Skip to main content

REVIEW article

Front. Cell. Infect. Microbiol., 09 September 2022
Sec. Molecular Bacterial Pathogenesis
This article is part of the Research Topic Drug-resistant Mycobacterium tuberculosis View all 21 articles

How Mycobacterium tuberculosis drug resistance has shaped anti-tubercular drug discovery

Amala Bhagwat&#x;Amala BhagwatAditi Deshpande&#x;Aditi DeshpandeTanya Parish*Tanya Parish*
  • Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States

Drug resistance is an increasing problem for the treatment of tuberculosis. The prevalence of clinical isolates with pre-existing resistance needs to be considered in any drug discovery program. Non-specific mechanisms of resistance such as increased efflux or decreased permeability need to be considered both in developing individual drug candidates and when designing novel regimens. We review a number of different approaches to develop new analogs and drug combinations or improve efficacy of existing drugs that may overcome or delay the appearance of clinical resistance. We also discuss the need to fully characterize mechanisms of resistance and cross- resistance to existing drugs to ensure that novel drugs will be clinically effective.

Introduction

Tuberculosis (TB) remains a major global health problem causing ~1.5 million deaths in 2020 (World Health Organization, 2021). Treatment of TB is complicated owing to the unique capacity of the causative bacterium (Mycobacterium tuberculosis), to survive within the human body. Although the bacilli are unable to replicate in acidic or hypoxic environments, such as found in the granuloma, M. tuberculosis can persist in these environments for lengthy periods. Latent TB infection (LTBI), in which the bacilli remain in the body without clinical symptoms poses a unique problem for the diagnosis and treatment of TB. In addition, drug resistance is a major problem which can result from both inherent and acquired resistance mechanisms. Thus TB control programs require both new drugs to overcome existing resistance. and rapid detection tests for drug resistance.

Although treatment of TB became possible with the discovery of streptomycin, there are few antibiotics available for modern use. The major frontline drugs for drug-susceptible TB, isoniazid and rifampicin have been supplemented with the recent addition of the new agents active against drug resistant TB, such as bedaquiline (a diarylquinolone), delaminid and pretomanid (nitroimidazoles) and repurposing of linezolid (an oxazolidinone), but the pipeline is still inadequate (Libardo et al., 2018; Oh et al., 2021). As for other antibiotics, resistance to streptomycin was observed very soon after its clinical use. Early clinical trials demonstrated the utility of combination regimens (Fox, 1979; Mitchison, 1985; Grosset, 1989) with the additional advantage that resistance to multiple agents is harder to acquire. However, given the length of time the frontline agents have been in use, a rise in drug resistance has been seen for rifampicin and isoniazid both singly and in combination (multi-drug resistance or MDR). Extensively drug resistant (XDR) strains are resistant to rifampicin, isoniazid and a fluoroquinolone.

Drug resistance in TB is largely mediated by chromosomal mutations, as there are no reports of horizontal gene transfer. However, there are multiple routes by which the bacilli can become resistant, not all of which involve mutation of the drug target. Mutations in the target which reduce or alleviate drug-binding do occur, as does mutation in the promoter leading to increased target expression. Drug inactivation, such as with the beta lactamases that degrade the beta lactams or modifying enzymes such as acetyl/methyl transferases are another resistance mechanism reported in M. tuberculosis (Zaunbrecher et al., 2009; Kurz and Bonomo, 2012). Other mechanisms which do not involve chromosomal mutation have been noted in vitro such as mistranslation of proteins leading to phenotypic resistance to rifampicin (Javid et al., 2014). In addition, changes in drug uptake or efflux are non-specific processes which can affect sensitivity to multiple drugs from the same or different chemical classes. M. tuberculosis has a lipid-rich outer cell wall which imparts intrinsic resistance by acting as a permeability barrier, and changes in cell wall composition can affect drug uptake (Jarlier and Nikaido, 1994). M. tuberculosis also has a variety of efflux systems which, if up-regulated, can lead to resistance (Rodrigues et al., 2017). For pro-drugs, such as isoniazid, occurrence of mutation in the activating enzymes can lead to drug resistance (Zhang et al., 1992; Zhang and Yew, 2009; Seifert et al., 2015). Given the variety of resistance mechanisms and the possibility of resistance to multiple drugs, an evaluation of the resistance mechanisms for new drugs is an important component of the drug discovery process, alongside the standard considerations of pre-existing resistance and resistance frequency.

The need for lengthy treatments (>6 months for drug sensitive TB) with multiple agents poses issues with adherence that can lead to the selection of resistant subpopulations during treatment. There is an urgent need to develop agents with new mechanisms that are not affected by pre-existing resistance, but also to shorten the duration of TB treatment to restrict the emergence of resistance. Thus, drug discovery for TB has been strongly shaped by the prevalence of existing resistance mechanisms, as well as the rate of resistance appearing in the clinic.

Drug discovery for TB has adopted several approaches which attempt to develop new agents to address the issue of pre-existing resistance and to the appearance of new resistance mechanisms. Several approaches have been used including: (i) Generating analogs which overcome resistance by binding to the target in a different fashion; (ii) Restoring sensitivity to antibiotics using booster or adjunct molecules; (iii) Using combinations to generate new regimens to minimize the appearance of resistance. We will review recent successes in these approaches and address some of the additional factors that should be considered when developing new agents (Table 1).

TABLE 1
www.frontiersin.org

Table 1 Examples of resistance mechanisms to current TB drugs and approaches to overcome resistance (references in text).

Development of analogs of existing drugs

The standard drug regimen for TB has a high success rate for cure when used with drug sensitive strains. Thus, there has been a lot of emphasis on developing new analogs of these successful antibiotics, but which can overcome pre-existing resistance.

New RNA polymerase inhibitors

The DNA-dependent RNA polymerase is the target of the frontline drug rifampicin. The majority of clinical resistance results from mutation in a hotspot of 81bp in the coding region of the target RpoB (Telenti et al., 1993; Mboowa et al., 2014; Zaw, 2018). Mutations in clinical isolates which result in changes in the hydrogen bonding and van der Waals interaction between RpoB and rifampicin are associated with clinical resistance (Li et al., 2021). Knowledge of the binding mechanism can be used to design novel derivatives which retain binding or to find molecules that bind to different sites on the RNA polymerase. Other members of the rifamycin class such as rifampin, rifapentine and rifabutin have the same pharmacophore which can result in cross-resistance (Alfarisi et al., 2017; Tiberi et al., 2017; Farhat et al., 2019). For example, H526C mutations lead to resistance to both rifampicin and rifabutin (Cavusoglu et al., 2004). Molecules with alternative binding sites/modes are of interest, for example fidaxomicin has in vitro activity against M. tuberculosis and a class of N-aroyl-N-aryl-phenyl-alaninamides were identified that bind to RNA polymerase and inhibit M. tuberculosis without cross-resistance (Lin et al., 2017; Kirsch et al., 2022). Development of these alternative RNA polymerase inhibitors could supplant rifampicin in a regimen and overcome clinical resistance.

New InhA inhibitors

Isoniazid is one of the earliest anti-tubercular drugs and works via inhibition of InhA, a component of FAS-II (fatty acid synthase) involved in synthesis of mycolic acids, key cell wall components. Isoniazid is a prodrug which is activated intracellularly by the KatG catalase-peroxidase (Zhang et al., 1992). The activated molecule forms an adduct with NAD(H) at the active site of the enzyme (Banerjee et al., 1994; Rawat et al., 2003). There are multiple routes to isoniazid resistance: (i) mutations in KatG (most commonly S315T) which reduce its enzymatic activity leading to lack of activation of isoniazid; (ii) mutations in the target InhA which lead to lack of binding (Tseng et al., 2015); and (iii) mutations in the promoter region which lead to increased expression of InhA (Seifert et al., 2015). A combination of mutations in the promoter and InhA are often seen clinically with highly resistant strains (Seifert et al., 2015).

In order to generate analogs which overcome resistance, the development of direct InhA inhibitors which do not require activation shows promise. Early work on triclosan and its derivatives confirmed that it was possible to develop alternative inhibitors for InhA (Armstrong et al., 2020; Rodriguez et al., 2020; Chetty et al., 2021) and multiple scaffolds, as well as a natural product, have been identified which can inhibit InhA (Pan and Tonge, 2012). These newer analogs generally do not require activation and bind directly to InhA, thus they can overcome resistance due to KatG and InhA mutation. A series of hydroxy-pyridones which do not require activation are active against common isoniazid resistant clinical strains (Manjunatha and Smith, 2015), as are several classes of thiadiazoles which inhibit InhA directly (Šink et al., 2015; Martínez-Hoyos et al., 2016). In addition, diazaborines which do not require activation or binding to NADH have been developed (Xia et al., 2018) which are active against isoniazid resistant clinical isolates. These also demonstrate good activity against both replicating and non-replicating bacteria (Flint et al., 2020) suggesting they might be able both to overcome pre-existing resistance and shorten therapy by eliminating persistent organisms. The natural product pyridomycin also targets InhA, as a competitive binder for NADH and is active against most clinically-resistant isolates (Hartkoorn et al., 2012). In addition to overcoming existing resistance new analogs which do not require activation would have a lower frequency of resistance, so drug resistance in the clinic would likely appear more slowly. This has been demonstrated in animal models, where the diazborine AN12855 had a lower frequency of resistance in mice as compared to INH (Robertson et al., 2019).

New gyrase inhibitors

Fluoroquinolones are broad-spectrum antibiotics with bactericidal activity which target DNA gyrase and DNA topoisomerase. In M. tuberculosis, DNA gyrase is the sole target, since it lacks the topoisomerase (Nagaraja et al., 2017; Aubry et al., 2004) Fluoroquinolones are attractive since they have activity against replicating, non-replicating and intracellular M. tuberculosis. Resistance to fluoroquinolones in M. tuberculosis is due to mutations in DNA gyrase (Avalos et al., 2015); high level resistance is generally conferred by mutation in the GyrA subunit in the quinolone resistance determining region covering codons 74-113 (Soudani et al., 2010; Singh et al., 2015; Singh et al., 2021; Chaoui et al., 2018). A single mutation can lead to resistance to the entire class of fluoroquinolones, therefore novel agents with different binding modes would be useful.

One approach to overcome resistance encoded by gyrA mutations, has been to identify novel scaffolds that target gyrase in vitro. Examples include the naphthyridone/aminopiperidines (Gibson et al., 2019) and alkoxytriazoloquinolones (Carta et al., 2019). The spiropyrimidinetrione series has activity against M. tuberculosis strains with mutations in gyrase suggesting a potential to overcome fluoroquinolone resistance (Basarab et al., 2022). In addition, the possibility of targeting GyrB has been addressed (Stokes et al., 2020); for example, the aminopyrazinamides and 2-amino-5-phenylthiophene-3-carboxamide (Shirude et al., 2013; Saxena et al., 2015) which target GyrB have good potency in vitro.

Restoring/improving the activity of existing agents

M. tuberculosis is intrinsically resistant (or can become resistant) to several classes of antibiotics via expression of drug-metabolizing enzymes. The bacilli also have efflux systems which can minimize intracellular accumulation and target engagement. Examples of efforts to overcome these intrinsic resistance mechanisms are described below and may lead to new strategies for prolonging the useful life of an antibiotic and/or reducing the required dose.

Ethionamide (ETH) is a prodrug which is activated by M. tuberculosis EthA to form an NAD-adduct which binds to InhA and inhibits mycolic acid synthesis (similar to the mode of action of isoniazid) (Vannelli et al., 2002). EthA, a flavin mono-oxygenase, is negatively regulated by the transcriptional regulator EthR. Inhibition of EthR leads to up-regulation of EthA which increases the activity of ETH. Small molecule inhibitors which disrupt EthR-DNA binding are able to “boost” the activity of ETH significantly, leading to activity in vivo at reduced doses (Willand et al., 2009) and could improve the clinical utility of ETH.

Ethionamide efficacy can also be “boosted” by the N-acylated 4-phenylpiperidine series (Flipo et al., 2022). These molecules interact with the VirS transcriptional regulator leading to the increased expression of MymA, a monooxygenase which activates ethionamide. This approach was successful in overcoming ethionamide resistance due to EthA mutations in vitro and in an animal model of infection.

M. tuberculosis is intrinsically resistant to beta lactams due to the expression of beta lactamase, but this can be reversed by the addition of beta lactamase inhibitors. For example, meropenem is highly effective in vitro when combined with clavulanate, as are the cephalosporins (Hugonnet et al., 2009; Ramón-García et al., 2016). The clinical effectiveness of meropenem is less clear, due to tolerability issues (De Jager et al., 2022), but this has led to an increased effort to find new beta lactams (Gold et al., 2022).

The M. tuberculosis acetyltransferase Eis can modify aminoglycosides thereby inactivating them (Willby et al., 2016). Increased expression of the enzyme leads to kanamycin resistance (Zaunbrecher et al., 2009) whereas inactivation of Eis restores kanamycin sensitivity. Several series of Eis inhibitors have been identified Willby et al., 2016; Punetha et al., 2020; (Punetha et al., 2021). Although kanamycin is unlikely to be used clinically since it is not orally available, this approach does lend proof of concept to the idea that targeting antibiotic modifying enzymes can overcome intrinsic resistance.

Drug efflux is a common mechanism of intrinsic resistance in many bacterial species, and M. tuberculosis encodes many efflux systems (Louw et al., 2009; Rodrigues et al., 2017). Differences in the expression or activity of efflux pumps in clinical isolates has been linked to resistance and over-expression of several systems (mmr, mmpL7, Rv1285c, p55 and efpA) was noted in response to drug treatment (Machado et al., 2017). Increased efflux is linked to antibiotic tolerance and the development of drug resistance (Pasipanodya and Gumbo, 2011). Thus targeting efflux and/or specific efflux pumps has been proposed as a way to improve efficacy of drugs and reduce resistance, although inhibiting efflux non-specifically can have issues with selectivity and/or toxicity (Rodrigues et al., 2020). Inhibitors of the EfpA efflux pump were recently identified (Johnson et al., 2019). EfpA plays a role in antibiotic tolerance in mycobacteria since its over-expression led to decreased uptake of several antibiotics including moxifloxacin (Rai and Mehra, 2021). Thus inhibitors of this system might have a dual function, since inhibition of EfpA inhibits growth, but could also prevent induction of tolerance.

Using combinations to reduce resistance

Combination regimens

The general consensus in anti-bacterial drug discovery is that the appearance of resistance occurs within a decade of widespread use for any new drug. If resistance can be delayed, this prolongs the useful life of a new drug. Standard TB therapy consists of a four drug regimen, partly because the drugs are insufficient on their own, but also because the combination of drugs can be very effective in delaying the appearance of resistance. Since the majority of target-based resistance is due to chromosomal mutations in M. tuberculosis, combining drugs is an effective way to reduce the frequency of resistance (since bacteria would need to be resistant to more than one agent simultaneously at the outset). Thus the development of new regimens, rather than individual drugs, is standard practice for TB. However, there are still additional considerations for generating the best regimens. In particular, the resistance mechanism(s) for each drug in the regimen needs to be different. Combining drugs which hit different targets is not sufficient to prevent cross-resistance, due to the possibility of non-specific resistance mechanisms. Recent experience using monotherapy with bedaquiline has demonstrated that low level clinical resistance can appear quickly and that it can involve non-specific mechanisms, such as increased drug efflux (see below). Therefore considering the susceptibility of novel agents to common resistance mechanisms is important.

Dual targeting molecules

An alternative approach to developing individual agents for a combination regimen is to develop agents that simultaneously inhibit more than one target. This has been proposed both for targets from the same family as well as for targets with different active sites. For example, uridine derivatives that target multiple Mur enzymes (involved in the same pathway of peptidoglycan synthesis) have been identified (Kumari et al., 2022), as well as “ionized non-classical antifolates” that target both dihydrofolate reductase and thymidylate synthase (Hajian et al., 2019); the thiophene carboxamide IMB-T130 which targets both tyrosyl-tRNA synthetase and dehydroquinate synthase (Zhu et al., 2015; Zhu et al., 2018); and SQ109 which targets both MmpL3 and respiration (Kai et al., 2014; Li et al., 2014, 3). Although this approach could be useful to reduce the frequency of resistance to a single agent it may pose difficulties with respect to optimization for multiple targets, dosing and pharmacokinetics due to variation in the expression level, essentiality and vulnerability of the targets.

Overcoming drug tolerance and eradicating persistent organisms

Antibiotic tolerance is assumed to be one of the major reasons that TB therapy takes many months; the persistence of genotypically sensitive, but phenotypically resistant bacilli may be a consequence of the physiological state(s) induced by host-induced stresses such as acidic pH, hypoxia or nutrient starvation (Mandal et al., 2019). Antibiotic tolerance is a precursor to the appearance of drug resistant bacilli since it allows for extended periods of survival in fluctuating concentrations of antimicrobial agents. Therefore, developing novel drugs that can shorten therapy would be a major advance in preventing or delaying the appearance of resistant isolates in the clinic. A number of groups have conducted high throughput screens to identify agents which target non-replicating organisms induced by different in vitro stresses including hypoxia, low pH, nitric oxide, cholesterol and nutrient starvation, as well as multi-stress models combining these (reviewed in (Parish, 2020). Such screens have identified numerous scaffolds for investigation. The most advanced compound GSK286, which was identified in a macrophage screen, targets cholesterol metabolism and is currently in a Phase I clinical trial (GlaxoSmithKline, 2022; Nuermberger et al., 2022).

The impact of broad resistance mechanisms on early drug discovery

Bedaquiline, a member of class of diarylquinolines, inhibits ATP generation by binding to the C subunit of F0-F1 of the ATP synthase. High level resistance results from mutations in AtpE which reduce binding affinity. However, other mechanisms of resistance are found including mutations in the transcriptional repressor Rv0678 (efflux pump regulator) (Andries et al., 2014) and pepQ (Hartkoorn et al., 2014; Almeida et al., 2016). Mutations in Rv0678 lead to upregulation of the MmpL5/MmpS5 efflux system and increased efflux of the drug. Since this system also effluxes other drug classes, including azoles, clofazimine and macozinone (Hartkoorn et al., 2014; Chen et al., 2022; Guo et al., 2022), the appearance of these mutations in clinical isolates will lead to cross-resistance to multiple antimycobacterial classes. Similarly mutations in pepQ result in resistance to other agents such as macozinone (Chen et al., 2022; Guo et al., 2022), This underscores the need to determine mechanisms of resistance for new agents for both low-level and high-level resistance. In addition, since Rv0678 mutations occur in clinical isolates (Andries et al., 2014), mutant strains with these SNPs should form part of any clinical isolate panel used for routine testing during drug discovery.

Determining mechanisms of resistance during the discovery phase

Phenotypic screening has been very successful in identifying new scaffolds for development. The disadvantage of whole cell screens is that the target is not known from the outset, so much effort has been put into developing target identification and validation methods. One of the most commonly-used methods is to isolate resistant mutants and characterize the chromosomal mutations. This can provide valuable information about potential target(s) and insight into the mechanism(s) of resistance. In these studies, the major focus has been on determining the frequency of resistance and of identifying mutations that lead to high level resistance.

Identification of the target and mutations that affect inhibitor binding can be invaluable in designing new analogs. However, there can be a disconnect between the mutations found in vitro and those that arise in vivo during treatment. For example, complete loss of KatG activity results in attenuation of M. tuberculosis but is the most common mechanism of isoniazid resistance isolated in vitro. In contrast, mutations which reduce the activity of KatG are more often seen in vivo (Vilchèze and Jacobs, 2014). Similarly the spectrum of mutations seem for linezolid are different in vitro from in vivo (Lee et al., 2012; McNeil et al., 2017). In clinical isolates of M. tuberculosis, resistance-conferring mutations are often accompanied by compensatory mutations that increase the overall fitness of the pathogen by restoring the activity of the drug target (Alame Emane et al., 2021). High level resistance can result from multiple mutations in the drug target which may affect binding and/or activity. As noted above, non-specific resistance mechanisms can also lead to low level resistance. Thus identifying mechanisms of resistance that arise using both in vitro and in vivo using relevant infection models are important to include in drug discovery efforts, as well as testing against a large panel of isogenic strains and clinical isolates.

Conclusion

Drug discovery for tuberculosis is notoriously difficult due to the nature of the bacterium and the pathology of the disease. The existence of resistance in clinical isolates and the probability of resistance developing to new agents in the clinic poses further restraints on drug development. Several approaches to deal with the prevalence of clinically-resistant isolates have been tried including the development of analogs of existing frontline drugs and potentiation of the efficacy of existing drugs. The development of novel combination regimens aims to reduce the appearance of resistance. In practical terms, during the development of novel anti-microbials, a wide range of clinical isolates carrying known resistance-associated mutations should form part of a screening panel. Ideally, such a panel would also include strains with decreased permeability and increased efflux. In addition, a full characterization of mutations that lead to low level and high-level resistance in vitro and in vivo should form part of the characterization of any drug candidate.

Author contributions

All authors wrote and edited the manuscript. All authors contributed to the article and approved the submitted version.

Funding

This work was funded by NIAID of the National Institutes of Health under award number R01AI129360 and the Department of Defense Office of the Congressionally Directed Medical Research Programs under award number PR191269.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Alame Emane, A. K., Guo, X., Takiff, H. E., Liu, S. (2021). Drug resistance, fitness and compensatory mutations in mycobacterium tuberculosis. Tuberculosis 129, 102091. doi: 10.1016/j.tube.2021.102091

PubMed Abstract | CrossRef Full Text | Google Scholar

Alfarisi, O., Alghamdi, W. A., Al-Shaer, M. H., Dooley, K. E., Peloquin, C. A. (2017). Rifampin vs. rifapentine: what is the preferred rifamycin for tuberculosis? Expert Rev. Clin. Pharmacol. 10, 1027–1036. doi: 10.1080/17512433.2017.1366311

PubMed Abstract | CrossRef Full Text | Google Scholar

Almeida, D., Ioerger, T., Tyagi, S., Li, S.-Y., Mdluli, K., Andries, K., et al. (2016). Mutations in pepQ confer low-level resistance to bedaquiline and clofazimine in mycobacterium tuberculosis. Antimicrob. Agents Chemother. 60, 4590–4599. doi: 10.1128/AAC.00753-16

PubMed Abstract | CrossRef Full Text | Google Scholar

Andries, K., Villellas, C., Coeck, N., Thys, K., Gevers, T., Vranckx, L., et al. (2014). Acquired resistance of mycobacterium tuberculosis to bedaquiline. PloS One 9, e102135. doi: 10.1371/journal.pone.0102135

PubMed Abstract | CrossRef Full Text | Google Scholar

Armstrong, T., Lamont, M., Lanne, A., Alderwick, L. J., Thomas, N. R. (2020). Inhibition of mycobacterium tuberculosis InhA: Design, synthesis and evaluation of new di-triclosan derivatives. Bioorg Med. Chem. 28, 115744. doi: 10.1016/j.bmc.2020.115744

PubMed Abstract | CrossRef Full Text | Google Scholar

Aubry, A., Pan, X.-S., Fisher, L. M., Jarlier, V., Cambau, E. (2004). Mycobacterium tuberculosis DNA gyrase: interaction with quinolones and correlation with antimycobacterial drug activity. Antimicrob. Agents Chemother. 48, 1281–88. doi: 10.1128/AAC.48.4.1281-1288.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Avalos, E., Catanzaro, D., Catanzaro, A., Ganiats, T., Brodine, S., Alcaraz, J., et al. (2015). Frequency and geographic distribution of gyrA and gyrB mutations associated with fluoroquinolone resistance in clinical mycobacterium tuberculosis isolates: a systematic review. PloS One 10, e0120470. doi: 10.1371/journal.pone.0120470

PubMed Abstract | CrossRef Full Text | Google Scholar

Banerjee, A., Dubnau, E., Quemard, A., Balasubramanian, V., Um, K. S., Wilson, T., et al. (1994). inhA, a gene encoding a target for isoniazid and ethionamide in mycobacterium tuberculosis. Science 263, 227–230. doi: 10.1126/science.8284673

PubMed Abstract | CrossRef Full Text | Google Scholar

Basarab, G. S., Ghorpade, S., Gibhard, L., Mueller, R., Njoroge, M., Peton, N., et al. (2022). Spiropyrimidinetriones: a class of DNA gyrase inhibitors with activity against mycobacterium tuberculosis and without cross-resistance to fluoroquinolones. Antimicrobial Agents Chemotherapy 66, e02192–e02121. doi: 10.1128/aac.02192-21

CrossRef Full Text | Google Scholar

Carta, A., Bua, A., Corona, P., Piras, S., Briguglio, I., Molicotti, P., et al. (2019). Design, synthesis and antitubercular activity of 4-alkoxy-triazoloquinolones able to inhibit the m. tuberculosis DNA gyrase. Eur. J. Medicinal Chem. 161, 399–415. doi: 10.1016/j.ejmech.2018.10.031

CrossRef Full Text | Google Scholar

Cavusoglu, C., Karaca-Derici, Y., Bilgic, A. (2004). In-vitro activity of rifabutin against rifampicin-resistant mycobacterium tuberculosis isolates with known rpoB mutations. Clin. Microbiol. Infection 10, 662–665. doi: 10.1111/j.1469-0691.2004.00917.x

CrossRef Full Text | Google Scholar

Chaoui, I., Oudghiri, A., El Mzibri, M. (2018). Characterization of gyrA and gyrB mutations associated with fluoroquinolone resistance in mycobacterium tuberculosis isolates from Morocco. J. Global Antimicrobial Resistance 12, 171–174. doi: 10.1016/j.jgar.2017.10.003

CrossRef Full Text | Google Scholar

Chen, X., Li, Y., Wang, B., Lu, Y. (2022). Identification of mutations associated with macozinone-resistant in mycobacterium tuberculosis. Curr. Microbiol. 79, 205. doi: 10.1007/s00284-022-02881-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Chetty, S., Armstrong, T., Sharma Kharkwal, S., Drewe, W. C., De Matteis, C. I., Evangelopoulos, D., et al. (2021). New InhA inhibitors based on expanded triclosan and di-triclosan analogues to develop a new treatment for tuberculosis. Pharm. (Basel) 14, 361. doi: 10.3390/ph14040361

CrossRef Full Text | Google Scholar

De Jager, V., Gupte, N., Nunes, S., Barnes, G. L., van Wijk, R. C., Mostert, J., et al. (2022). Early bactericidal activity of meropenem plus clavulanate (with or without rifampin) for tuberculosis: The COMRADE randomized, phase 2A clinical trial. Am. J. Respir. Crit. Care Med. 205, 1228–1235. doi: 10.1164/rccm.202108-1976OC

PubMed Abstract | CrossRef Full Text | Google Scholar

Farhat, M. R., Sixsmith, J., Calderon, R., Hicks, N. D., Fortune, S. M., Murray, M. (2019). Rifampicin and rifabutin resistance in 1003 mycobacterium tuberculosis clinical isolates. J. Antimicrob. Chemother. 74, 1477–1483. doi: 10.1093/jac/dkz048

PubMed Abstract | CrossRef Full Text | Google Scholar

Flint, L., Korkegian, A., Parish, T. (2020). InhA inhibitors have activity against non-replicating mycobacterium tuberculosis. PloS One 15, e0239354. doi: 10.1371/journal.pone.0239354

PubMed Abstract | CrossRef Full Text | Google Scholar

Flipo, M., Frita, R., Bourotte, M., Martínez-Martínez, M. S., Boesche, M., Boyle, G. W., et al. (2022). The small-molecule SMARt751 reverses mycobacterium tuberculosis resistance to ethionamide in acute and chronic mouse models of tuberculosis. Sci. Trans. Med. 14, eaaz6280. doi: 10.1126/scitranslmed.aaz6280

CrossRef Full Text | Google Scholar

Fox, W. (1979). The chemotherapy of pulmonary tuberculosis: A review. Chest 76, 785–796. doi: 10.1378/chest.76.6.785

PubMed Abstract | CrossRef Full Text | Google Scholar

Gibson, E. G., Oviatt, A. A., Cacho, M., Neuman, K. C., Chan, P. F., Osheroff, N. (2019). Bimodal actions of a Naphthyridone/Aminopiperidine-based antibacterial that targets gyrase and topoisomerase IV. Biochemistry 58, 4447–4455. doi: 10.1021/acs.biochem.9b00805

PubMed Abstract | CrossRef Full Text | Google Scholar

GlaxoSmithKline (2022) A randomised, double blind (Sponsor unblinded), placebo-controlled, first time in human study to evaluate the safety, tolerability and pharmacokinetics of single and repeat oral doses and the food effect of GSK2556286 in healthy adult participants (clinicaltrials.gov). Available at: https://clinicaltrials.gov/ct2/show/NCT04472897 (Accessed June 15, 2022).

Google Scholar

Gold, B., Zhang, J., Quezada, L. L., Roberts, J., Ling, Y., Wood, M., et al. (2022). Identification of β-lactams active against Mycobacterium tuberculosis by a consortium of pharmaceutical companies and academic institutions. ACS Infect. Dis. 8, 557–573. doi: 10.1021/acsinfecdis.1c00570

PubMed Abstract | CrossRef Full Text | Google Scholar

Grosset, J. H. (1989). Present status of chemotherapy for tuberculosis. Rev. Infect. Dis. 11 Suppl 2, S347–S352. doi: 10.1093/clinids/11.supplement_2.s347

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, Q., Bi, J., Lin, Q., Ye, T., Wang, Z., Wang, Z., et al. (2022). Whole genome sequencing identifies novel mutations associated with bedaquiline resistance in mycobacterium tuberculosis. Front. Cell Infect. Microbiol. 12. doi: 10.3389/fcimb.2022.807095

CrossRef Full Text | Google Scholar

Hajian, B., Scocchera, E., Shoen, C., Krucinska, J., Viswanathan, K., G-Dayanandan, N., et al. (2019). Drugging the folate pathway in mycobacterium tuberculosis: The role of multi-targeting agents. Cell Chem. Biol. 26, 781–791.e6. doi: 10.1016/j.chembiol.2019.02.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Hartkoorn, R. C., Sala, C., Neres, J., Pojer, F., Magnet, S., Mukherjee, R., et al. (2012). Towards a new tuberculosis drug: pyridomycin – nature’s isoniazid. EMBO Mol. Med. 4, 1032–1042. doi: 10.1002/emmm.201201689

PubMed Abstract | CrossRef Full Text | Google Scholar

Hartkoorn, R. C., Uplekar, S., Cole, S. T. (2014). Cross-resistance between clofazimine and bedaquiline through upregulation of MmpL5 in mycobacterium tuberculosis. Antimicrob. Agents Chemother. 58, 2979–2981. doi: 10.1128/AAC.00037-14

PubMed Abstract | CrossRef Full Text | Google Scholar

Hugonnet, J.-E., Tremblay, L. W., Boshoff, H. I., Barry, C. E., Blanchard, J. S. (2009). Meropenem-clavulanate is effective against extensively drug-resistant mycobacterium tuberculosis. Science 323, 1215–1218. doi: 10.1126/science.1167498

PubMed Abstract | CrossRef Full Text | Google Scholar

Jarlier, V., Nikaido, H. (1994). Mycobacterial cell wall: structure and role in natural resistance to antibiotics. FEMS Microbiol. Lett. 123, 11–18. doi: 10.1111/j.1574-6968.1994.tb07194.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Javid, B., Sorrentino, F., Toosky, M., Zheng, W., Pinkham, J. T., Jain, N., et al. (2014). Mycobacterial mistranslation is necessary and sufficient for rifampicin phenotypic resistance Proc Natl Acad Sci USA. 111(3), 1132–1137. doi: 10.1073/pnas.1317580111.

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnson, E. O., LaVerriere, E., Office, E., Stanley, M., Meyer, E., Kawate, T., et al. (2019). Large-Scale chemical-genetics yields new m. tuberculosis inhibitor classes. Nature 571, 72–78. doi: 10.1038/s41586-019-1315-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Kai, L., Lici, A. S-B., Xinxin, F., Ashutosh, U., Venugopal, P, Benoit, L., et al. (2014). Multitarget drug discovery for tuberculosis and other infectious diseases. J. medicinal Chem. 57(7), 3126–39. doi: 10.1021/jm500131s.

CrossRef Full Text | Google Scholar

Kirsch, S. H., Haeckl, F. P. J., Müller, R. (2022). Beyond the approved: target sites and inhibitors of bacterial RNA polymerase from bacteria and fungi. Nat. Prod. Rep. 39(6), 1226–1263. doi: 10.1039/D1NP00067E

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumari, M., Waseem, M., Subbarao, N. (2022). Discovery of multi-target mur enzymes inhibitors with anti-mycobacterial activity through a scaffold approach. J. Biomol Struct. Dyn 1–22. doi: 10.1080/07391102.2022.2040593

CrossRef Full Text | Google Scholar

Kurz, S. G., Bonomo, R. A. (2012). Reappraising the use of β-lactams to treat tuberculosis. Expert Rev. Anti Infect. Ther. 10, 999–1006. doi: 10.1586/eri.12.96

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, M., Lee, J., Carroll, M. W., Choi, H., Min, S., Song, T., et al. (2012). Linezolid for treatment of chronic extensively drug-resistant tuberculosis. N Engl. J. Med. 367, 1508–1518. doi: 10.1056/NEJMoa1201964

PubMed Abstract | CrossRef Full Text | Google Scholar

Libardo, M. D. J., Boshoff, H. I. M., Barry, C. E. (2018). The present state of the tuberculosis drug development pipeline. Curr. Opin. Pharmacol. 42, 81–94. doi: 10.1016/j.coph.2018.08.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, M.-C., Lu, J., Lu, Y., Xiao, T.-Y., Liu, H.-C., Lin, S.-Q., et al. (2021). rpoB mutations and effects on rifampin resistance in mycobacterium tuberculosis. Infect. Drug Resist. 14, 4119–4128. doi: 10.2147/IDR.S333433

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, W., Mandal, S., Degen, D., Liu, Y., Ebright, Y. W., Li, S., et al. (2017). Structural basis of mycobacterium tuberculosis transcription and transcription inhibition. Mol. Cell 66, 169–179.e8. doi: 10.1016/j.molcel.2017.03.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, W., Upadhyay, A., Fontes, F. L., North, E. J., Wang, Y., Crans, D. C., et al. (2014). Novel insights into the mechanism of inhibition of MmpL3, a target of multiple pharmacophores in mycobacterium tuberculosis. Antimicrob. Agents Chemother. 58, 6413–6423. doi: 10.1128/AAC.03229-14

PubMed Abstract | CrossRef Full Text | Google Scholar

Louw, G. E., Warren, R. M., Gey van Pittius, N. C., McEvoy, C. R. E., Van Helden, P. D., Victor, T. C. (2009). A balancing act: efflux/influx in mycobacterial drug resistance. Antimicrob. Agents Chemother. 53, 3181–3189. doi: 10.1128/AAC.01577-08

PubMed Abstract | CrossRef Full Text | Google Scholar

Machado, D., Coelho, T. S., Perdigão, J., Pereira, C., Couto, I., Portugal, I., et al. (2017). Interplay between mutations and efflux in drug resistant clinical isolates of mycobacterium tuberculosis. Front. Microbiol. 8. doi: 10.3389/fmicb.2017.00711

PubMed Abstract | CrossRef Full Text | Google Scholar

Mandal, S., Njikan, S., Kumar, A., Early, J. V., Parish, T. (2019). The relevance of persisters in tuberculosis drug discovery. Microbiol. (Reading) 165, 492–499. doi: 10.1099/mic.0.000760

CrossRef Full Text | Google Scholar

Manjunatha, U. H., Smith, P. W. (2015). Perspective: Challenges and opportunities in TB drug discovery from phenotypic screening. Bioorg Med. Chem. 23, 5087–5097. doi: 10.1016/j.bmc.2014.12.031

PubMed Abstract | CrossRef Full Text | Google Scholar

Martínez-Hoyos, M., Perez-Herran, E., Gulten, G., Encinas, L., Álvarez-Gómez, D., Alvarez, E., et al. (2016). Antitubercular drugs for an old target: GSK693 as a promising InhA direct inhibitor. EBioMedicine 8, 291–301. doi: 10.1016/j.ebiom.2016.05.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Mboowa, G., Namaganda, C., Ssengooba, W. (2014). Rifampicin resistance mutations in the 81 bp RRDR of rpoB gene in mycobacterium tuberculosis clinical isolates using Xpert®MTB/RIF in Kampala, Uganda: a retrospective study. BMC Infect. Dis. 14, 481. doi: 10.1186/1471-2334-14-481

PubMed Abstract | CrossRef Full Text | Google Scholar

McNeil, M. B., Dennison, D. D., Shelton, C. D., Parish, T. (2017). In vitro isolation and characterization of oxazolidinone-resistant mycobacterium tuberculosis. Antimicrob. Agents Chemother. 61, e01296–e01217. doi: 10.1128/AAC.01296-17

PubMed Abstract | CrossRef Full Text | Google Scholar

Mitchison, D. A. (1985). The action of antituberculosis drugs in short-course chemotherapy. Tubercle 66, 219–225. doi: 10.1016/0041-3879(85)90040-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Nagaraja, V., Godbole, A. A., Henderson, S. R., Maxwell, A. (2017). DNA Topoisomerase I and DNA gyrase as targets for TB therapy. Drug Discovery Today 22, 510–518. doi: 10.1016/j.drudis.2016.11.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Nuermberger, E. L., Martínez-Martínez, M. S., Sanz, O., Urones, B., Esquivias, J., Soni, H., et al. (2022). GSK2556286 is a novel antitubercular drug candidate effective in vivo with the potential to shorten tuberculosis treatment. Antimicrob Agents Chemother. 66 (6), e00132-22 doi: 10.1128/aac.00132-22

CrossRef Full Text | Google Scholar

Oh, S., Trifonov, L., Yadav, V. D., Barry, C. E., Boshoff, H. I. (2021). Tuberculosis drug discovery: A decade of hit assessment for defined targets. Front. Cell. Infect. Microbiol. 11. doi: 10.3389/fcimb.2021.611304

CrossRef Full Text | Google Scholar

Pan, P., Tonge, P. J. (2012). Targeting InhA, the FASII enoyl-ACP reductase: SAR studies on novel inhibitor scaffolds. Curr. Top. Med. Chem. 12, 672–693. doi: 10.2174/156802612799984535

PubMed Abstract | CrossRef Full Text | Google Scholar

Parish, T. (2020). In vitro drug discovery models for mycobacterium tuberculosis relevant for host infection. Expert Opin. Drug Discovery 15, 349–358. doi: 10.1080/17460441.2020.1707801

CrossRef Full Text | Google Scholar

Pasipanodya, J. G., Gumbo, T. (2011). A new evolutionary and pharmacokinetic-pharmacodynamic scenario for rapid emergence of resistance to single and multiple anti-tuberculosis drugs. Curr. Opin. Pharmacol. 11, 457–463. doi: 10.1016/j.coph.2011.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Punetha, A., Green, K. D., Garzan, A., Thamban Chandrika, N., Willby, M. J., Pang, A. H., et al. (2021). Structure-based design of haloperidol analogues as inhibitors of acetyltransferase eis from mycobacterium tuberculosis to overcome kanamycin resistance. RSC Med. Chem. 12, 1894–1909. doi: 10.1039/d1md00239b

PubMed Abstract | CrossRef Full Text | Google Scholar

Punetha, A., Ngo, H. X., Holbrook, S. Y. L., Green, K. D., Willby, M. J., Bonnett, S. A., et al. (2020). Structure-guided optimization of inhibitors of acetyltransferase eis from mycobacterium tuberculosis. ACS Chem. Biol. 15, 1581–1594. doi: 10.1021/acschembio.0c00184

PubMed Abstract | CrossRef Full Text | Google Scholar

Rai, D., Mehra, S. (2021). The mycobacterial efflux pump EfpA can induce high drug tolerance to many antituberculosis drugs, including moxifloxacin, in mycobacterium smegmatis. Antimicrob. Agents Chemother. 65, e0026221. doi: 10.1128/AAC.00262-21

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramón-García, S., González del Río, R., Villarejo, A. S., Sweet, G. D., Cunningham, F., Barros, D., et al. (2016). Repurposing clinically approved cephalosporins for tuberculosis therapy. Sci. Rep. 6, 34293. doi: 10.1038/srep34293

PubMed Abstract | CrossRef Full Text | Google Scholar

Rawat, R., Whitty, A., Tonge, P. J. (2003). The isoniazid-NAD adduct is a slow, tight-binding inhibitor of InhA, the mycobacterium tuberculosis enoyl reductase: adduct affinity and drug resistance. Proc. Natl. Acad. Sci. U.S.A. 100, 13881–13886. doi: 10.1073/pnas.2235848100

PubMed Abstract | CrossRef Full Text | Google Scholar

Robertson, G. T., Ektnitphong, V. A., Scherman, M. S., McNeil, M. B., Dennison, D., Korkegian, A., et al (2019). Efficacy and Improved Resistance Potential of a Cofactor-Independent InhA Inhibitor of Mycobacterium tuberculosis in the C3HeB/FeJ Mouse Model. Antimicrob. Agents Chemother. 63, e02071–18. doi: 10.1128/AAC.02071-18

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodrigues, L., Cravo, P., Viveiros, M. (2020). Efflux pump inhibitors as a promising adjunct therapy against drug resistant tuberculosis: a new strategy to revisit mycobacterial targets and repurpose old drugs. Expert Rev. Anti Infect. Ther. 18, 741–757. doi: 10.1080/14787210.2020.1760845

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodrigues, L., Parish, T., Balganesh, M., Ainsa, J. A. (2017). Antituberculosis drugs: reducing efflux=increasing activity. Drug Discovery Today 22, 592–599. doi: 10.1016/j.drudis.2017.01.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodriguez, F., Saffon, N., Sammartino, J. C., Degiacomi, G., Pasca, M. R., Lherbet, C. (2020). First triclosan-based macrocyclic inhibitors of InhA enzyme. Bioorg Chem. 95, 103498. doi: 10.1016/j.bioorg.2019.103498

PubMed Abstract | CrossRef Full Text | Google Scholar

Saxena, S., Samala, G., Renuka, J., Sridevi, J. P., Yogeeswari, P., Sriram, D. (2015). Development of 2-amino-5-phenylthiophene-3-carboxamide derivatives as novel inhibitors of mycobacterium tuberculosis DNA GyrB domain. Bioorganic Medicinal Chem. 23, 1402–1412. doi: 10.1016/j.bmc.2015.02.032

CrossRef Full Text | Google Scholar

Seifert, M., Catanzaro, D., Catanzaro, A., Rodwell, T. C. (2015). Genetic mutations associated with isoniazid resistance in mycobacterium tuberculosis: A systematic review. PloS One 10, e0119628. doi: 10.1371/journal.pone.0119628

PubMed Abstract | CrossRef Full Text | Google Scholar

Shirude, P. S., Madhavapeddi, P., Tucker, J. A., Murugan, K., Patil, V., Basavarajappa, H., et al. (2013). Aminopyrazinamides: Novel and specific GyrB inhibitors that kill replicating and nonreplicating mycobacterium tuberculosis. ACS Chem. Biol. 8, 519–523. doi: 10.1021/cb300510w

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, P., Jain, A., Dixit, P., Prakash, S., Jaiswal, I., Venkatesh, V., et al. (2015). Prevalence of gyrA and b gene mutations in fluoroquinolone-resistant and -sensitive clinical isolates of mycobacterium tuberculosis and their relationship with MIC of ofloxacin. J. Antibiotics 68, 63–66. doi: 10.1038/ja.2014.95

CrossRef Full Text | Google Scholar

Singh, P. K., Singh, U., Jain, A. (2021). Emergence of specific gyrA mutations associated high-level fluoroquinolone-resistant mycobacterium tuberculosis among multidrug-resistant tuberculosis cases in north India. Microbial Drug Resistance 27, 647–651. doi: 10.1089/mdr.2020.0240

PubMed Abstract | CrossRef Full Text | Google Scholar

Šink, R., Sosič, I., Živec, M., Fernandez-Menendez, R., Turk, S., Pajk, S., et al. (2015). Design, synthesis, and evaluation of new thiadiazole-based direct inhibitors of enoyl acyl carrier protein reductase (InhA) for the treatment of tuberculosis. J. Med. Chem. 58, 613–624. doi: 10.1021/jm501029r

PubMed Abstract | CrossRef Full Text | Google Scholar

Soudani, A., Hadjfredj, S., Zribi, M., Messaoud, T., Masmoudi, A., Majed, B., et al. (2010). First report of molecular characterization of fluoroquinolone-resistant mycobacterium tuberculosis isolates from a Tunisian hospital. Clin. Microbiol. Infection 16, 1454–1457. doi: 10.1111/j.1469-0691.2010.03087.x

CrossRef Full Text | Google Scholar

Stokes, S. S., Vemula, R., Pucci, M. J. (2020). Advancement of GyrB inhibitors for treatment of infections caused by mycobacterium tuberculosis and non-tuberculous mycobacteria. ACS Infect. Dis. 6, 1323–1331. doi: 10.1021/acsinfecdis.0c00025

PubMed Abstract | CrossRef Full Text | Google Scholar

Telenti, A., Imboden, P., Marchesi, F., Lowrie, D., Cole, S., Colston, M. J., et al. (1993). Detection of rifampicin-resistance mutations in mycobacterium tuberculosis. Lancet 341, 647–650. doi: 10.1016/0140-6736(93)90417-f

PubMed Abstract | CrossRef Full Text | Google Scholar

Tiberi, S., Buchanan, R., Caminero, J. A., Centis, R., Arbex, M. A., Salazar, M., et al. (2017). The challenge of the new tuberculosis drugs. La Presse Médicale 46, e41–e51. doi: 10.1016/j.lpm.2017.01.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Tseng, S.-T., Tai, C.-H., Li, C.-R., Lin, C.-F., Shi, Z.-Y. (2015). The mutations of katG and inhA genes of isoniazid-resistant mycobacterium tuberculosis isolates in Taiwan. J. Microbiology Immunol. Infection 48, 249–255. doi: 10.1016/j.jmii.2013.08.018

CrossRef Full Text | Google Scholar

Vannelli, T. A., Dykman, A., Ortiz de Montellano, P. R. (2002). The antituberculosis drug ethionamide is activated by a flavoprotein monooxygenase∗. J. Biol. Chem. 277, 12824–12829. doi: 10.1074/jbc.M110751200

PubMed Abstract | CrossRef Full Text | Google Scholar

Vilchèze, C., Jacobs, W. R. (2014). Resistance to isoniazid and ethionamide in mycobacterium tuberculosis: Genes, mutations, and causalities. Microbiol. Spectr. 2, MGM2-0014–2013. doi: 10.1128/microbiolspec.MGM2-0014-2013

CrossRef Full Text | Google Scholar

Willand, N., Dirié, B., Carette, X., Bifani, P., Singhal, A., Desroses, M., et al. (2009). Synthetic EthR inhibitors boost antituberculous activity of ethionamide. Nat. Med. 15, 537–544. doi: 10.1038/nm.1950

PubMed Abstract | CrossRef Full Text | Google Scholar

Willby, M. J., Green, K. D., Gajadeera, C. S., Hou, C., Tsodikov, O. V., Posey, J. E., et al. (2016). Potent inhibitors of acetyltransferase eis overcome kanamycin resistance in mycobacterium tuberculosis. ACS Chem. Biol. 11, 1639–1646. doi: 10.1021/acschembio.6b00110

PubMed Abstract | CrossRef Full Text | Google Scholar

World Health Organization (2021) Global tuberculosis report 2021. Available at: https://www.who.int/publications-detail-redirect/9789240037021 (Accessed January 17, 2022).

Google Scholar

Xia, Y., Zhou, Y., Carter, D. S., McNeil, M. B., Choi, W., Halladay, J., et al. (2018). Discovery of a cofactor-independent inhibitor of mycobacterium tuberculosis InhA. Life Sci. Alliance 1, e201800025. doi: 10.26508/lsa.201800025

PubMed Abstract | CrossRef Full Text | Google Scholar

Zaunbrecher, M. A., Sikes, R. D., Metchock, B., Shinnick, T. M., Posey, J. (2009). Overexpression of the chromosomally encoded aminoglycoside acetyltransferase eis confers kanamycin resistance in mycobacterium tuberculosis. E. Proc. Natl. Acad. Sci. U.S.A. 106, 20004–20009. doi: 10.1073/pnas.0907925106

CrossRef Full Text | Google Scholar

Zaw, M. T. (2018). Mutations inside rifampicin-resistance determining region of rpoB gene associated with rifampicin-resistance in mycobacterium tuberculosis. J. Infection Public Health 11, 605–610. doi: 10.1016/j.jiph.2018.04.005.

CrossRef Full Text | Google Scholar

Zhang, Y., Heym, B., Allen, B., Young, D., Cole, S. (1992). The catalase-peroxidase gene and isoniazid resistance of mycobacterium tuberculosis. Nature 358, 591–593. doi: 10.1038/358591a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Yew, W. W. (2009). Mechanisms of drug resistance in mycobacterium tuberculosis. Int. J. Tuberculosis Lung Dis. 13, 1320–1330

Google Scholar

Zhu, N., Lin, Y., Li, D., Gao, N., Liu, C., You, X., et al. (2015). Identification of an anti-TB compound targeting the tyrosyl-tRNA synthetase. J. Antimicrobial Chemotherapy 70, 2287–2294. doi: 10.1093/jac/dkv110

CrossRef Full Text | Google Scholar

Zhu, N., Wang, X., Li, D., Lin, Y., You, X., Jiang, J., et al. (2018). IMB-T130 targets 3-dehydroquinate synthase and inhibits mycobacterium tuberculosis. Sci. Rep. 8, 17439. doi: 10.1038/s41598-018-35701-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Mycobacterium tuberculosis, antibiotic resistance, drug discovery, antibiotic tolerance, antibacterial

Citation: Bhagwat A, Deshpande A and Parish T (2022) How Mycobacterium tuberculosis drug resistance has shaped anti-tubercular drug discovery. Front. Cell. Infect. Microbiol. 12:974101. doi: 10.3389/fcimb.2022.974101

Received: 20 June 2022; Accepted: 18 August 2022;
Published: 09 September 2022.

Edited by:

Ben Gold, Weill Cornell Medicine, United States

Reviewed by:

Michael Berney, Albert Einstein College of Medicine, United States
Gregory T. Robertson, Colorado State University, United States

Copyright © 2022 Bhagwat, Deshpande and Parish. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Tanya Parish, tanya.parish@seattlechildrens.org

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.