubMed] [Google Scholar]

18. Conradie F, Diacon AH, Ngubane N, Howell P, Everitt D, Crook AM, et al. Treatment of highly drug-resistant
pulmonary tuberculosis. N Engl J Med. 2020;382((10)):893–902. [PMC free article] [PubMed] [Google Scholar]

19. World Health Organization Rapid communication key changes to the treatment of drug-resistant tuberculosis.
World Health Organization. 2022.

20. World Health Organization Consolidated guidelines on drug-resistant tuberculosis treatment. 2019.
[WHO/CDS/TB/2019.7] [PubMed]

21. Garcia-Prats AJ, Svensson EM, Weld ED, Schaaf HS, Hesseling AC. Current status of pharmacokinetic and
safety studies of multidrug-resistant tuberculosis treatment in children. Int J Tuberc Lung Dis. 2018;22((5)):15–
23. [PubMed] [Google Scholar]

22. World Health Organization WHO consolidated guidelines on tuberculosis module 5: management of
tuberculosis in children and adolescents [PubMed]

23. Gao M, Gao J, Xie L, Wu G, Chen W, Chen Y, et al. Early outcome and safety of bedaquiline-containing
regimens for treatment of MDR- and XDR-TB in China a multicentre study. Clin Microbiol Infect.
2021;27((4)):597–602. [PubMed] [Google Scholar]
24. Barvaliya SV, Desai MK, Panchal JR, Solanki RN. Early treatment outcome of bedaquiline plus optimised
background regimen in drug resistant tuberculosis patients. Indian J Tuberc. 2020;67((2)):222–230. [PubMed]
[Google Scholar]

25. Kang H, Jo KW, Jeon D, Yim JJ, Shim TS. Interim treatment outcomes in multidrug-resistant tuberculosis
using bedaquiline and/or delamanid in South Korea. Respir Med. 2020;167:105956. [PubMed] [Google Scholar]

26. Mohr-Holland E, Reuter A, Furin J, Garcia-Prats A, De Azevedo V, Mudaly V, et al. Injectable-free regimens
containing bedaquiline, delamanid, or both for adolescents with rifampicin-resistant tuberculosis in Khayelitsha,
South Africa. EClinicalMedicine. 2020;20:100290. [PMC free article] [PubMed] [Google Scholar]

27. Kempker RR, Mikiashvili L, Zhao Y, Benkeser D, Barbakadze K, Bablishvili N, et al. Clinical outcomes
among patients with drug-resistant tuberculosis receiving bedaquiline or delamanid containing regimens. Clin
Infect Dis. 2019 [PMC free article] [PubMed] [Google Scholar]

28. Olayanju O, Esmail A, Limberis J, Dheda K. A regimen containing bedaquiline and delamanid compared to
bedaquiline in patients with drug-resistant tuberculosis. Eur Respir J. 2020;55((1)):1901181. [PubMed] [Google
Scholar]

29. Mbuagbaw L, Guglielmetti L, Hewison C, Bakare N, Bastard M, Caumes E, et al. Outcomes of bedaquiline
treatment in patients with multidrug-resistant tuberculosis. Emerg Infect Dis. 2019;25((5)):936–943. [PMC free
article] [PubMed] [Google Scholar]

30. Loveday M, Hughes J, Sunkari B, Master I, Hlangu S, Reddy T, et al. Maternal and infant outcomes among
pregnant women treated for multidrug/rifampicin-resistant tuberculosis in South Africa. Clin Infect Dis. 2020
[PMC free article] [PubMed] [Google Scholar]

31. Akkerman OW, Odish OF, Bolhuis MS, De Lange WC Kremer HP, Luijckx GJ. Pharmacokinetics of
bedaquiline in cerebrospinal fluid and serum in multidrug-resistant tuberculous meningitis. Clin Infect Dis.
2016;62((4)):523–524. [PubMed] [Google Scholar]

32. Upton CM, Steele CI, Maartens G, Diacon AH, Wiesner L, Dooley KE. Pharmacokinetics of bedaquiline in
cerebrospinal fluid (CSF) in patients with pulmonary tuberculosis (TB) J Antimicrob Chemother.
2022;77((6)):1720–1724. [PMC free article] [PubMed] [Google Scholar]

33. Kadura S, King N, Nakhoul M, Zhu H, Theron G, Koser CU, et al. Systematic review of mutations associated
with resistance to the new and repurposed Mycobacterium tuberculosis drugs bedaquiline delamanid and
pretomanid. J Antimicrob Chemother. 2020;75((8)):2031–2043. [PMC free article] [PubMed] [Google Scholar]

34. 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)):2979–2981.
[PMC free article] [PubMed] [Google Scholar]

35. Somoskovi A, Bruderer V, Homke R, Bloemberg GV, Bottger EC. A mutation associated with clofazimine and
bedaquiline cross-resistance in MDR-TB following bedaquiline treatment. Eur Respir J. 2015;45((2)):554–557.
[PubMed] [Google Scholar]
36. Matsumoto M, Hashizume H, Tomishige T, Kawasaki M, Tsubouchi H, Sasaki H, et al. OPC-67683 a nitro-
dihydro-imidazooxazole derivative with promising action against tuberculosis in vitro and in mice. PLoS Med.
2006;3((11)):e466. [PMC free article] [PubMed] [Google Scholar]

37. Liu Y, Matsumoto M, Ishida H, Ohguro K, Yoshitake M, Gupta R, et al. Delamanid from discovery to its use
for pulmonary multidrug-resistant tuberculosis (MDR-TB) Tuberculosis. 2018;111:20–30. [PubMed] [Google
Scholar]

38. Stinson K, Kurepina N, Venter A, Fujiwara M, Kawasaki M, Timm J, et al. MIC of delamanid (OPC-67683)
against Mycobacterium tuberculosis clinical isolates and a proposed critical concentration. Antimicrob Agents
Chemother. 2016;60((6)):3316–3322. [PMC free article] [PubMed] [Google Scholar]

39. Diacon AH, Dawson R, Hanekom M, Narunsky K, Venter A, Hittel N, et al. Early bactericidal activity of
delamanid (OPC-67683) in smear-positive pulmonary tuberculosis patients. Int J Tuberc Lung Dis.
2011;15((7)):949–954. [PubMed] [Google Scholar]

40. Sasahara K, Shimokawa Y, Hirao Y, Koyama N, Kitano K, Shibata M, 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)):1267–1276. [PubMed] [Google Scholar]

41. Mallikaarjun S, Wells C, Petersen C, Paccaly A, Shoaf SE, Patil S, et al. Delamanid coadministered with
antiretroviral drugs or antituberculosis drugs shows No clinically relevant drug-drug interactions in healthy
subjects. Antimicrob Agents Chemother. 2016;60((10)):5976–5985. [PMC free article] [PubMed] [Google
Scholar]

42. Bahuguna A, Rawat DS. An overview of new antitubercular drugs drug candidates and their targets. Med Res
Rev. 2020;40((1)):263–292. [PubMed] [Google Scholar]

43. Gler MT, Skripconoka V, Sanchez-Garavito E, Xiao H, Cabrera-Rivero JL, Vargas-Vasquez DE, et al.
Delamanid for multidrug-resistant pulmonary tuberculosis. N Engl J Med. 2012;366((23)):2151–2160. [PubMed]
[Google Scholar]

44. von Groote-Bidlingmaier F Patientia R, Sanchez E, Balanag V, Jr, Ticona E, Segura P, 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;7((3)):249–259. [PubMed] [Google Scholar]

45. Hewison C, Ferlazzo G, Avaliani Z, Hayrapetyan A, Jonckheere S, Khaidarkhanova Z, et al. Six-month


response to delamanid treatment in MDR TB patients. Emerg Infect Dis. 2017;23((10)):1746–1748. [PMC free
article] [PubMed] [Google Scholar]

46. Pontali E, Sotgiu G, Tiberi S, Tadolini M, Visca D, D'Ambrosio L, et al. Combined treatment of drug-resistant
tuberculosis with bedaquiline and delamanid a systematic review. Eur Respir J. 2018;52((1)):1800934. [PubMed]
[Google Scholar]

47. World Health Organization WHO consolidated guidelines on tuberculosis module 4: treatment: drug-resistant
tuberculosis treatment World Health Organization. 2020. [PubMed]
48. Li Y, Sun F, Zhang W. Bedaquiline and delamanid in the treatment of multidrug-resistant tuberculosis
promising but challenging. Drug Dev Res. 2019;80((1)):98–105. [PMC free article] [PubMed] [Google Scholar]

49. The Global Alliance for TB Drug Developmet (TB Alliance) Pretomanid tablets US prescribing information.
[Available from: https://www.fda.gov/

50. Food and Drug Administration FDA approves new drug for treatment-resistant forms of tuberculosis that
affects the lungs. 2019.

51. Pretomanid EM. FGK (pretomanid) an overview of Pretomanid FGK and why it is authorised in the EU
European Medicines Agency. 2020 [Google Scholar]

52. Manjunatha U, Boshoff HI, Barry CE. The mechanism of action of PA-824 novel insights from transcriptional
profiling. Commun Integr Biol. 2009;2((3)):215–218. [PMC free article] [PubMed] [Google Scholar]

53. Singh R, Manjunatha U, Boshoff HI, Ha YH, Niyomrattanakit P, Ledwidge R, et al. PA-824 kills
nonreplicating Mycobacterium tuberculosis by intracellular NO release. Science. 2008;322((5906)):1392–1395.
[PMC free article] [PubMed] [Google Scholar]

54. Lenaerts AJ, Gruppo V, Marietta KS, Johnson CM, Driscoll DK, Tompkins NM, 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)):2294–2301. [PMC free article] [PubMed] [Google Scholar]

55. Xu J, Li SY, Almeida DV, Tasneen R, Barnes-Boyle K, Converse PJ, et al. Contribution of pretomanid to novel
regimens containing bedaquiline with either linezolid or moxifloxacin and pyrazinamide in murine models of
tuberculosis. Antimicrob Agents Chemother. 2019;63((5)):e00021–e00119. [PMC free article] [PubMed] [Google
Scholar]

56. Feuerriegel S, Köser CU, Baù D, Rüsch-Gerdes S, Summers DK, Archer JAC, et al. Impact of Fgd1 and ddn
diversity in Mycobacterium tuberculosis complex on in vitro susceptibility to PA-824. Antimicrob Agents
Chemother. 2011;55((12)):5718–5722. [PMC free article] [PubMed] [Google Scholar]

57. Haver HL, Chua A, Ghode P, Lakshminarayana SB, Singhal A, Mathema B, et al. Mutations in genes for the
F420 biosynthetic 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)):5316–5323. [PMC free article] [PubMed] [Google Scholar]

58. Dooley KE, Luetkemeyer AF, Park JG, Allen R, Cramer Y, Murray S, et al. Phase I safety and
pharmacogenetics study of the antituberculosis drug PA-824 with concomitant lopinavir-ritonavir or rifampin.
Antimicrob Agents Chemother. 2014;58((9)):5245–5252. [PMC free article] [PubMed] [Google Scholar]

59. Diacon AH, Dawson R, von Groote-Bidlingmaier F Symons G, Venter A, Donald PR, 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)):943–953. [PubMed] [Google Scholar]

60. Dawson R, Diacon AH, Everitt D, van Niekerk C Donald PR, Burger DA, et al. Efficiency and safety of the
combination of moxifloxacin pretomanid (PA-824) and pyrazinamide during the first 8 weeks of antituberculosis
treatment a phase 2b, open-label, partly randomised trial in patients with drug-susceptible or drug-resistant
pulmonary tuberculosis. Lancet. 2015;385((9979)):1738–1747. [PubMed] [Google Scholar]

61. Tweed CD, Dawson R, Burger DA, Conradie A, Crook AM, Mendel CM, et al. Bedaquiline and pyrazinamide
during the first 8 weeks of treatment of patients with drug-susceptible or drug-resistant pulmonary tuberculosis a
multicentre, open-label, partially randomised, phase 2b trial. Lancet Respir Med. 2019;7((12)):1048–1058. [PMC
free article] [PubMed] [Google Scholar]

62. Clinical trial results offer hope to DR-TB patients with short, effective treatment [press release] London. 20-10-
2021 2021.

63. Ignatius EH, Abdelwahab MT, Hendricks B, Gupte N, Narunsky K, Wiesner L, et al. Pretomanid
pharmacokinetics in the presence of rifamycins interim results from a randomized trial among patients with
tuberculosis. Antimicrob Agents Chemother. 2021;65((2)):e01196–e01220. [PMC free article] [PubMed] [Google
Scholar]

64. Nedelman JR, Salinger DH, Subramoney V, Woolson R, Wade K, Li M, et al. An exposure-response
perspective on the clinical dose of pretomanid. Antimicrob Agents Chemother. 2020;65((1)):e01121–e01220.
[PMC free article] [PubMed] [Google Scholar]

65. Slee AM, Wuonola MA, McRipley RJ, Zajac I, Zawada MJ, Bartholomew PT. Oxazolidinones a new class of
synthetic antibacterial agents in vitro and in vivo activities of DuP 105 and DuP 721. Antimicrob Agents
Chemother. 1987;31((11)):1791–1797. [PMC free article] [PubMed] [Google Scholar]

66. Rich M, Jaramillo E. Guidelines for the programmatic management of drug-resistant tuberculosis. World
Health Organization. 2006 [PubMed] [Google Scholar]

67. Barbachyn MR, Hutchinson DK, Brickner SJ, Cynamon MH, Kilburn JO, Klemens SP, et al. Identification of a
novel oxazolidinone (U-100480) with potent antimycobacterial activity. J Med Chem. 1996;39((3)):680–685.
[PubMed] [Google Scholar]

68. De Vriese AS Coster RV, Smet J, Seneca S, Lovering A, van Haute LL, et al. Linezolid-induced inhibition of
mitochondrial protein synthesis. Clin Infect Dis. 2006;42((8)):1111–1117. [PubMed] [Google Scholar]

69. Kloss P, Xiong L, Shinabarger DL, Mankin AS. Resistance mutations in 23 S rRNA identify the site of action
of the protein synthesis inhibitor linezolid in the ribosomal peptidyl transferase center. J Mol Biol.
1999;294((1)):93–101. [PubMed] [Google Scholar]

70. Clinical and Laboratory Standards Institute (CLSI) Wayne M24-A2. 2nd ed. Susceptibility testing of
mycobacteria and other aerobic actinomycetes; p. p. PA2011. [PubMed] [Google Scholar]

71. World Health Organization Technical report on critical concentrations for drug susceptibility testing of
medicines used in the treatment of drug-resistant tuberculosis World Health Organization. 2018. Report No.
WHO/CDS/TB/2018.5.

72. EUCAST Linezolid/Mycobacterium tuberculosis MIC distribution reference database. Available from
http://www.eucast.org.
73. Alcalá L, Ruiz-Serrano MJ, Pérez-Fernández Turégano C, García de Viedma D, Díaz-Infantes M, Marín-
Arriaza M. in vitro activities of linezolid against clinical isolates of Mycobacterium tuberculosis that are
susceptible or resistant to first-line antituberculous drugs. Antimicrob Agents Chemother. 2003;47((1)):416–417.
[PMC free article] [PubMed] [Google Scholar]

74. Cynamon MH, Klemens SP, Sharpe CA, Chase S. Activities of several novel oxazolidinones against
Mycobacterium tuberculosis in a murine model. Antimicrob Agents Chemother. 1999;43((5)):1189–1191. [PMC
free article] [PubMed] [Google Scholar]

75. Drusano GL, Neely MN, Yamada WM, Maynard M, Duncanson B, Brown D. The combination of fosfomycin
plus meropenem is synergistic for Pseudomonas aeruginosa PAO1 in a hollow-fiber infection model. Antimicrob
Agents Chemother. 2018;62((12)):e01682–e01718. [PMC free article] [PubMed] [Google Scholar]

76. Villani P, Regazzi MB, Marubbi F, Viale P, Pagani L, Cristini F, et al. Cerebrospinal fluid linezolid
concentrations in postneurosurgical central nervous system infections. Antimicrob Agents Chemother.
2002;46((3)):936–937. [PMC free article] [PubMed] [Google Scholar]

77. Conte JE, Golden JA, Kipps J, Zurlinden E. Intrapulmonary pharmacokinetics of linezolid. Antimicrob Agents
Chemother. 2002;46((5)):1475–1480. [PMC free article] [PubMed] [Google Scholar]

78. Li Y, Huang H, Dong W, Lan T, Fan J, Wen S, et al. Penetration of linezolid into bone tissue 24 h after
administration in patients with multidrug-resistant spinal tuberculosis. PLoS One. 2019;14((10)):e0223391. [PMC
free article] [PubMed] [Google Scholar]

79. Stalker DJ, Jungbluth GL. Clinical pharmacokinetics of linezolid a novel oxazolidinone antibacterial. Clin
Pharmacokinet. 2003;42((13)):1129–1140. [PubMed] [Google Scholar]

80. Jungbluth GL, Welshman IR, Hopkins NK. Linezolid pharmacokinetics in pediatric patients an overview.
Pediatr Infect Dis J. 2003;22((9 Suppl l)):S153–S157. [PubMed] [Google Scholar]

81. Slatter JG, Stalker DJ, Feenstra KL, Welshman IR, Bruss JB, Sams JP, et al. Pharmacokinetics and excretion of
linezolid following an oral dose of [14C] linezolid to healthy human subjects. Drug Metab Dispos.
2001;29((8)):1136–1145. [PubMed] [Google Scholar]

82. Alffenaar JW, Kosterink JG, van Altena R, van der Werf TS, Uges DR, Proost JH. Limited sampling strategies
for therapeutic drug monitoring of linezolid in patients with multidrug-resistant tuberculosis. Ther Drug Monit.
2010;32((1)):97–101. [PubMed] [Google Scholar]

83. Srinivas NR, Syed M. Applicability of a single time point strategy for the prediction of area under the
concentration curve of linezolid in patients superiority of ctrough- over cmax-derived linear regression models.
Drugs R D. 2016;16((1)):69–79. [PMC free article] [PubMed] [Google Scholar]

84. Dietze R, Hadad DJ, McGee B, Molino LP, Maciel EL, Peloquin CA, et al. Early and extended early
bactericidal activity of linezolid in pulmonary tuberculosis. Am J Respir Crit Care Med. 2008;178((11)):1180–
1185. [PMC free article] [PubMed] [Google Scholar]

85. Lee M, Lee J, Carroll MW, Choi H, Min S, Song T, et al. Linezolid for treatment of chronic extensively drug-
resistant tuberculosis. N Engl J Med. 2012;367((16)):1508–1518. [PMC free article] [PubMed] [Google Scholar]
86. Esmail A, Oelofse S, Lombard C, Perumal R, Mbuthini L, Goolam Mahomed A, et al. An all-oral 6-month
regimen for multidrug-resistant tuberculosis a multicenter, randomized controlled clinical trial (the NExT study)
Am J Respir Crit Care Med. 2022;205((10)):1214–1227. [PubMed] [Google Scholar]

87. Conradie F, Everitt D, Olugbosi M, Wills G, Fabiane S, Timm J, et al. High rate of successful outcomes
treating highly resistant TB in the ZeNix study of pretomanid bedaquiline and alternative doses and durations of
linezolid. J Int AIDS Soc. 2021;24((S4)):70–71. [Google Scholar]

88. Nyang'wa B, Kazounis E, Motta I, Dodd M, Fielding K, Berry C. TB-PRACTECAL results 24 week all-oral
regimens for rifampicin resistant tuberculosis (CROI Abstract 79) Special issue: abstracts from the 2022
conference on retroviruses and opportunistic Infections Top Antiv Med. 2022 [Google Scholar]

89. Lee M, Mok J, Kim DK, Shim TS, Koh W-J, Jeon D, et al. Delamanid and pyrazinamide for the treatment of
patients with fluoroquinolone-sensitive multidrug-resistant tuberculosis (Treatment Shortening of MDR-TB Using
Existing and New Drugs, MDR-END) study protocol for a phase II/III, multicenter, randomized, open-label
clinical trial. Trials. 2019;20((1)):57–110. [PMC free article] [PubMed] [Google Scholar]

90. Sotgiu G, Centis R, D'Ambrosio L, Alffenaar J-WC, Anger HA, Caminero JA, et al. Efficacy safety and
tolerability of linezolid containing regimens in treating MDR-TB and XDR-TB systematic review and meta-
analysis. Eur Respir J. 2012;40((6)):1430–1442. [PubMed] [Google Scholar]

91. Imperial MZ, Nedelman JR, Conradie F, Savic RM. Proposed linezolid dosing strategies to minimize adverse
events for treatment of extensively drug-resistant tuberculosis. Clin Infect Dis. 2022;74((10)):1736–1747. [PMC
free article] [PubMed] [Google Scholar]

92. Bolhuis MS, Tiberi S, Sotgiu G, De Lorenzo S Kosterink JG, van der Werf TS, et al. Linezolid tolerability in
multidrug-resistant tuberculosis a retrospective study. Eur Respir J. 2015;46((4)):1205–1207. [PubMed] [Google
Scholar]

93. Wasserman S, Brust JCM, Abdelwahab MT, Little F, Denti P, Wiesner L, et al. Linezolid toxicity in patients
with drug-resistant tuberculosis a prospective cohort study. J Antimicrob Chemother. 2022;77((4)):1146–1154.
[PMC free article] [PubMed] [Google Scholar]

94. Richter E, Rüsch-Gerdes S, Hillemann D. First linezolid-resistant clinical isolates of Mycobacterium


tuberculosis. Antimicrob Agents Chemother. 2007;51((4)):1534–1536. [PMC free article] [PubMed] [Google
Scholar]

95. Wasserman S, Louw G, Ramangoaela L, Barber G, Hayes C, Omar SV, et al. Linezolid resistance in patients
with drug-resistant TB and treatment failure in South Africa. J Antimicrob Chemother. 2019;74((8)):2377–2384.
[PMC free article] [PubMed] [Google Scholar]

96. McNeil MB, Dennison DD, Shelton CD, Parish T. In vitro isolation and characterization of oxazolidinone-
resistant Mycobacterium tuberculosis. Antimicrob Agents Chemother. 2017;61((10)):e01296–e01317. [PMC free
article] [PubMed] [Google Scholar]

97. Barry V, Belton J, Conalty M, Denneny J, Edward DO, sulli-Van JF, et al. A New series of phenazines (rimino-
compounds) with high antituberculosis activity. Nature. 1957;179((4568)):1013–1015. [PubMed] [Google
Scholar]
98. Reddy VM, Nadadhur G, Daneluzzi D, O'Sullivan JF, Gangadharam PR. Antituberculosis activities of
clofazimine and its new analogs B4154 and B4157. Antimicrob Agents Chemother. 1996;40((3)):633–636. [PMC
free article] [PubMed] [Google Scholar]

99. Cholo MC, Mothiba MT, Fourie B, Anderson R. Mechanisms of action and therapeutic efficacies of the
lipophilic antimycobacterial agents clofazimine and bedaquiline. J Antimicrob Chemother. 2017;72((2)):338–353.
[PubMed] [Google Scholar]

100. Beites T, O'Brien K, Tiwari D, Engelhart CA, Walters S, Andrews J, et al. Plasticity of the Mycobacterium
tuberculosis respiratory chain and its impact on tuberculosis drug development. Nat Commun. 2019;10((1)):4970–
4912. [PMC free article] [PubMed] [Google Scholar]

101. Brunton LL, Chabner B, Knollmann BC. New York, NY, USA: McGraw-Hill Education; 2018. Goodman &
Gilman's the pharmacological basis of therapeutics. [Google Scholar]

102. Swanson RV, Adamson J, Moodley C, Ngcobo B, Ammerman NC, Dorasamy A, et al. Pharmacokinetics and
pharmacodynamics of clofazimine in a mouse model of tuberculosis. Antimicrob Agents Chemother.
2015;59((6)):3042–3051. [PMC free article] [PubMed] [Google Scholar]

103. Almeida D, Ioerger T, Tyagi S, Li SY, Mdluli K, Andries K, et al. Mutations in pepQ confer low-level
resistance to bedaquiline and clofazimine in Mycobacterium tuberculosis. Antimicrob Agents Chemother.
2016;60((8)):4590–4599. [PMC free article] [PubMed] [Google Scholar]

104. Xu J, Wang B, Hu M, Huo F, Guo S, Jing W, et al. Primary clofazimine and bedaquiline resistance among
isolates from patients with multidrug-resistant tuberculosis. Antimicrob Agents Chemother. 2017;61((6)):e00239–
e00317. [PMC free article] [PubMed] [Google Scholar]

105. van Ingen J, Simons S, De Zwaan R, van der Laan T, Kamst-van Agterveld M, Boeree MJ, et al. Comparative
study on genotypic and phenotypic second-line drug resistance testing of Mycobacterium tuberculosis complex
isolates. J Clin Microbiol. 2010;48((8)):2749–2753. [PMC free article] [PubMed] [Google Scholar]

106. Ammerman NC, Swanson RV, Tapley A, Moodley C, Ngcobo B, Adamson J, et al. Clofazimine has delayed
antimicrobial activity against Mycobacterium tuberculosis both in vitro and in vivo. J Antimicrob Chemother.
2017;72((2)):455–461. [PubMed] [Google Scholar]

107. Ammerman NC, Swanson RV, Bautista EM, Almeida DV, Saini V, Omansen TF, et al. Impact of clofazimine
dosing on treatment shortening of the first-line regimen in a mouse model of tuberculosis. Antimicrob Agents
Chemother. 2018;62((7)):e00636–e00718. [PMC free article] [PubMed] [Google Scholar]

108. Swanson RV, Ammerman NC, Ngcobo B, Adamson J, Moodley C, Dorasamy A, et al. Clofazimine
contributes sustained antimicrobial activity after treatment cessation in a mouse model of tuberculosis
chemotherapy. Antimicrob Agents Chemother. 2016;60((5)):2864–2869. [PMC free article] [PubMed] [Google
Scholar]

109. Saini V, Ammerman NC, Chang YS, Tasneen R, Chaisson RE, Jain S, et al. Treatment-shortening effect of a
novel regimen combining clofazimine and high-dose rifapentine in pathologically distinct mouse models of
tuberculosis. Antimicrob Agents Chemother. 2019;63((6)):e00388–e00419. [PMC free article] [PubMed] [Google
Scholar]
110. Nix DE, Adam RD, Auclair B, Krueger TS, Godo PG, Peloquin CA. Pharmacokinetics and relative
bioavailability of clofazimine in relation to food orange juice and antacid. Tuberculosis. 2004;84((6)):365–373.
[PubMed] [Google Scholar]

111. Horita Y, Doi N. Comparative study of the effects of antituberculosis drugs and antiretroviral drugs on
cytochrome P450 3A4 and P-glycoprotein. Antimicrob Agents Chemother. 2014;58((6)):3168–3176. [PMC free
article] [PubMed] [Google Scholar]

112. George J. Metabolism and interactions of antileprosy drugs. Biochem Pharmacol. 2020;177:113993.
[PubMed] [Google Scholar]

113. Abdelwahab MT, Wasserman S, Brust JC, Gandhi NR, Meintjes G, Everitt D, et al. Clofazimine
pharmacokinetics in patients with TB dosing implications. J Antimicrob Chemother. 2020;75((11)):3269–3277.
[PMC free article] [PubMed] [Google Scholar]

114. Faraj A, Svensson RJ, Diacon AH, Simonsson USH. Drug effect of clofazimine on persisters explains an
unexpected increase in bacterial load in patients. Antimicrob Agents Chemother. 2020;64((5)):e01905–e01919.
[PMC free article] [PubMed] [Google Scholar]

115. Tang S, Yao L, Hao X, Liu Y, Zeng L, Liu G, et al. Clofazimine for the treatment of multidrug-resistant
tuberculosis prospective, multicenter, randomized controlled study in China. Clin Infect Dis. 2015;60((9)):1361–
1367. [PubMed] [Google Scholar]

116. Fox GJ, Benedetti A, Cox H, Koh W-J, Viiklepp P, Ahuja S, et al. Group 5 drugs for multidrug-resistant
tuberculosis individual patient data meta-analysis. Eur Respir J. 2017;49((1)):1600993. [PubMed] [Google
Scholar]

117. Ahmad N, Ahuja SD, Akkerman OW, Alffenaar JWC, Anderson LF, et al.Collaborative. Group for the Meta-
Analysis of Individual Patient Data in MDR-TB treatment–2017 Treatment correlates of successful outcomes in
pulmonary multidrug-resistant tuberculosis an individual patient data meta-analysis. Lancet.
2018;392((10150)):821–834. [PMC free article] [PubMed] [Google Scholar]

118. MSF TB-Practecal Clinical Trial MSF. 2022.

119. Hwang TJ, Dotsenko S, Jafarov A, Weyer K, Falzon D, Lunte K, et al. Safety and availability of clofazimine
in the treatment of multidrug and extensively drug-resistant tuberculosis analysis of published guidance and meta-
analysis of cohort studies. BMJ Open. 2014;4((1)):e004143. [PMC free article] [PubMed] [Google Scholar]

120. Lan Z, Ahmad N, Baghaei P, Barkane L, Benedetti A, Brode SK, et al. Drug-associated adverse events in the
treatment of multidrug-resistant tuberculosis an individual patient data meta-analysis. Lancet Respir Med.
2020;8((4)):383–394. [PMC free article] [PubMed] [Google Scholar]

121. Abdelwahab MT, Court R, Everitt D, Diacon AH, Dawson R, Svensson EM, et al. Effect of clofazimine
concentration on QT prolongation in patients treated for tuberculosis. Antimicrob Agents Chemother.
2021;65((7)):e0268720. [PMC free article] [PubMed] [Google Scholar]

122. Mitchison DA. Role of individual drugs in the chemotherapy of tuberculosis. Int J Tuberc Lung Dis.
2000;4((9)):796–806. [PubMed] [Google Scholar]
123. Jayaram R, Gaonkar S, Kaur P, Suresh BL, Mahesh BN, Jayashree R, et al. Pharmacokinetics-
pharmacodynamics of rifampin in an aerosol infection model of tuberculosis. Antimicrob Agents Chemother.
2003;47((7)):2118–2124. [PMC free article] [PubMed] [Google Scholar]

124. Gumbo T, Louie A, Deziel MR, Liu W, Parsons LM, Salfinger M, et al. Concentration-dependent
Mycobacterium tuberculosis killing and prevention of resistance by rifampin. Antimicrob Agents Chemother.
2007;51((11)):3781–3788. [PMC free article] [PubMed] [Google Scholar]

125. De Steenwinkel JE, Aarnoutse RE, De Knegt GJ, Ten Kate MT, Teulen M, Verbrugh HA, et al. Optimization
of the rifampin dosage to improve the therapeutic efficacy in tuberculosis treatment using a murine model. Am J
Respir Crit Care Med. 2013;187((10)):1127–1134. [PubMed] [Google Scholar]

126. Rosenthal IM, Tasneen R, Peloquin CA, Zhang M, Almeida D, Mdluli KE, et al. Dose-ranging comparison of
rifampin and rifapentine in two pathologically distinct murine models of tuberculosis. Antimicrob Agents
Chemother. 2012;56((8)):4331–4340. [PMC free article] [PubMed] [Google Scholar]

127. Hu Y, Liu A, Ortega-Muro F, Alameda-Martin L, Mitchison D, Coates A. High-dose rifampicin kills


persisters, shortens treatment duration, and reduces relapse rate in vitro and in vivo. Front Microbiol. 2015;6:641.
[PMC free article] [PubMed] [Google Scholar]

128. Boeree MJ, Diacon AH, Dawson R, Narunsky K, du Bois J, Venter A, et al. A dose-ranging trial to optimize
the dose of rifampin in the treatment of tuberculosis. Am J Respir Crit Care Med. 2015;191((9)):1058–1065.
[PubMed] [Google Scholar]

129. Aarnoutse RE, Kibiki GS, Reither K, Semvua HH, Haraka F, Mtabho CM, et al. Pharmacokinetics and
bacteriological response of rifampin administered at 600 and 1 200 milligrams daily in patients with pulmonary
tuberculosis. Antimicrob Agents Chemother. 2017;61((11)):e01054–e01117. [PMC free article] [PubMed]
[Google Scholar]

130. Ruslami R, Ganiem AR, Dian S, Apriani L, Achmad TH, van der Ven AJ, et al. Intensified regimen
containing rifampicin and moxifloxacin for tuberculous meningitis an open-label, randomised controlled phase 2
trial. Lancet Infect Dis. 2013;13((1)):27–35. [PubMed] [Google Scholar]

131. Stott KE, Pertinez H, Sturkenboom MGG, Boeree MJ, Aarnoutse R, Ramachandran G, et al.
Pharmacokinetics of rifampicin in adult TB patients and healthy volunteers a systematic review and meta-analysis.
J Antimicrob Chemother. 2018;73((9)):2305–2313. [PMC free article] [PubMed] [Google Scholar]

132. Svensson RJ, Svensson EM, Aarnoutse RE, Diacon AH, Dawson R, Gillespie SH, et al. Greater early
bactericidal activity at higher rifampicin doses revealed by modeling and clinical trial simulations. J Infect Dis.
2018;218((6)):991–999. [PubMed] [Google Scholar]

133. Niemi M, Backman JT, Fromm MF, Neuvonen PJ, Kivistö KT. Pharmacokinetic interactions with rifampicin
clinical relevance. Clin Pharmacokinet. 2003;42((9)):819–850. [PubMed] [Google Scholar]

134. Boeree MJ, Heinrich N, Aarnoutse R, Diacon AH, Dawson R, Rehal S, et al. High-dose rifampicin and
SQ109 for treating tuberculosis a multi-arm, multi-stage randomised controlled trial. Lancet Infect Dis.
2017;17((1)):39–49. [PMC free article] [PubMed] [Google Scholar]
135. Ruslami R, Nijland HMJ, Alisjahbana B, Parwati I, van Crevel R Aarnoutse RE. Pharmacokinetics and
tolerability of a higher rifampin dose versus the standard dose in pulmonary tuberculosis patients. Antimicrob
Agents Chemother. 2007;51((7)):2546–2551. [PMC free article] [PubMed] [Google Scholar]

136. Steingart KR, Jotblad S, Robsky K, Deck D, Hopewell PC, Huang D. Higher-dose rifampin for the treatment
of pulmonary tuberculosis a systematic review. Int J Tuberc Lung Dis. 2011;15((3)):305–316. [PubMed] [Google
Scholar]

137. Jindani A, Borgulya G, De Patiño IW Gonzales T, De Fernandes RA Shrestha B, et al. A randomised Phase II
trial to evaluate the toxicity of high-dose rifampicin to treat pulmonary tuberculosis. Int J Tuberc Lung Dis.
2016;20((6)):832–838. [PubMed] [Google Scholar]

138. Velásquez GE, Brooks MB, Coit JM, Pertinez H, Vargas Vásquez D, Sánchez Garavito E, et al. Efficacy and
safety of high-dose rifampin in pulmonary tuberculosis. A randomized controlled trial. Am J Respir Crit Care
Med. 2018;198((5)):657–666. [PMC free article] [PubMed] [Google Scholar]

139. Te Brake LH, de Jager V, Narunsky K, Vanker N, Svensson EM, Phillips PP, et al. Increased bactericidal
activity but dose-limiting intolerability at 50 mg·kg(−1) rifampicin. Eur Respir J. 2021;58((1)):2000955. [PMC
free article] [PubMed] [Google Scholar]

140. Svensson EM, Dian S, Te Brake L, Ganiem AR, Yunivita V, van Laarhoven A, et al. Model-based meta-
analysis of rifampicin exposure and mortality in Indonesian tuberculous meningitis trials. Clin Infect Dis.
2020;71((8)):1817–1823. [PMC free article] [PubMed] [Google Scholar]

141. Heemskerk AD, Bang ND, Mai NTH, Chau TTH, Phu NH, Loc PP, et al. Intensified antituberculosis therapy
in adults with tuberculous meningitis. N Engl J Med. 2016;374((2)):124–134. [PubMed] [Google Scholar]

142. World Health Organization Guidance for national tuberculosis programmes on the management of
tuberculosis in children World Health Organization. 2014. Report No 9241548746. [PubMed]

143. Thee S, Detjen A, Wahn U, Magdorf K. Rifampicin serum levels in childhood tuberculosis. Int J Tuberc Lung
Dis. 2009;13((9)):1106–1111. [PubMed] [Google Scholar]

144. World Health Organization Rapid advice treatment of tuberculosis in children 2010. 2012. [PubMed]

145. Donald PR, Maritz JS, Diacon AH. The pharmacokinetics and pharmacodynamics of rifampicin in adults and
children in relation to the dosage recommended for children. Tuberculosis. 2011;91((3)):196–207. [PubMed]
[Google Scholar]

146. Donald PR. Antituberculosis drug-induced hepatotoxicity in children. Pediatr Rep. 2011;3((2)):e16–e17.
[PMC free article] [PubMed] [Google Scholar]

147. Aruldhas BW, Hoglund RM, Ranjalkar J, Tarning J, Mathew SK, Verghese VP, et al. Optimization of dosing
regimens of isoniazid and rifampicin in children with tuberculosis in India. Br J Clin Pharmacol.
2019;85((3)):644–654. [PMC free article] [PubMed] [Google Scholar]
148. Wobudeya EC, Hesseling AC, Mave V, Hissar S, Turkova A, Crook AM, Gibb DM. On behalf of the SHINE
trial team. editors. Shorter treatment for minimal tuberculosis in children main findings from the SHINE trial The
Union. 2020 [Google Scholar]

149. World health organization WHO operational handbook on tuberculosis module 4: treatment: drug-susceptible
tuberculosis treatment. 2022. [PubMed]

150. Garcia-Prats AJ, Svensson EM, Winckler J, Draper HR, Fairlie L, van der Laan LE, et al. Pharmacokinetics
and safety of high-dose rifampicin in children with TB the Opti-Rif trial. J Antimicrob Chemother.
2021;76((12)):3237–3246. [PMC free article] [PubMed] [Google Scholar]

151. Dheda K, Lenders L, Magombedze G, Srivastava S, Raj P, Arning E, et al. Drug-penetration gradients
associated with acquired drug resistance in patients with tuberculosis. Am J Respir Crit Care Med.
2018;198((9)):1208–1219. [PMC free article] [PubMed] [Google Scholar]

152. Prideaux B, Via LE, Zimmerman MD, Eum S, Sarathy J, O'brien P, et al. The association between sterilizing
activity and drug distribution into tuberculosis lesions. Nat Med. 2015;21((10)):1223–1227. [PMC free article]
[PubMed] [Google Scholar]

153. Phillips PP, Dooley KE, Gillespie SH, Heinrich N, Stout JE, Nahid P, et al. A new trial design to accelerate
tuberculosis drug development the phase IIC Selection Trial with Extended Post-treatment follow-up (STEP) BMC
Med. 2016;14((1)):51–111. [PMC free article] [PubMed] [Google Scholar]

154. Priftin (rifapentine) Tablets - highlights of prescribing information Initial U.S. Approval 1998. Last visited
17-05-2021 https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/021024s009lbl.pdf.

155. Dickinson JM, Mitchison DA. in vitro properties of rifapentine (MDL473) relevant to its use in intermittent
chemotherapy of tuberculosis. Tubercle. 1987;68((2)):113–118. [PubMed] [Google Scholar]

156. Mor N, Simon B, Mezo N, Heifets L. Comparison of activities of rifapentine and rifampin against
Mycobacterium tuberculosis residing in human macrophages. Antimicrob Agents Chemother. 1995;39((9)):2073–
2077. [PMC free article] [PubMed] [Google Scholar]

157. Keung AC, Owens RC, Jr, Eller MG, Weir SJ, Nicolau DP, Nightingale CH. Pharmacokinetics of rifapentine
in subjects seropositive for the human immunodeficiency virus a phase I study. Antimicrob Agents Chemother.
1999;43((5)):1230–1233. [PMC free article] [PubMed] [Google Scholar]

158. Weiner M, Bock N, Peloquin CA, Burman WJ, Khan A, Vernon A, et al. Pharmacokinetics of rifapentine at
600 and 1 200 mg during once-weekly tuberculosis therapy. Am J Respir Crit Care Med. 2004;169((11)):1191–
1197. [PubMed] [Google Scholar]

159. Dooley KE, Bliven-Sizemore EE, Weiner M, Lu Y, Nuermberger EL, Hubbard WC, et al. Safety and
pharmacokinetics of escalating daily doses of the antituberculosis drug rifapentine in healthy volunteers. Clin
Pharmacol Ther. 2012;91((5)):881–888. [PMC free article] [PubMed] [Google Scholar]

160. Weiner M, Savic RM, Kenzie WRM, Wing D, Peloquin CA, Engle M, et al. Rifapentine pharmacokinetics
and tolerability in children and adults treated once weekly with rifapentine and isoniazid for latent tuberculosis
infection. J Pediatr Infect Dis Soc. 2014;3((2)):132–145. [PubMed] [Google Scholar]
161. Keung A, Eller MG, McKenzie KA, Weir SJ. Single and multiple dose pharmacokinetics of rifapentine in
man part II. Int J Tuberc Lung Dis. 1999;3((5)):437–444. [PubMed] [Google Scholar]

162. Alfarisi O, Alghamdi WA, Al-Shaer MH, Dooley KE, Peloquin CA. Rifampin vs. rifapentine what is the
preferred rifamycin for tuberculosis? Expert Rev Clin Pharmacol. 2017;10((10)):1027–1036. [PubMed] [Google
Scholar]

163. Litjens CHC, Aarnoutse RE, van Ewijk-Beneken Kolmer EW Svensson EM, Colbers A, Burger DM, et al.
Protein binding of rifampicin is not saturated when using high-dose rifampicin. J Antimicrob Chemother.
2019;74((4)):986–990. [PubMed] [Google Scholar]

164. Hibma JE, Radtke KK, Dorman SE, Jindani A, Dooley KE, Weiner M, et al. Rifapentine population
pharmacokinetics and dosing recommendations for latent tuberculosis infection. Am J Respir Crit Care Med.
2020;202((6)):866–877. [PMC free article] [PubMed] [Google Scholar]

165. Keung A, Reith K, Eller MG, McKenzie KA, Cheng L, Weir SJ. Enzyme induction observed in healthy
volunteers after repeated administration of rifapentine and its lack of effect on steady-state rifapentine
pharmacokinetics part I. Int J Tuberc Lung Dis. 1999;3((5)):426–436. [PubMed] [Google Scholar]

166. Dooley K, Flexner C, Hackman J, Peloquin CA, Nuermberger E, Chaisson RE, et al. Repeated administration
of high-dose intermittent rifapentine reduces rifapentine and moxifloxacin plasma concentrations. Antimicrob
Agents Chemother. 2008;52((11)):4037–4042. [PMC free article] [PubMed] [Google Scholar]

167. Li AP, Reith MK, Rasmussen A, Gorski JC, Hall SD, Xu L, et al. Primary human hepatocytes as a tool for the
evaluation of structure-activity relationship in cytochrome P450 induction potential of xenobiotics evaluation of
rifampin, rifapentine and rifabutin. Chem Biol Interact. 1997;107((1–2)):17–30. [PubMed] [Google Scholar]

168. Zvada SP, Denti P, Geldenhuys H, Meredith S, van As D Hatherill M, et al. Moxifloxacin population
pharmacokinetics in patients with pulmonary tuberculosis and the effect of intermittent high-dose rifapentine.
Antimicrob Agents Chemother. 2012;56((8)):4471–4473. [PMC free article] [PubMed] [Google Scholar]

169. Podany AT, Bao Y, Swindells S, Chaisson RE, Andersen JW, Mwelase T, et al. Efavirenz pharmacokinetics
and pharmacodynamics in HIV-infected persons receiving rifapentine and isoniazid for tuberculosis prevention.
Clin Infect Dis. 2015;61((8)):1322–1327. [PMC free article] [PubMed] [Google Scholar]

170. Swindells S, Ramchandani R, Gupta A, Benson CA, Leon-Cruz J, Mwelase N, et al. One month of
rifapentine plus isoniazid to prevent HIV-related tuberculosis. N Engl J Med. 2019;380((11)):1001–1011. [PMC
free article] [PubMed] [Google Scholar]

171. Martinson NA, Barnes GL, Moulton LH, Msandiwa R, Hausler H, Ram M, et al. New regimens to prevent
tuberculosis in adults with HIV infection. N Engl J Med. 2011;365((1)):11–20. [PMC free article] [PubMed]
[Google Scholar]

172. Sterling TR, Villarino ME, Borisov AS, Shang N, Gordin F, Bliven-Sizemore E, et al. Three months of
rifapentine and isoniazid for latent tuberculosis infection. N Engl J Med. 2011;365((23)):2155–2166. [PubMed]
[Google Scholar]
173. Sterling TR, Scott NA, Miro JM, Calvet G, La Rosa A, Infante R, et al. Three months of weekly rifapentine
and isoniazid for treatment of Mycobacterium tuberculosis infection in HIV-coinfected persons. AIDS.
2016;30((10)):1607–1615. [PMC free article] [PubMed] [Google Scholar]

174. Njie GJ, Morris SB, Woodruff RY, Moro RN, Vernon AA, Borisov AS. Isoniazid-rifapentine for latent
tuberculosis infection a systematic review and meta-analysis. Am J Prev Med. 2018;55((2)):244–252. [PMC free
article] [PubMed] [Google Scholar]

175. Sandul AL, Nwana N, Holcombe JM, Lobato MN, Marks S, Webb R, et al. High rate of treatment completion
in program settings with 12-dose weekly isoniazid and rifapentine for latent Mycobacterium tuberculosis infection.
Clin Infect Dis. 2017;65((7)):1085–1093. [PMC free article] [PubMed] [Google Scholar]

176. Gao XF, Li J, Yang ZW, Li YP. Rifapentine vs. rifampicin for the treatment of pulmonary tuberculosis a
systematic review. Int J Tuberc Lung Dis. 2009;13((7)):810–819. [PubMed] [Google Scholar]

177. Dorman SE, Savic RM, Goldberg S, Stout JE, Schluger N, Muzanyi G, et al. Daily rifapentine for treatment
of pulmonary tuberculosis. A randomized dose-ranging trial. Am J Respir Crit Care Med. 2015;191((3)):333–343.
[PMC free article] [PubMed] [Google Scholar]

178. Rosenthal IM, Zhang M, Williams KN, Peloquin CA, Tyagi S, Vernon AA, et al. Daily dosing of rifapentine
cures tuberculosis in three months or less in the murine model. PLoS Med. 2007;4((12)):e344. [PMC free article]
[PubMed] [Google Scholar]

179. Dorman SE, Nahid P, Kurbatova EV, Phillips PPJ, Bryant K, Dooley KE, et al. Four-month rifapentine
regimens with or without moxifloxacin for tuberculosis. N Engl J Med. 2021;384((18)):1705–1718. [PMC free
article] [PubMed] [Google Scholar]

180. Seidel H, Bittner J. Darstellung der βm Aminosalicysäure. Monatsh Chem. 1902;23:431–433. [Google
Scholar]

181. Donald PR, Diacon AH. Para-aminosalicylic acid the return of an old friend. Lancet Infect Dis.
2015;15((9)):1091–1099. [PubMed] [Google Scholar]

182. Abulfathi AA, Donald PR, Adams K, Svensson EM, Diacon AH, Reuter H. The pharmacokinetics of para-
aminosalicylic acid and its relationship to efficacy and intolerance. Br J Clin Pharmacol. 2020;86((11)):2123–
2132. [PMC free article] [PubMed] [Google Scholar]

183. Dubovsky H. History of Medicine-Correspondence with a pioneer Jürgen Lehmann (1898-1989) producer of
tl” le first effective antituberculosis specific. South Afr Med J. 1991;79((1)):48–50. [PubMed] [Google Scholar]

184. Lehmann J. Para-aminosalicylic acid in the treatment of tuberculosis. Lancet. :194615–194616. [PubMed]
[Google Scholar]

185. Brunton L, Knollmann B, Hilal-Dandan RG. New York City: USA McGraw-hill Education; 2018. Gilman's
the pharmacological basis of therapeutics. [Google Scholar]

186. Parvez MM, Shin HJ, Jung JA, Shin JG. Evaluation of para-aminosalicylic acid as a substrate of multiple
solute carrier uptake transporters and possible drug interactions with nonsteroidal anti-inflammatory drugs in
vitro. Antimicrob Agents Chemother. 2017;61((5)):e02392–e02416. [PMC free article] [PubMed] [Google
Scholar]

187. Jacobus Pharmaceutical Company I Paser 2010. Available from https://medlibrary.org/lib/rx/meds/paser/

188. Peloquin CA, Henshaw TL, Huitt GA, Berning SE, Nitta AT, James GT. Pharmacokinetic evaluation of para-
aminosalicylic acid granules. Pharmacotherapy. 1994;14((1)):40–46. [PubMed] [Google Scholar]

189. Heifets LB. Boca Raton, FL: CRC Press; 1991. Antituberculosis drugs antimicrobial activity in vitro. Drug
susceptibility in the chemotherapy of mycobacterial infections; pp. p.13–58. [Google Scholar]

190. Singh B, Mitchison DA. The bactericidal activities of combinations of streptomycin p-aminosalicylic acid
(PAS) oxytetracycline (terramycin) and viomycin against Mycobacterium tuberculosis. J Gen Microbiol.
1955;13((1)):176–184. [PubMed] [Google Scholar]

191. Bartmann K. Antituberculosis drugs Springer Berlin Heidelberg. 1988 [Google Scholar]

192. Wan SH, Pentikäinen PJ, Azarnoff DL. Bioavailability of aminosalicylic acid and its various salts in humans
III absorption from tablets. J Pharm Sci. 1974;63((5)):708–711. [PubMed] [Google Scholar]

193. Pentikäinen PJ, Wan SH, Azarnoff DL. Bioavailability of aminosalicylic acid and its various salts in humans
IV comparison of four brands of the sodium salt. J Pharm Sci. 1974;63((9)):1431–1434. [PubMed] [Google
Scholar]

194. Adams KT, Donald PR, Abulfathi AA, Diacon AH, Stander MA, Reuter H. Pharmacokinetics of para-
aminosalicylic acid and its 2 major metabolites a potential relationship to the development of gastrointestinal
intolerance. J Clin Pharmacol. 2020;60((4)):489–494. [PubMed] [Google Scholar]

195. Lehmann J. The role of the metabolism of P-Aminosalicylic acid (PAS) in the treatment of tuberculosis.
Interaction with the metabolism of Isonicotinic acid Hydrazide (INH) and the synthesis of cholesterol. Scand J
Respir Dis. 1969;50((3)):169–185. [PubMed] [Google Scholar]

196. Sy SK, De Kock L Diacon AH, Werely CJ, Xia H, Rosenkranz B, et al. N-acetyltransferase genotypes and the
pharmacokinetics and tolerability of para-aminosalicylic acid in patients with drug-resistant pulmonary
tuberculosis. Antimicrob Agents Chemother. 2015;59((7)):4129–4138. [PMC free article] [PubMed] [Google
Scholar]

197. Peloquin CA, Zhu M, Adam RD, Singleton MD, Nix DE. Pharmacokinetics of para-aminosalicylic acid
granules under four dosing conditions. Ann Pharmacother. 2001;35((11)):1332–1338. [PubMed] [Google Scholar]

198. Yue WY, Cohen SS. Toleration and absorption of sodium para-aminosalicylate and para-aminosalicylic acid
(neopasalate) comparison with other forms of para-aminosalicylic acid. Dis Chest. 1966;49((2)):165–174.
[PubMed] [Google Scholar]

199. Jones JS. Intravenous PAS in relation to pulmonary tuberculotherapy with an account of 1, 145 transfusions.
Br J Tuberc Dis Chest. 1954;48((4)):286–297. [PubMed] [Google Scholar]

200. Charles F. The treatment of pulmonary tuberculosis with intravenous PAS-infusions. Tubercle.
1955;36((2)):40–44. [PubMed] [Google Scholar]
201. The chemotherapy of tuberculosis Br Med J. 1950;2((4688)):1102–1103. [PMC free article] [PubMed]
[Google Scholar]

202. Treatment of pulmonary tuberculosis with streptomycin and para-amino-salicylic acid; a Medical Research
Council investigation Br Med J. 1950. pp. 1073–1085. [PMC free article] [PubMed]

203. The prevention of streptomycin resistance by combined chemotherapy; a Medical Research Council
investigation Br Med J. 1952;1((4769)):1157–1162. [PMC free article] [PubMed] [Google Scholar]

204. Various combinations of isoniazid with streptomycin or with PAS in the treatment of pulmonary tuberculosis;
seventh report to the Medical Research Council by their Tuberculosis Chemotherapy Trials Committee Br Med J.
1955;1:435. [PMC free article] [PubMed] [Google Scholar]

205. Jindani A, Aber VR, Edwards EA, Mitchison DA. The early bactericidal activity of drugs in patients with
pulmonary tuberculosis. Am Rev Respir Dis. 1980;121((6)):939–949. [PubMed] [Google Scholar]

206. Para-aminosalicylic acid treatment in pulmonary tuberculosis Am Rev tuberculosis. 1950;61((5)):597–612.


[PubMed] [Google Scholar]

207. Wissenschaftliche Arbeitsgemeinschaft für die Therapie von Lungenkrankheiten WATL Cooperative
controlled trial of thiocarlide (DATC) PAS and bedrest alone in short-term single-drug treatment in retreated
cavitary pulmonary tuberculosis. Beitr Klin Tuberk. 1969;139((2)):115–139. [PubMed] [Google Scholar]

208. de Kock L, Sy SK, Rosenkranz B, Diacon AH, Prescott K, Hernandez KR, et al. Pharmacokinetics of para-
aminosalicylic acid in HIV-uninfected and HIV-coinfected tuberculosis patients receiving antiretroviral therapy
managed for multidrug-resistant and extensively drug-resistant tuberculosis. Antimicrob Agents Chemother.
2014;58((10)):6242–6250. [PMC free article] [PubMed] [Google Scholar]

209. World Health Organization Gear up to end TB introducing the end TB strategy World Health Organization.
2015.

You might also like