Abstract
Drug-resistant tuberculosis is a substantial health-care concern worldwide. Despite culture-based methods being considered the gold standard for drug susceptibility testing, molecular methods provide rapid information about the Mycobacterium tuberculosis mutations associated with resistance to anti-tuberculosis drugs. This consensus document was developed on the basis of a comprehensive literature search, by the TBnet and RESIST-TB networks, about reporting standards for the clinical use of molecular drug susceptibility testing. Review and the search for evidence included hand-searching journals and searching electronic databases. The panel identified studies that linked mutations in genomic regions of M tuberculosis with treatment outcome data. Implementation of molecular testing for the prediction of drug resistance in M tuberculosis is key. Detection of mutations in clinical isolates has implications for the clinical management of patients with multidrug-resistant or rifampicin-resistant tuberculosis, especially in situations when phenotypic drug susceptibility testing is not available. A multidisciplinary team including clinicians, microbiologists, and laboratory scientists reached a consensus on key questions relevant to molecular prediction of drug susceptibility or resistance to M tuberculosis, and their implications for clinical practice. This consensus document should help clinicians in the management of patients with tuberculosis, providing guidance for the design of treatment regimens and optimising outcomes.
Introduction
According to the latest estimates by WHO1, tuberculosis affected 10·6 million people and caused 1·6 million deaths in 2021. That these deaths continue to occur despite the availability of curative antimicrobial regimens is a testimony to the difficulty of diagnosing disease and designing, administering, and monitoring tuberculosis treatment. The rise of tuberculosis resistance to isoniazid and rifampicin has seriously complicated treatment for the estimated 450 000 people each year who develop disease resistance to these first-line antimycobacterial agents.1 Definitions of resistance, including multidrug-resistant (MDR) and extensively drug-resistant (XDR) tuberculosis are shown in table 1.
Table 1:
2006 WHO definitions2 | 2021 WHO definitions3 | |
---|---|---|
RR tuberculosis | Rifampicin | Rifampicin |
MDR tuberculosis | Rifampicin; isoniazid | Rifampicin; isoniazid |
Pre-extensively drug-resistant tuberculosis | Not defined | MDR or RR tuberculosis; fluoroquinolone* |
Extensively drug-resistant tuberculosis | MDR or RR tuberculosis; a fluoroquinolone*; second-line injectable drugs† | MDR or RR tuberculosis; a fluoroquinolone*; and bedaquiline or linezolid, or both |
RR=rifampicin-resistant. MDR=multidrug-resistant.
Fluoroquinolone (ie, levofloxacin or moxifloxacin).
Second-line injectable drugs (ie, amikacin, capreomycin, and kanamycin).
Development of three essential capacities regarding tuberculosis treatment will be needed to bring the disease under control while minimising the emergence of resistance to currently available agents: optimising regimen composition, optimising regimen duration, and minimising toxicity from the components of the regimen. This consensus statement is directed at advancing the understanding of the first of these, namely optimising regimen composition.
Despite the attractiveness of a one-size-fits-all regimen strategy, having a single regimen that can be used for all patients with drug-resistant tuberculosis is difficult for several reasons. First, there will always be patients who cannot tolerate the usual agents and will need to have individualised regimens that address their intolerance. Second, development of resistance to the new agents, although hopefully minimised, can never be completely prevented; thus, some patients will require treatment with regimens that do not contain some of the more effective agents as the organisms causing their disease will be resistant to these agents. Third, in some patients, the use of concomitant medications, such as antiretroviral agents, will preclude the use of certain antimycobacterial compounds because of drug–drug interactions. Finally, compensatory mutations and epistasis render resistant clinical strains differentially susceptible to different second-line agents. Thus, prompt and accurate identification of the susceptibility pattern of a patient’s Mycobacterium tuberculosis infecting strain is essential.
For many decades, M tuberculosis susceptibility testing has been done in specialised laboratories with culture-based methods. These methods are limited by the slow growth of the organisms, such that results are not available for many weeks after disease detection. This delay leads to uncertainty for clinicians during the first weeks of treatment, a crucial time period for getting the disease under control and preventing further tuberculosis transmission. Fortunately, some developments in rapid sequencing and molecular identification of critical resistance mutations have led to the ability to predict drug susceptibility or resistance within hours to days after diagnosis with high diagnostic accuracy.4,5
We summarise progress made in developing tools to enable rapid identification of resistance to the agents used in tuberculosis treatment. As such, we update our consensus statement on this topic from 2016, with the substantial progress that has been made since publication.6 Taken together, we believe that this updated consensus provides important and clinically useful guidance to aid the design of effective tuberculosis treatment regimens and to ensure optimal therapy outcomes.
Methods
This document has been produced by physicians, microbiologists, molecular biologists, and clinical epidemiologists of the TBnet (http://www.tbnet.eu) and RESIST-TB (http://www.resisttb.org) networks to reach a consensus in reporting standards in the clinical use of M tuberculosis molecular drug susceptibility testing (DST) results. Chapter leaders reviewed the available literature, and the targeted search for evidence included hand-searching journals and searching electronic databases including MEDLINE and PubMed. The search targeted articles published in English, before June 1, 2022, related to the evolution of drug resistance in clinical isolates, principles of phenotypic and genotypic drug resistance testing, and the impact of mutations detected by genotypic tests on phenotypic DST and clinical outcomes. In addition, relevant documents published by WHO were included. Consensus statements were developed in a stepwise approach, as previously described (panel 1).6–10
Panel 1: Stepwise consensus statements.
Step 1: preliminary proposals for key recommendations were drafted by the coordinating authors (JD, CL, CRHJr). All coauthors were asked to provide alternative statements
Step 2: alternative statements were collected from coauthors
Step 3: coauthors were asked to select one preferred statement among the alternative statements. The coauthors were masked to the vote
Step 4: for each recommendation, the statement that received the most votes was selected for inclusion in the manuscript
Step 5: all coauthors were asked to indicate their agreement, disagreement, or whether they preferred to abstain from a decision
Evolution of drug resistance in clinical M tuberculosis isolates
M tuberculosis bacteria are continuing to develop resistance to both old and new drugs via spontaneous mutations of the bacterial chromosome.11 Mutations include mostly single-nucleotide polymorphisms, but insertions and deletions can also occur. Horizontal gene transfer (eg, mediated by plasmids) has no role in the evolution of drug resistance in M tuberculosis.12 The most common drug resistance mechanisms comprise the modification of the drug target itself, increased expression of the drug target, downregulation or abrogation of prodrug-activating enzymes, upregulation of drug efflux pumps, altered permeability, and phase variation.12,13 The probability of acquiring drug resistance through spontaneous mutations and selection depends on the antibiotic (eg, the estimated frequency for rifampicin is 10−8, and 10−6 for isoniazid).14 Moreover, studies suggest that mutation rates can differ between strains of different M tuberculosis lineages, which influences the probability of acquiring drug-resistant mutations.15–17
When suboptimal treatment regimens are used, drug-resistant M tuberculosis subpopulations can be selected in a patient.18–21 The main reasons for ineffective therapy include improper intake of drug regimens, addition of single drugs to ineffective treatment regimens, inadequate drug supplies, quality of the anti-tuberculosis drugs, individual differences in pharmacokinetics and pharmacodynamics,22 intrinsic strain differences, drug tolerance, and low-level resistance.23,24 Some research highlights the heterogeneity of antibiotic concentrations within the tuberculous granuloma.25 A lack of granuloma vascularisation and diffusion barriers towards necrotic foci can lead to drastically altered pharmacodynamics and pharmacokinetic correlations of individual antibiotics as compared with blood plasma measurements.25,26 The resulting heterogeneity of granuloma micro-environments in a single patient probably drives the microevolution of M tuberculosis subpopulations, which can harbour different drug resistance mutations and can coexist in separate lesions.27–29 This intrapatient M tuberculosis diversity also influences the performance of drug resistance assays, and should therefore be considered in routine diagnostics and treatment.29–31
Diagnostic delays and misdiagnosis of patients with MDR tuberculosis can further amplify the evolution of resistance and virulence determinants in M tuberculosis strains.32–34 Outbreaks of drug-resistant strains have been observed in different settings globally,35–37 suggesting that these strains have few or no fitness costs, challenging the previous dogma of a transmission detriment of MDR M tuberculosis strains. However, no-cost or low-cost resistance mutations dearly exist, and secondary mutations can occur that compensate for the initial fitness defect of resistant mutants.38–40 This molecular interaction is an example of epistasis, and might also influence antibiotic resistance and compensatory evolution in strains of different M tuberculosis lineages.41,42 In multiple studies, compensatory mechanisms were suggested to be associated with an increased transmission risk, eventually leading to the expansion of MDR clones in different areas worldwide.35–37,43
Principles of phenotypic drug resistance testing
Mycobacterial phenotypic DST assesses the ability of the organism to grow in the presence of the antibiotic. The various phenotypic DST methods are detailed elsewhere.44–46 The commonly used proportion method relies on the premise that if more than 1% of the organisms in a given population is resistant to the critical concentration of a drug, the population is defined as being resistant. These critical concentrations separate the susceptible populations from resistant ones, and are specific to the phenotypic DST method and media.
Mycobacterial phenotypic DST is technically difficult, and its interpretation is challenged by the molecular assays’ detection of resistance mutations. Laboratory errors occur, and appropriate quality-management systems and proficiency testing are vital.46–48 Phenotypic DST accuracy is also influenced by the prevalence of resistant strains and the level of this resistance. In settings where low-level resistant strains prevail, phenotypic DST can correlate less well with clinical outcomes, in contrast to settings where high-level resistance is common.49 In isolates with low-level resistance, high doses of drugs such as rifampicin, isoniazid, and fluoroquinolones can be clinically effective.50
Two systematic reviews of the evidence supporting critical concentrations in phenotypic DST testing were done by WHO.51,52 Although many of the current critical concentrations were reaffirmed, important changes were proposed, most notably that the critical concentration of rifampicin for testing on solid 7H10 medium and in Mycobacteria Growth Indicator Tube medium (Becton Dickinson, Franklin Lakes, NJ, USA) be lowered from 1 mg/L to 0·5 mg/L. WHO also introduced clinical breakpoints for moxifloxacin. Clinical breakpoints are the concentrations delineating strains that might still respond to therapy with higher doses of the agent. The Clinical and Laboratory Standards Institute also defined clinical breakpoints for isoniazid.45 Other important observations include few exploitable published data for critical concentrations of newer drugs such as bedaquiline;53–55 and scarce data about reproducibility for these newer drugs. As more data become available, critical concentrations will need to be re-evaluated.
The reduced critical concentration of rifampicin could enable the detection of borderline mutations in the rpoB gene, which are considered to be of clinical relevance but only result in modest minimum inhibitory concentration increase. This reduced critical concentration of rifampicin would only reduce rather than eliminate the discordance between genotype and phenotype. Minimum inhibitory concentration distributions of bacterial populations with low-level resistance phenotypes might overlap with the minimum inhibitory concentration distributions of wild-type isolates, resulting in poor reproducibility of categorical phenotypic DST. This overlap could be also the case for other drugs like bedaquiline and levofloxadn (see section on molecular testing).
Although the use of critical concentrations to distinguish susceptible isolates from non-susceptible isolates is appealing as it is well standardised, more granular phenotypic DST data (such as minimum inhibitory concentrations with broth microdilution) could facilitate better understanding of the correlation between phenotypic DST, genotype, and treatment outcome—especially if associated with pharmacokinetic and pharmacodynamics data. Hence, quantitative drug susceptibility measures need to be implemented in diagnostic mycobacteriology.56,57 The first objective of the newly formed European Committee on Antimicrobial Susceptibility Testing subcommittee on antimycobacterial susceptibility testing (EUCAST-AMST) was to set a reference method for minimum inhibitory concentration determination of M tuberculosis. This standardised method58,59 will be used for both new and old anti-tuberculosis drugs to assess epidemiological cutoffs. This method will allow new compounds to be evaluated and compared, and will offer a basis for calibrating phenotypic DST methods used in the field, commertial or not, against a standard reference method.60 In addition, the CRyPTIC Consortium61 presented the epidemiological cutoffs for 13 anti-tuberculosis compounds, including bedaquiline and delamanid, from clinical isolates collected worldwide, for a more widespread adoption of the broth microdilution test. Finally, a 2022 WHO document provided a standardised methodology to facilitate the improvement of the broth microdilution plate method for clinical use, which represents a promising solution for comprehensive and quality-assured phenotypic DST.62
Principles of genotypic DST
The fundamental principle of genotypic DST is to predict phenotypic resistance on the basis of genotype (ie, the genetic variants detected). Drug resistance in M tuberculosis strains is due to variations of their genome (either single-nucleotide polymorphisms or insertions and deletions) mapping to spedfic genetic loci.63 Silent mutations are usually not responsible for resistance, with few documented exceptions.64 Importantly, not all mutations found at the genetic loci involved in drug resistance are responsible for a resistant phenotype, as phylogenetic markers and neutral polymorphisms can also be located in these regions.65–69 Whereas for several anti-tuberculosis drugs the association between sequence variations and phenotypic resistance development is well documented, our knowledge remains incomplete for others.63 The frequency of resistance mutations observed in clinical M tuberculosis strains varies by resistance genes, local epidemiology, and M tuberculosis population structure in different geographical settings.70,71 Generally, drug resistance in M tuberculosis is emerging at strain level, but there are a few exceptions where intrinsic resistance is found in strains of a specific phylogenetic clade.65,72,73
In the past 15 years, rapid commercial genotypic DST assays have become available to detect drug resistance in clinical M tuberculosis strains by interrogating the presence of known resistance-conferring variants (table 2).5,75–80 Performance of these genotypic DST tests, and implications for their use in different application scenarios are outlined in WHO guidelines.81,82 The line probe assay GenoType MTBDRsl VER 2.0 (HAIN Lifescience/Bruker, Nehren, Germany) and the cartridge-based Xpert MTB/XDR (Cepheid, Sunnyvale, CA, USA) are the only assays currently endorsed for the detection of resistance to fluoroquinolones, thus allowing the identification of pre-extensively drug-resistant (pre-XDR) tuberculosis according to the updated definition.3,82,83 Of note, the line probe assay Nipro Genoscholar PZA-TB II (Nipro Corporation, Osaka, Japan) is able to detect pncA mutations associated with pyrazinamide resistance.82,84,85
Table 2:
Xpert MTB/RIF (Cepheid, Sunnyvale, CA, USA) | Xpert MTB/RIF Ultra (Cepheid, Sunnyvale, CA, USA) | Truenat MTB-Rif Dx (MolBio Diagnostics, Goa, India) | GenoType MTBDRplus VER 2.0 (HAIN Lifescience/Bruker, Nehren, Germany) | Nipro NTM+MDRTB detection kit 2 (Nipro Corporation, Osaka, Japan) | GenoType MTBDRsl VER 2.0 (HAIN Lifescience/Bruker, Nehren, Germany) | Xpert MTB/XDR (Cepheid, Sunnyvale, CA, USA) | Nipro Genoscholar PZA-TB II (Nipro Corporation, Osaka, Japan) | Deeplex Myc-TB* (Genoscreen, Lille, France) | Next Gen-RDST* (Illumina, San Diego, CA, USA) | AmpliSeq TB* | |
---|---|---|---|---|---|---|---|---|---|---|---|
Technology | Cartridge-based real-time PCR and molecular beacons | Cartridge-based real-time PCR and sloppy molecular beacons | Chip-based real-time PCR | Line probe assay | Line probe assay | Line probe assay | Cartridge-based real-time PCR and sloppy molecular beacons | Line probe assay | tNGS | tNGS | tNGS |
Genotypic DST approach | Negative hybridisation of wild-type probes | Mutation-specific melting temperature shifts analysis | Melting temperature shifts analysis | Negative hybridisation of wild-type probes or positive hybridisation of specific mutation probes | Negative hybridisation of wild-type probes or positive hybridisation of specific mutation probes | Negative hybridisation of wild-type probes or positive hybridisation of specific mutation probes | Mutation-specific melting temperature shifts analysis | Negative hybridisation of wild-type probes | Specific identification of mutations by sequencing | Single molecule-overlapping reads | Specific identification of mutations by sequencing |
First-line | |||||||||||
Rifampicin (RR) | rpoB RRDR | rpoB RRDR | rpoB RRDR | rpoB RRDR | rpoB RRDR | .. | .. | .. | rpoB fragment (incl. cod. 170,491, and RRDR) | rpoB RRDR | rpoB full gene |
Isoniazid (MDR) | .. | .. | .. | katG codon 315; inhA promoter region | katG codon 315 region; inhA promoter region | .. | katG codon 315 region; inhA promoter region; ahpC promoter region; fabG1 codon 203 region | .. | katG fragment (including codon 315); ahpC fragment (including promoter region); inhA fragment (including promoter region, and codon 94); fabG1 fragment (including promoter region) | katG fragment (including codon 315); inhA promoter region | katG full gene; inhA full gene |
Group A | |||||||||||
Levofloxacin and moxifloxacin (pre-XDR) | .. | .. | .. | .. | .. | gyrA QRDR; gyrB QRDR | gyrA QRDR; gyrB QRDR | .. | gyrA QRDR; gyrB QRDR | gyrA QRDR | gyrA full gene |
Bedaquiline (XDR) | .. | .. | .. | .. | .. | .. | .. | .. | Rv0678 full gene | .. | .. |
Linezolid (XDR) | .. | .. | .. | .. | .. | .. | .. | .. | rplC fragment; rrl fragment | .. | .. |
Group B | |||||||||||
Clofazimine | .. | .. | .. | .. | .. | .. | .. | .. | Rv0678 full gene | .. | .. |
Cycloserine | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. |
Terizidone | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. |
Group C | |||||||||||
Ethambutol | .. | .. | .. | .. | .. | .. | .. | .. | embB fragment (including codon 306) | .. | embB full gene |
Delamanid | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. |
Pretoman id | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. |
Pyrazinamide | .. | .. | .. | .. | .. | .. | .. | pncA full gene | pncA full gene | .. | pncA full gene |
Imipenem-cilastatin | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. |
Meropenem | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. |
Amikacin | .. | .. | .. | .. | .. | rrs position 1401, 1402, and 1484 | rrs 1400 region | .. | rrs full gene | rrs 1400 and 1484 regions | .. |
Streptomycin | .. | .. | .. | .. | .. | .. | .. | .. | rpsL fuIl gene; gidB full gene | .. | rpsL full gene |
Ethionamide and prothionamide | .. | .. | .. | inhA promoter region | .. | .. | inhA promoter region | .. | inhA fragment (including promoter region, and codon 94); fabG1 fragment (including promoter region); ethA full gene | .. | .. |
P-aminosalicylic acid | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. | .. |
No longer recommended | |||||||||||
Kanamycin | .. | .. | .. | .. | .. | rrs position 1401, 1402, and 1484; eis promoter region | rrs 1400 region; eis promoter region | .. | rrs full gene; eis fragment (including promoter region) | rrs 1400 and 1484 regions; eis promoter region | eis full gene |
Capreomycin | .. | .. | .. | .. | .. | rrs position 1401, 1402, and 1484 | rrs 1400 region | .. | rrs full gene; tlyA full gene | rrs 1400 and 1484 regions | .. |
The table focuses on WHO-endorsed methods, and main tNGS assays currently available. Drugs are grouped according to WHO.74 MDR, pre-XDR, and XDR tuberculosis are defined according to WHO.3 Target regions are indicative; for detailed positions covered please refer to the specific manufacturers’ instruction manuals. tNGS=targeted next-generation sequencing. DST=drug susceptibility testing. RR=rifampicin resistance. MDR=multidrug-resistant, pre-XDR=pre-extensively drug-resistant. XDR=extensively drug-resistant. RRDR=rifampicin resistance-determining region. QRDR=quinolone resistance-determining region.
None of the targeted next-generation sequencing approaches are currently endorsed by a WHO recommendation.
Knowledge about the genetic basis of resistance to new or repurposed drugs such as bedaquiline, linezolid, clofazimine, and delamanid remains poor, as is our capability for phenotypic DST.86–88 No rapid molecular commercial assay exists for these drugs, and genotypic DST can be done by sequencing technologies only. In this context, the targeted sequencing assay Deeplex Myc-TB (Genoscreen, Lille, France) is able to provide genotypic DST for 13 drugs, including linezolid, bedaquiline, and clofazimine, by targeting the full sequences of 18 loci associated with drug resistance.89,90 Preliminary studies show that, directly from clinical specimens, Deeplex Myc-TB can frequently be successful, reporting complete resistance predictions in sputum samples with acid-fast bacilli visible on microscopy.89–91 Similar targeted next-generation sequencing approaches exist; however, they are not yet targeting new or repurposed drugs (Next Gen-RDST,92 AmpliSeq TB93). Compared with whole-genome sequencing, targeted next-generation sequencing has the potential to decentralise sequencing-based genotypic DST for individual patient care, especially if coupled with versatile technological and bioinformatic platforms.31,91,94–96 The different genotypic DST assays have varied turnaround times, ranging from 1 working day for Xpert and line probe assay under optimal conditions, to 7–10 working days for targeted next-generation sequencing under operational conditions, when applied to clinical specimens (table 3).
Table 3:
Under optimal conditions | Under operational conditions* | |
---|---|---|
Xpert (Cepheid, Sunnyvale, CA, USA) | 1 working day | 1–2 working days |
Line probe assay | 1 working day | 2–4 working days |
Amplicon sequencing (Deeplex, [Genoscreen, Lille, France]) | 2–3 working days | 7–10 working days |
Whole-genome sequencing | 2–3 working days | 7–10 working days |
Once sample has arrived at the laboratory.
Notably, the technical success with a specimen depends on the number of bacilli, as a low inoculum increases the risk of technical failure. When genotypic DST assays are applied to a positive culture, the probability of obtaining a result increases, but so does the turnaround time.
Current genotypic DST assays cannot be used for treatment monitoring as they detect DNA from both live and dead bacteria, thus a positive result does not imply the viability of the pathogen.97 Similarly, the detection of heteroresistance in clinical specimens by the use of WHO-endorsed rapid molecular tests vary from below 10% to above 75%, depending on the assay and target considered.83,98,99 By contrast, targeted next-generation sequencing assays allow for the detection of even minor subpopulations due to the high coverage they provide.89,100,101 A cost evaluation of the workflow of whole-genome sequencing in eight laboratories in Europe and North America calculated the costs to be 7% cheaper than phenotypic DST.102 In settings with a low tuberculosis burden, the routine use of Xpert shortened the time to tuberculosis diagnosis and treatment, whereas whole-genome sequencing shortened the time to DST and treatment modification when necessary, reducing treatment and hospitalisation costs. The combined use of these two methods was the most cost-effective option and allowed for faster appropriate treatment, and this subsequently reduces transmission, benefiting health, and reducing future treatment costs.103 In India, molecular tests used in the diagnosis of MDR-tuberculosis, such as line probe assays and Xpert, were reported to have considerable advantages for the programmatic management of drug-resistant tuberculosis, including speed, standardisation of testing, potentially high throughput, and reduced laboratory biosafety requirements. They also appeared to be cost effective and helped in detecting missing cases.104 Cost-effectiveness studies of sequencing tests in low-income settings with high tuberculosis burden are needed.
In 2022, a budget impact analysis was done to estimate the costs of introduction and routine use of next-generation sequencing in the National Tuberculosis Programme of Moldova, a country with a high burden of MDR tuberculosis.105 According to the model, next-generation sequencing adoption would require expanded resources compared with conventional phenotypic DST. We are convinced that despite these initial investments, DST strategies using next-generation sequencing will also prove to be cost-effective. However, much needs to be done for the implementation of molecular DST, especially in countries of high tuberculosis burden. A recent study by the TBnet106 documented substantial gaps in the availability of genotypic DST for anti-tuberculosis drugs in the European Region of WHO. These results are especially worrisome in view of the roll-out of the BPaLM regimen (a combination of bedaquiline, pretomanid, linezolid, and moxifloxacin), as many countries currently have no capacity for DST of key components of this regimen.
Relationship between results of genotypic and phenotypic drug resistance testing and clinical outcomes
There are different ways to study the clinical implications of drug resistance mutations. The simplest one is to show that in a case of treatment failure, there is associated drug resistance either phenotypically (ie, minimum inhibitory concentration increase) or genotypically (ie, appearance of a mutation) at the time of tuberculosis diagnosis. This technique was used in the 1960s to show that the phenotypic proportion method correctly identified strains associated with treatment failure.107 It was also initially used to show that rifampicin resistance was associated with rpoB mutations.108 For some genes (table 4), such a correlation has been shown.
Table 4:
Gene | High-confidence mutations associated with drug resistance | Sensitivity of the high-confidence mutations as predictors of phenotypic drug susceptibility111 | Drug mutations for which a dose increase might be considered to overcome resistance at standard dosing* | Effect on treatment outcome | |
---|---|---|---|---|---|
First-line drug | |||||
Rifampicin | rpoB | Most commonly Ser450Leu but >20 other mutations described | 93·8% | Asp435Tyr, His445Leu†, Leu452Pro, Leu430Pro, His445Asn, Ile491Phe | rpoB mutations associated with unfavourable first-line regimen treatment outcome; no difference between disputed and undisputed mutations;49,112,114 rpoB mutations have no effect on MDR treatment outcome115 |
Isoniazid | inhA-mabA | −15 c/t+Ilel94Thr, −15 c/t+Ser94Ala | 91·2%‡ | −15 c/t | inhA mutations increase relapse after first-line treatment and reduce early bactericidal activity of isoniazid 5 mg/kg; inhA −15 c/t mutation has more of an effect than other mutations in inhA116,118; inhA mutations have no effect on MDR treatment outcome (short or long regimen)115,119,120 |
Isoniazid | katG | Ser315Ile, Ser315Asn, Ser315Thr | 91·2%‡ | .. | katG 315 mutations are associated with unfavourable first-line regimen treatment outcome (treatment failure or death) and relapse, have more of an effect than inhA mutations,116,115 and have no effect on MDR treatment outcome (short or long regimen)115,120 |
Group A drug | |||||
Bedaquiline | Rv0678 | Gln22Leu,Thr33Ala, Ser63Arg, Ile67PheSer, Arg72Trp, Arg135Gly, Leu136Pro | 0%§ | l85ins_Gln† | Some studies show a negative effect of initial bedaquiline resistance in relation to Rv0678 mutations on treatment outcome whereas others do not; the emergence of Rv0678 mutations during treatment is associated with worse treatment outcome121–123 |
Bedaquiline | atpE | Asp28Gly, Asp28Val, Ala63Pro | 0%§ | .. | Emergence of atpE mutations during treatment is not always associated with worse treatment outcome124,125 |
Bedaquiline | pepQ | Insufficient data | 0%§ | .. | Unknown |
Levofloxacin and moxifloxacin | gyrA | Gly88Cys, Asp94Gly, Asp94His, Asp94Asn, Asp94Tyr | ~85% | Asp89Asn, Ala90Val, Ser91Pro, Asp94Ala | gyrA 94 mutations delay sputum conversion of MDR tuberculosis regimen; gyrA mutations are selected in case of ofloxacin treatment failure and can predict MDR tuberculosis treatment outcome as efficiently as phenotypic drug susceptibility testing; for gyrA mutations associated with unfavourable MDR tuberculosis treatment outcome, the higher the associated fluoroquinolone minimum inhibitory concentration, the higher the negative effect on treatment outcome (failure or relapse Asp94Asn>Asp94Gly>Ala90Val>Asp94Ala)116,126–130 |
Levofloxacin and moxifloxacin | gyrB | .. | .. | Asp461His†, Asp461Asn†, Asp499Asp†, Ala504Val† | The clinical effect of gyrB mutations on MDR tuberculosis treatment outcome has not been shown but has been shown in a murine model119,131 |
Linezolid | rplC | Cys154Arg | 38·2%¶ | .. | rplC mutations are selected in case of linezolid regimen treatment failure132,133 |
Linezolid | rrl | 2299 g/t, 2814 g/t | 38·2%¶ | .. | rrl mutations are selected in case of linezolid regimen treatment failure132,133 |
Group B drug | |||||
Clofazimine | Rv0678 | Gln22Leu,Thr33Ala, Ser63Arg, Ile67PheSer, Arg72Ter, Gly25Asp, Leu44Pro, Arg135Gly, Leu136Pro, Ser68Arg, >30 mutations described | 0% | .. | Unknown |
Clofazimine | pepQ | Insufficient data | .. | .. | Unknown |
Cycloserine | alr | −8 c/t, Met319Thr, Tyr364Asp, Tyr364Cys, Arg373Leu,Arg373Gly | .. | .. | Unknown |
Amikacin | rrs | 1401 a/g, 1484 a/t | 77·3%‖ | 1402 c/t | rrs mutations are selected in case of kanamycin or capreomycin treatment failure, and predict 4-month sputum culture conversion as efficiently as phenotypic drug susceptibility testing; rrs mutations are not associated with MDR tuberculosis treatment outcome119,126,129,134 |
Amikacin | eis | .. | 77·3%‖ | −14 c/t | eis mutations are not associated with MDR tuberculosis treatment outcome119,129 |
Group C drug | |||||
Streptomycin | rpsL | Lys43Arg, Lys43Thr, Lys88Gln, Lys88Arg | 82·4%** | .. | rpsL mutations predict 4-month sputum culture conversion as efficiently as phenotypic drug susceptibility testing134 |
Streptomycin | rrs | 514 a/c, 514 a/t, 462 c/t, 513 c/t, 517 c/t | 82·4%** | .. | Unknown |
Delamanid | fbiA | Asp49Tyr, Lys250Stop | 6·l%†† | .. | Unknown |
Delamanid | ddn | Trp88Stop | 6·l%†† | .. | Unknown |
Ethambutol | embB | Me306lle, Met306Val, Asp354Ala, Gly406Asp, Gly406Cys, Gly406Ser, Gln497Arg | 86·7%†† | .. | embB mutations have not been associated with M DR tuberculosis treatment outcome in one study119 |
Ethambutol | embC-embA | −8 c/t, −12 c/t, −16 c/t (often in linkage with embB mutations) | 86·7%†† | .. | Unknown |
Ethionamide and prothionamide | inhA | −15 c/t, Ser94Ala | 75·7%§§ | .. | inhA mutations have no impact on MDR tuberculosis treatment outcome (short or long regimen)115,119,120 |
Ethionamide and prothionamide | ethA | Pooled frameshifts and premature stop codons | 75·7%§§ | .. | ethA mutations are selected in case of ethionamide treatment failure, and are associated with unfavourable MDR tuberculosis treatment outcome119,126 |
Imipenem and meropenem | Unknown | Insufficient data | .. | .. | Unknown |
P-aminosali cylic acid | folC | Glu153Ala, Glu153Gly, Ser150Gly, Phe152Ser, Ile43Thr, Ile43Ala, Glu40Gly | .. | .. | Unknown |
P-aminosali cylic acid | ribD | −12 g/a | .. | .. | Unknown |
P-aminosali cylic acid | thyA | .. | .. | .. | thyA mutations are selected in case of p-aminosalicylic acid treatment failure126 |
Pyrazinamide | pncA | >300 mutations described | 72·3% | Val180Ile†,Ala170Val†, Asp110GIy†, Ser65Ala†, Glu37Val† | pncA mutations are associated with delayed sputum culture conversion of MDR tuberculosis treatment; the negative impact of pncA mutations on MDR tuberculosis treatment outcome has been shown in one study but not in two others119,129,135 |
MDR=multidrug resistant.
Agent should not count among active drugs.
Additional data needed.
When inhA-mabA and katG are studied.
When Rv0678, atpE, and pepQ are studied.
When rplC and rrl are studied.
When rrs and eis are studied.
When rpsL and rrs are studied.
When fbiA and ddn are studied.
When embB and embC-embA are studied.
When inhA and ethA are studied.
However, for clinical use, the question is not only whether the mutation correctly identifies a strain with an elevated minimum inhibitory concentration, but also whether this mutation is associated with treatment failure, thus prompting the clinician to consider a change in the treatment regimen. Given that tuberculosis treatment relies on drug combinations, this second demonstration requires that the antibiotic showing a resistant phenotype associated with the mutation has an important impact on the outcome of the drug combination. This second demonstration is true for both drug-susceptible tuberculosis treatment regimens and MDR or rifampicin-resistant tuberculosis regimens, as shown in WHO’s meta-analysis that indicated not all second-line drugs have the same impact on treatment outcome.136 Hence, the drug resistance mutations that will have a strong effect on treatment outcome are those modulating the most effective antibiotics. Another factor to consider is the differential impact of mutations on the minimum inhibitory concentration as highlighted previously. Again, the probability of showing a differential impact on treatment outcome will depend on both the mutation-specific minimum inhibitory concentration effect and on the intrinsic activity of the drug. The more effective or mycobactericidal the drug, and the higher the impact of the mutation on the minimum inhibitory concentration, the more likely the effect on treatment outcome. We have reviewed studies linking the treatment outcome with the presence of a drug mutation (table 4). Of note, the lack of a clinical impact is by no means conclusive; it might only mean that although the mutation reduces the activity of the drug, other drugs in the regimen could facilitate clinical cure. The relationship between mutations and their clinical implications are discussed within the context of each drug type or class.
Another notable benefit of the use of molecular DST is proper initial distinguishing of patients with tuberculosis according to detected resistance patterns. This benefit is extremely important in countries with high MDR and XDR tuberculosis burden, with poor capacity for airborne infection isolation in single-occupant hospital rooms. Taking into consideration molecular DST results can facilitate an optimal distribution of isolation wards and prevent nosocomial transmission of drug-resistant M tuberculosis strains, even when it has low relevance in treatment decisions.
For isoniazid, katG 315 mutations have a higher negative impact on first-line treatment outcome than inhA mutations. Although not analysed in the meta-analysis of individual patient data due to low numbers in the dataset,137 and not listed as a group C drug in recent WHO guidance138, high-dose isoniazid is still used in children, in patients in whom an effective regimen cannot otherwise be constructed (due to lack of drug access, adverse events, resistance to other agents, etc), and in patients with low-level resistance predicted by inhA mutations and other mutations in katG.118,139 Although high-dose isoniazid is still being used as part of an all-oral 9-month bedaquiline-based MDR or rifampicin-resistant tuberculosis regimen in countries like South Africa, and as part of the 9–11-month injectable-based regimen140,141 previously endorsed by WHO in 2019 and 2020 guidance,80,138 this regimen will soon be replaced by alternative 6-month pan-oral regimens like BPaLM.142
For rifampicin, more than 20 mutations in rpoB could have a negative effect on first-line treatment outcome, such as the Ile491Phe mutation found in Eswatini. As approximately 5% of mutations fall outside of the rpoB gene hotspot region, repeat genotypic or phenotypic susceptibility testing should be considered in patients with genotypically rifampicin-susceptible tuberculosis who do not respond to rifampicin-based treatment at 2–3 months. It is also important to consider nonadherence and the possibility of heteroresistance, and to check the adequacy ofblood rifampicin concentrations.143,144
For fluoroquinolones, the gyrA mutations Asp94Asn, Asp94Gly, Ala90Val, and Asp94Ala have a negative effect on MDR or rifampicin-resistant tuberculosis treatment outcome, and that effect is closely linked to the minimum inhibitory concentration generated by the associated mutation.119,126–130 Fluoroquinolone resistance implies pre-XDR tuberculosis. Results from the ZeNix trial145 suggest that successful treatment of MDR or rifampicin-resistant tuberculosis can also be achieved in approximately 90% of patients treated for 6 months with bedaquiline (200 mg twice per day for 14 days, and 200 mg thrice per week thereafter), pretomanid (200 mg once per day), and linezolid (600 mg once per day), without fluoroquinolones. With a conventional treatment regimen that does not include all of the three BPaL drugs, fluoroquinolone resistance is associated with a poorer prognosis.146 Treatment outcomes in fluoroquinolone-resistant MDR or rifampicin-resistant tuberculosis can possibly (but presently unclearly) be improved without these three medicines, if additional group C medicines are added to have at least five active drugs in the treatment regimen. For ethionamide, multiple ethA mutations throughout the gene have a negative effect on MDR tuberculosis treatment outcome.119,126
For bedaquiline, data are scarce about primary or acquired resistance in clinical cohorts and its effect on outcomes. In South Africa, the proportion of patients with a successful outcome was 72% (794/1103) among those with bedaquiline susceptibility compared with 57% (21/37) among those with bedaquiline resistance.123 However, we know that addition of bedaquiline to a background regimen, or when used in combination with other drugs, is associated with a reduction in mortality and improved outcomes.141,147 Thus, clinically significant resistance to bedaquiline would be expected to imply a worse prognosis. There are discrepant results regarding Rv0678 mutations and treatment outcome, probably due to the low minimum inhibitory concentration increase. Unfortunately, phenotypic testing is required to ascertain the presence of bedaquiline resistance, as high-confidence mutations in the atpE and Rv0678 genes are likely to predict less than 10% of phenotypically defined resistance.148 After considering phenotypical cross-resistance with clofazimine86,149 (which is quite frequent), a regimen containing four or five effective drugs would need to be constructed in patients with isolates resistant to bedaquiline.
For linezolid, rrl mutations 2814 g/t and 2270 g/t, and rplC mutation Cysl54Arg are associated with treatment failure. The high-confidence mutations currently predict approximately 75% of phenotypically defined resistance.86 Thus, the addition of such mutations to standardised molecular testing platforms would be useful. When linezolid resistance occurs in combination with fluoroquinolone resistance, it defines XDR tuberculosis, and the design of an appropriate individualised regimen is required.
Although the phase 3 delamanid trial150 did not show improved treatment outcomes, many authorities consider the drug useful for the treatment of patients with resistance beyond MDR or rifampicin-resistant tuberculosis. Mutations encoding drug resistance that predict almost 90% of delamanid resistance involve six genes, and there is emerging evidence of acquired delamanid resistance.151 Incorporation of genotypic testing into standardised molecular platforms would be useful. Pretomanid is now part of recommended 6-month pan-oral regimens, but there are no clear cutoffs, and critical concentration is not yet defined for performing phenotypic DST.55 Nonetheless, a low proportion of phenotypically XDR tuberculosis isolates were found to be genotypically resistant to delamanid and pretomanid.149
For pyrazinamide, there are many mutations in pncA that can confer drug resistance. Mutations in pncA have been related to delayed sputum culture conversion; however, the effect on MDR or rifampicin-resistant tuberculosis treatment outcome has been variable.119,129,135 For ethambutol, embB mutations do not appear to have an effect on treatment outcomes for MDR or rifampicin-resistant tuberculosis.119
For injectables such as amikacin, kanamycin, and capreomycin, the most commonly detected mutations are rrs 1401 a/g, 1402 c/t, 1484 a/t, and eis −14 c/t. No mutations have been shown to have an effect on treatment outcomes for MDR or rifampicin-resistant tuberculosis, which is consistent with the meta-analysis that showed these drugs have poor activity against MDR or rifampicin-resistant tuberculosis.152 Nowadays, amikacin has little applicability but can still be widely used when access to bedaquiline and linezolid is restricted. It can also form part of the regimen in patients with XDR tuberculosis, or resistance beyond XDR tuberculosis when treatment options are scarce. Although an all-oral regimen is recommended by WHO,142 many patients in tuberculosis-endemic countries do not yet have access to such regimens, making such recommendations unrealistic.1,11
Thus, some mutations that compromise the activity of major drugs have an impact on treatment outcome. In this sense, a classification to stratify mutations in order of importance for regimen design and choice of drugs would be useful. For this, the CRyPTIC Consortium, ReSeqTB, contributors to WHO’s surveillance programme, multinational tuberculosis researchers, and public health bodies provided whole-genome sequencing and associated anonymised metadata. Algorithms for identifying variants associated and not associated with resistant phenotypes were adapted from approaches developed by the multinational CRyPTIC Consortium, and the confidence-grading method developed the Seq&Treat project153. The consortium concluded that for levofloxacin, moxifloxacin, and ethambutol, the mutations’ pooled sensitivity was more than 80%. Specificity was over 95% for all drugs except ethionamide (91·4%), moxifloxacin (91·6%), and ethambutol (93·3%). Only two resistance mutations were identified for bedaquiline, delamanid, clofazimine, and linezolid as prevalence of phenotypic resistance was low for these drugs. Finally, the WHO catalogue of mutations in M tuberculosis and their associations with drug resistance were published.154 Based on the evidence presented in this review, we developed consensus recommendations that should help clinicans in the management of patients with TB, through the optimised use of molecular drug resistance testing for M tuberculosis. Consensus recommendations are shown in panel 2.
Panel 2: Consensus recommendations.
-
Should rapid molecular testing for Mycobacterium tuberculosis drug resistance be the gold standard for detection of rifampicin resistance? When should it be done? Should it always be accompanied by phenotypic drug susceptibility testing (DST)?
Rapid molecular testing for the prediction of rifampicin resistance in M tuberculosis (by WHO-endorsed methods) should be done as part of the initial evaluations in the diagnosis of active tuberculosis. Of note, current routinely available rapid molecular testing methods for the prediction of rifampicin resistance in M tuberculosis are not covering all mutations in the rpoB gene that result in M tuberculosis drug resistance. Although not all detections of mutations in the rpoB gene have important implications for regions like Eswatini, these exceptions are of low clinical relevance in other geographical regions. Confirmatory culture-based DST for rifampicin should be done for genotypically susceptible isolates. Resistance inferred by the presence of unidentified mutations (eg, no wild type on rapid molecular testing) should be confirmed by culture-based DST. Identification of mutations recognised as markers of resistance (such as those listed in WHO’s catalogue of mutations) should not be further confirmed by conventional culture-based DST.
19 (76%) agree, five (20%) disagree, and one (4%) abstain.
-
Should rapid molecular testing for M tuberculosis drug resistance be the gold standard for detection of isoniazid resistance? When should it be done? Should it always be accompanied by phenotypic DST?
Isoniazid resistance is the most common drug resistance of M tuberculosis. Rapid molecular testing for the prediction of isoniazid resistance in M tuberculosis (by WHO-endorsed methods) should be done as part of the initial evaluations in the diagnosis of active tuberculosis. Confirmatory culture-based DST for isoniazid should be done for genotypically susceptible isolates. Resistance inferred by the presence of unidentified mutations (eg, no wild type on rapid molecular testing) should be confirmed by culture-based DST. Identification of mutations recognised as markers of resistance (such as those listed in WHO’s catalogue of mutations) should not be further confirmed by conventional culture-based DST.
19 (76%) agree, five (20%) disagree, and one (4%) abstain.
-
When is prediction of M tuberculosis drug resistance towards ethambutol and pyrazinamide recommended, and if so, by which methods?
Rapid molecular testing for the prediction of ethambutol and pyrazinamide resistance in M tuberculosis is not recommended as part of the initial evaluations in the diagnosis of active tuberculosis. However, due to the inherent limitations of culture-based DST for ethambutol and pyrazinamide, the identification of mutations recognised as markers of resistance (such as those listed in WHO’s catalogue of mutations) should rule out the need for further conventional culture-based DST.
20 (80%) agree, two (8%) disagree, and three (12%) abstain.
-
Which molecular methods are available for the prediction of second-line drug resistance to WHO group A, group B, and group C medicines?
The only molecular method available for prediction of resistance to all second-line drugs of WHO group A, group B, and group C is whole-genome sequencing. Amplicon sequencing (Deeplex [Genoscreen, Lille, France]) is available for prediction of resistance to fluoroquinolones, bedaquiline, linezolid, clofazimine, ethambutol, pyrazinamide, amikacin, streptomycin, and ethionamide. Line probe assays are available for prediction of resistance to fluoroquinolones, ethambutol, pyrazinamide, amikacin, and streptomycin. The Xpert MTB/XDR assay (Cepheid, Sunnyvale, CA, USA) can be used for prediction of resistance to isoniazid, fluoroquinolones, amikacin, and ethionamide.
23 (92%) agree, one (4%) disagree, and one (4%) abstain.
-
In which patients, on which specimens, and at which time after diagnosis of tuberculosis should molecular testing for the prediction of resistance to second-line anti-tuberculosis medicines be done?
Additional molecular testing for the prediction of drug resistance to second-line anti-tuberculosis medicines in M tuberculosis should be done as part of the evaluations in the diagnosis of active tuberculosis, as soon as resistance to rifampicin has been suggested by molecular methods, or when rifampicin resistance has been identified by phenotypic DST, on the earliest available specimen. This timing is especially important in patients from countries with a high burden of multidrug-resistant or rifampicin-resistant tuberculosis. Since phenotypic DST for several key second-line anti-tuberculosis medicines is not universally available, implementation of molecular testing for the prediction of drug resistance to second-line anti-tuberculosis medicines in M tuberculosis has high priority for the control of drug-resistant tuberculosis.
25 (100%) agree.
-
How fast can results of molecular drug susceptibility prediction be provided?
Under ideal circumstances, results that predict susceptibility or resistance of M tuberculosis towards rifampicin, isoniazid, fluoroquinolones, amikacin, and ethambutol can be obtained within 1 working day. Results of amplicon sequencing and whole-genome sequencing to predict resistance against second-line anti-tuberculosis medicines can be obtained within 2–3 working days. Under operational conditions, once a sample has arrived at the laboratory, rapid molecular testing results can be obtained within 1–2 working days, line probe assay results within 2–4 working days, and amplicon sequencing (Deeplex) and whole-genome sequencing results within 7–10 working days.
23 (92%) agree, one (4%) disagree, and one (4%) abstain.
-
How should the results of molecular DST be reported by the laboratory to the clinicians?
Results of molecular DST should be reported on standardised forms by the laboratory to the clinicians, including interpretation on the confidence of drug resistance prediction and on the level of drug resistance, when agreed-upon definitions for high-level or low-level resistance are available.
25 (100%) agree.
-
After starting a patient on treatment for tuberculosis based on molecular results, should the treatment regimen be changed if phenotypic DST provides a discrepant result?
In case of discrepant results between the genotypic prediction and phenotypic testing of M tuberculosis drug resistance, results should be discussed between the clinician and microbiologist to identify the cause of the discrepancy and the relevance for clinical decision making. Low-level drug resistance that is not identified by routine phenotypic DST might be identified by genotypic testing. At the same time, genotypic analysis might identify mutations that confer drug resistance, which leads to a low level of phenotypic drug resistance that could potentially be overcome by high dosages of medicines in clinical practice. By contrast, phenotypic DST might identify drug resistance caused by other, yet unknown, mechanisms not detected by genotypic analysis.
25 (100%) agree.
Conclusions
Molecular prediction of M tuberculosis drug susceptibility and resistance is currently revolutionising the management of patients with tuberculosis, especially in settings with a high burden of MDR or rifampicin-resistant tuberculosis. For the first time, in 2021, WHO issued a catalogue of mutations and their associations with antimicrobial resistance, to individualise and mirror the path taken almost two decades ago in the field of HIV infection. New technologies allow for targeted sequencing directly from sputum90 or stool155 specimens, providing the prospect of a much faster turnaround time for DST than for conventional bacteriological methods, but still allowing for the design of accurate MDR or rifampicin-resistant tuberculosis treatment regimens.31 As new drugs are now being marketed, it is essential to ensure that M tuberculosis does not acquire resistance against these new compounds shortly after they become available. In tandem with the roll-out of reliable diagnostic technologies that meet WHO target product profiles156 to comprehensively predict drug susceptibility and resistance, global availability of antimicrobial susceptibility testing157 is equally important. Ideally, severe mutations in the M tuberculosis genome should already be known before marketing newer drugs, thus enabling the identification of emerging drug resistance early on. The tools are becoming available—now is the time to bring them to the places where they are needed most.
Acknowledgments
The study was funded by the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement (823854; INNOVA4TB) and from COST (European Cooperation in Science and Technology) action (CA21164; ADVANCE-TB).
Footnotes
For more on TBnet see http://www.tbnet.eu
For more on RESIST-TB see http://www.resisttb.org
Declaration of interests
JD reports a technology licence to GenID (Germany), and honoraria for lectures from Oxford Immunotec (UK). EC reports support for attendance, accommodation, and travel for ECCMID 2022, Lisboa from European Society of Clinical Microbiology and Infectious Diseases (ESCMID), for annual ERL-TB net meetings from ERL-TB net (Network of National Reference Centers for Tuberculosis in Europe), and for the annual congress 2022, Bologna, from the European Society for Mycobacteriology; is a member of the Executive committee of ESCMID; and is chair of the subcommittee for antimycobacterial agents of EUCAST (European Committee on Antimicrobial Susceptibility Testing). MRF reports grants or contracts from NIH/NIAID (5R01AI155765 and 5R21AI154089) and consulting fees (paid to them) from FIND. LG is a member (unpaid) of the data safety monitoring board for the XACT-19 clinical trial in University of Cape Town, Cape Town, South Africa, and is co-principal investigator of two phase 3 clinical trials on shorter treatment for MDR-TB (endTB and endTB-Q), funded by Unitaid. BL reports grants or contracts from European Union, German Ministry for Education and Research (BMBF), Kultusministerkonferenz, German Centre for Infection Research, and Helmholtz Association, and unpaid leadership or fiduciary roles for DZIF IAB, DZIF Steering Committee transplant Cohort, and TBnet chair Epidemiology. SN reports support for this manuscript (eg, funding, provision of study materials, medical writing, and article processing charges) from BMBF (German Center for Infection Research), DFG (Excellenz Cluster Precision Medicine in Chronic Inflammation EXC 2167), and Leibniz Science Campus Evolutionary Medicine of the LUNG (EvoLUNG), and consulting fees from Illumina advisory board in 2022. NV reports grants or contracts for a study on bedaquiline from Janssen. CRH reports grants or contracts from NIH/NIAID (R01AI134430, DAA3-19-65672, R01AI147316 U01AI152980, and R01AI146555), and Centers for Disease Control and Prevention (NU38PS004651); consulting fees from Otsuka Pharmaceuticals; participation on a data safety monitoring board for SODUCU (PanACEA Sutezolid Dose-finding and Combination Evaluation), BEAT-Tuberculosis (Building Evidence for Advancing New treatment for tuberculosis), DECODE (PanACEA Delpazolid Dose-finding and Combination Development), and Médecins Sans Frontières; and a leadership or fiduciary role from the International Union Against Tuberculosis and Lung Diseases. CLa reports support for the present manuscript (eg, funding, provision of study materials, medical writing, and article processing charges) from DZIF (German Center of Infection Research); consulting fees from a consultation service to INSMED, a company that produced liposomal amikacin as an inhalative suspension for the treatment of non-tuberculous mycobacteria pulmonary disease (outside of the scope of this work); speakers’ honoraria from Insmed, Gilead, and Janssen (all outside of the scope of this work); is a member of the data safety board of trials from Médecins Sans Frontières (outside of the scope of this work); is supported by the German Center for Infection Research (DZIF); and acknowledges funding from the European Commission (anTBiotic EU-H2020 733079, ClicTB EDCTP2 RIA2017T-2030, stool4TB EDCTP2 RIAD2018-2511, and UNITE4TB EU-IMI 101007873). All other authors declare no competing interests.
Contributor Information
José Domínguez, Institut d’Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, CIBER Enfermedades Respiratorias, INNOVA4TB Consortium, Barcelona, Spain.
Martin J Boeree, Department of Lung Diseases, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands.
Emmanuelle Cambau, Centre National de Référence des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, Paris, France, APHP-Hôpital Bichat, Mycobacteriology Laboratory, INSERM, University Paris Cite, IAME UMR1137, Paris, France.
Dumitru Chesov, Department of Pneumology and Allergology, Nicolae Testemiţanu State University of Medicine and Pharmacy, Chisinau, Moldova, Division of Clinical Infectious Diseases, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany, Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany.
Francesca Conradie, Department of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa.
Vivian Cox, Centre for Infectious Disease Epidemiology and Research, School of Public Health and Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
Keertan Dheda, Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa; Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, UK.
Andrii Dudnyk, Department of Tuberculosis, Clinical Immunology and Allergy, National Pirogov Memorial Medical University, Vinnytsia, Ukraine, Public Health Center, Ministry of Health of Ukraine, Kyiv, Ukraine.
Maha R Farhat, Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA.
Sebastien Gagneux, Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health lnstitute, Allschwil, Switzerland, University of Basel, Basel, Switzerland.
Martin P Grobusch, Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Amsterdam, Netherlands.
Matthias I Gröschel, Department of Infectious Diseases and Respiratory Medicine, Charité—Universitätsmedizin Berlin, Berlin, Germany, Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
Lorenzo Guglielmetti, Sorbonne Université, INSERM, U1135, Centre d’Immunologie et des Maladies Infectieuses, (Cimi-Paris), APHP Sorbonne Université, Department of Bacteriology Hôpital Pitié-Salpêtrière, Centre National de Référence des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, Paris, France.
Irina Kontsevaya, Division of Clinical Infectious Diseases, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany, Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK.
Berit Lange, Department for Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany, German Centre for Infection Research, TI BBD, Braunschweig, Germany.
Frank van Leth, Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands, Amsterdam Public Health Research Institute, Amsterdam, Netherlands.
Christian Lienhardt, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK, UMI 233 IRD–U1175 INSERM - Université de Montpellier, Institut de Recherche pour le Développement, Montpellier, France.
Anna M Mandalakas, Division of Clinical Infectious Diseases, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany, Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany, Global TB Program, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA.
Florian P Maurer, National and Supranational Reference Center for Mycobacteria, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany, Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
Matthias Merker, Division of Evolution of the Resistome, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany.
Paolo Miotto, Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
Barbara Molina-Moya, Institut d’Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, CIBER Enfermedades Respiratorias, INNOVA4TB Consortium, Barcelona, Spain.
Florence Morel, Sorbonne Université, INSERM, U1135, Centre d’Immunologie et des Maladies Infectieuses, (Cimi-Paris), APHP Sorbonne Université, Department of Bacteriology Hôpital Pitié-Salpêtrière, Centre National de Référence des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, Paris, France.
Stefan Niemann, Division of Molecular and Experimental Mycobacteriology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany, Department of Human, Biological and Translational Medical Sciences, School of Medicine, University of Namibia, Windhoek, Namibia.
Nicolas Veziris, Sorbonne Université, INSERM, U1135, Centre d’Immunologie et des Maladies Infectieuses, (Cimi-Paris), APHP Sorbonne Université, Department of Bacteriology Hôpital Pitié-Salpêtrière, Centre National de Référence des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, Paris, France.
Andrew Whitelaw, Division of Medical Microbiology, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa, National Health Laboratory Service, Tygerberg Hospital, Cape Town, South Africa.
Charles R Horsburgh, Jr, Departments of Epidemiology, Biostatistics, Global Health and Medicine, Boston University Schools of Public Health and Medicine, Boston, MA, USA.
Christoph Lange, Division of Clinical Infectious Diseases, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany, Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany, Global TB Program, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA.
References
- 1.WHO. Global tuberculosis report 2022. Geneva: World Health Organization, 2022. [Google Scholar]
- 2.WHO. Report of the meeting of the WHO Global Task Force on XDR-TB: Geneva, Switzerland, 9–10 October 2006. Geneva: World Health Organization, 2006. [Google Scholar]
- 3.WHO. Meeting report of the WHO expert consultation on the definition of extensively drug-resistant tuberculosis, 27–29 October 2020. Geneva: World Health Organization, 2021. [Google Scholar]
- 4.Allix-Béguec C, Arandjelovic I, Bi L, et al. Prediction of susceptibility to first-line tuberculosis drugs by DNA sequencing. N Engl J Med 2018; 379:1403–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Molina-Moya B, Guglielmetti L, Bothamley G, van Leth F, Lange C, Dominguez J. Use and impact of molecular methods for detecting drug-resistant TB. Int J Tuberc Lung Dis 2021; 25:157–59. [DOI] [PubMed] [Google Scholar]
- 6.Dominguez J, Boettger EC, Cirillo D, et al. Clinical implications of molecular drug resistance testing for Mycobacterium tuberculosis: a TBNET/RESIST-TB consensus statement. Int J Tuberc Lung Dis 2016; 20: 24–42. [DOI] [PubMed] [Google Scholar]
- 7.Mack U, Migliori GB, Sester M, et al. LTBI: latent tuberculosis infection or lasting immune responses to M tuberculosis? A TBNET consensus statement. Eur Respir J 2009; 33: 956–73. [DOI] [PubMed] [Google Scholar]
- 8.Solovic I, Sester M, Gomez-Reino JJ, et al. The risk of tuberculosis related to tumour necrosis factor antagonist therapies: a TBNET consensus statement. Eur Respir J 2010; 36:1185–206. [DOI] [PubMed] [Google Scholar]
- 9.Bumbacea D, Arend SM, Eyuboglu F, et al. The risk of tuberculosis in transplant candidates and recipients: a TBNET consensus statement. Eur Respir J 2012; 40: 990–1013. [DOI] [PubMed] [Google Scholar]
- 10.Lange C, Abubakar I, Alffenaar JW, et al. Management of patients with multidrug-resistant/extensively drug-resistant tuberculosis in Europe: a TBNET consensus statement. Eur Respir J 2014; 44: 23–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chesov E, Chesov D, Maurer FP, et al. Emergence of bedaquiline resistance in a high tuberculosis burden country. Eur Respir J 2022; 59: 2100621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gygli SM, Borrell S, Trauner A, Gagneux S. Antimicrobial resistance in Mycobacterium tuberculosis: mechanistic and evolutionary perspectives. FEMS Microbiol Rev 2017; 41: 354–73. [DOI] [PubMed] [Google Scholar]
- 13.Nguyen TVA, Anthony RM, Bañuls AL, Nguyen TVA, Vu DH, Alffenaar JC. Bedaquiline resistance: its emergence, mechanism, and prevention. Clin Infect Dis 2018; 66:1625–30. [DOI] [PubMed] [Google Scholar]
- 14.Shimao T Drug resistance in tuberculosis control. Tubercle 1987; 68 (suppl): 5–18. [DOI] [PubMed] [Google Scholar]
- 15.Gagneux S, DeRiemer K, Van T, et al. Variable host-pathogen compatibility in Mycobacterium tuberculosis. Proc Natl Acad Sci USA 2006; 103: 2869–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ford CB, Shah RR, Maeda MK, et al. Mycobacterium tuberculosis mutation rate estimates from different lineages predict substantial differences in the emergence of drug-resistant tuberculosis. Nat Genet 2013;45:784–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Castro RAD, Ross A, Kamwela L, et al. The genetic background modulates the evolution of fluoroquinolone-resistance in Mycobacterium tuberculosis. Mol Biol Evol 2020; 37:195–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Merker M, Kohl TA, Roetzer A, et al. Whole genome sequencing reveals complex evolution patterns of multidrug-resistant Mycobacterium tuberculosis Beijing strains in patients. PLoS One 2013; 8: e82551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bloemberg GV, Keller PM, Stucki D, et al. Acquired resistance to bedaquiline and delamanid in therapy for tuberculosis. N Engl J Med 2015; 373:1986–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ley SD, de Vos M, Van Rie A, Warren RM. Deciphering within-host microevolution of Mycobacterium tuberculosis through whole-genome sequencing: the phenotypic impact and way forward. Microbiol Mol Biol Rev 2019; 83: e00062–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Sonnenkalb L, Strohe G, Dreyer V, et al. Microevolution of Mycobacterium tuberculosis subpopulations and heteroresistance in a patient receiving 27 years of tuberculosis treatment in Germany. Antimicrob Agents Chemother 2021; 65: e0252020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Müller B, Borrell S, Rose G, Gagneux S. The heterogeneous evolution of multidrug-resistant Mycobacterium tuberculosis. Trends Genet 2013; 29:160–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Colangeli R, Jedrey H, Kim S, et al. Bacterial factors that predict relapse after tuberculosis therapy. N Engl J Med 2018; 379:823–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Jones RM, Adams KN, Eldesouky HE, Sherman DR. The evolving biology of Mycobacterium tuberculosis drug resistance. Front Cell Infect Microbiol 2022; 12:1027394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Dartois V The path of anti-tuberculosis drugs: from blood to lesions to mycobacterial cells. Nat Rev Microbiol 2014; 12:159–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Sarathy JP, Zuccotto F, Hsinpin H, et al. Prediction of drug penetration in tuberculosis lesions. ACS Infect Dis 2016; 2: 552–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Moreno-Molina M, Shubladze N, Khurtsilava I, et al. Genomic analyses of Mycobacterium tuberculosis from human lung resections reveal a high frequency of polyclonal infections. Nat Commun 2021; 12: 2716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Castro RAD, Borrell S, Gagneux S. The within-host evolution of antimicrobial resistance in Mycobacterium tuberculosis. FEMS Microbiol Rev 2021; 45: fuaa071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chen Y, Ji L, Liu Q, et al. Lesion heterogeneity and long-term heteroresistance in multidrug-resistant tuberculosis. J Infect Dis 2021; 224: 889–93. [DOI] [PubMed] [Google Scholar]
- 30.Dheda K, Lenders L, Magombedze G, et al. Drug-penetration gradients associated with acquired drug resistance in patients with tuberculosis. Am J Respir Crit Care Med 2018; 198: 1208–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Grobbel HP, Merker M, Köhler N, et al. Design of multidrug-resistant tuberculosis treatment regimens based on DNA sequencing. Clin Infect Dis 2021; 73: 1194–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cox HS, Sibilia K, Feuerriegel S, et al. Emergence of extensive drug resistance during treatment for multidrug-resistant tuberculosis. N Engl J Med 2008; 359: 2398–400. [DOI] [PubMed] [Google Scholar]
- 33.Bonnet M, Pardini M, Meacci F, et al. Treatment of tuberculosis in a region with high drug resistance: outcomes, drug resistance amplification and re-infection. PLoS One 2011; 6: e23081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zignol M, Dara M, Dean AS, et al. Drug-resistant tuberculosis in the WHO European Region: an analysis of surveillance data. Drug Resist Updat 2013; 16: 108–15. [DOI] [PubMed] [Google Scholar]
- 35.Casali N, Nikolayevskyy V, Balabanova Y, et al. Evolution and transmission of drug-resistant tuberculosis in a Russian population. Nat Genet 2014; 46: 279–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Cohen KA, Abeel T, Manson McGuire A, et al. Evolution of extensively drug-resistant tuberculosis over four decades: whole genome sequencing and dating analysis of Mycobacterium tuberculosis isolates from KwaZulu-Natal. PLoS Med 2015; 12: el001880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Merker M, Barbier M, Cox H, et al. Compensatory evolution drives multidrug-resistant tuberculosis in Central Asia. eLife 2018; 7: e38200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sander P, Springer B, Prammananan T, et al. Fitness cost of chromosomal drug resistance-conferring mutations. Antimicrob Agents Chemother 2002; 46: 1204–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Böttger EC, Springer B. Tuberculosis: drug resistance, fitness, and strategies for global control. Eur J Pediatr 2008; 167: 141–48. [DOI] [PubMed] [Google Scholar]
- 40.Comas I, Borrell S, Roetzer A, et al. Whole-genome sequencing of rifampidn-resistant Mycobacterium tuberculosis strains identifies compensatory mutations in RNA polymerase genes. Nat Genet 2011; 44: 106–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Borrell S, Teo Y, Giardina F, et al. Epistasis between antibiotic resistance mutations drives the evolution of extensively drug-resistant tuberculosis. Evol Med Public Health 2013; 2013: 65–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Green AG, Vargas R, Marin MG, Freschi L, Xie J, Farhat MR. The discovery of genome-wide mutational dependence in naturally evolving populations. bioRxiv 2022; published online June 28. 10.1101/2022.06.24.497504 (preprint). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gygli SM, Loiseau C, Jugheli L, et al. Prisons as ecological drivers of fitness-compensated multidrug-resistant Mycobacterium tuberculosis. Nat Med 2021; 27: 1171–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Inderlied CBNK. Antimycobacterial agents: in vitro susceptibility testing and mechanisms of action and resistance. Chapter 5. In: Lorian V (ed). Antibiotics in laboratory medicine, 5th edn. Philadelphia, PA: Lippincott Williams and Wilkins, 2005:155–225. [Google Scholar]
- 45.Clinical and Laboratory Standards Institute. Susceptibility testing of mycobacteria, nocardiae, and other aerobic actinomycetes, 3rd edition. Wayne, PA: Clinical and Laboratory Standards Institute, 2018. [PubMed] [Google Scholar]
- 46.Cambau E, Rush-Gerdes S. First- and second-line susceptibility testing for Mycobacterium tuberculosis complex. In: European Centre for Disease Prevention and Control. Handbook on TB laboratory diagnostic methods for the European Union. Stockholm: European Centre for Disease Prevention and Control, 2018: 81–91. [Google Scholar]
- 47.Van Deun A, Wright A, Zignol M, Weyer K, Rieder HL. Drug susceptibility testing proficiency in the network of supranational tuberculosis reference laboratories. Int J Tuberc Lung Dis 2011; 15: 116–24. [PubMed] [Google Scholar]
- 48.Antimycobacterial Susceptibility Testing Group. Updating the approaches to define susceptibility and resistance to anti-tuberculosis agents: implications for diagnosis and treatment. Eur Respir J 2022; 59: 2200166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Van Deun A, Aung KJ, Bola V, et al. Rifampin drug resistance tests for tuberculosis: challenging the gold standard. J Clin Microbiol 2013; 51: 2633–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zuur MA, Pasipanodya JG, van Soolingen D, van der Werf TS, Gumbo T, Alffenaar JC. Intermediate susceptibility dose-dependent breakpoints for high-dose rifampin, isoniazid, and pyrazinamide treatment in multidrug-resistant tuberculosis programs. Clin Infect Dis 2018; 67: 1743–9. [DOI] [PubMed] [Google Scholar]
- 51.WHO. Technical report on critical concentrations for drug susceptibility testing of medicines used in the treatment of drug-resistant tuberculosis. Geneva: World Health Organization, 2018. [Google Scholar]
- 52.WHO. Technical report on critical concentrations for drug susceptibility testing of isoniazid and the rifamydns (rifampidn, rifabutin and rifapentine). Geneva: World Health Organization, 2021. [Google Scholar]
- 53.Torrea G, Coeck N, Desmaretz C, et al. Bedaquiline susceptibility testing of Mycobacterium tuberculosis in an automated liquid culture system. J Antimicrob Chemother 2015; 70: 2300–05. [DOI] [PubMed] [Google Scholar]
- 54.Kaniga K, Hasan R, Jou R, et al. Bedaquiline drug resistance emergence assessment in multidrug-resistant tuberculosis (MDR-TB): a 5-year prospective in vitro surveillance study of bedaquiline and other second-line drug susceptibility testing in MDR-TB isolates. J Clin Microbiol 2022; 60: e0291920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Bateson A, Ortiz Canseco J, McHugh TD, et al. Ancient and recent differences in the intrinsic susceptibility of Mycobacterium tuberculosis complex to pretomanid. J Antimicrob Chemother 2022; 77: 1685–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Böttger EC. The ins and outs of Mycobacterium tuberculosis drug susceptibility testing. Clin Microbiol Infect 2011; 17: 1128–34. [DOI] [PubMed] [Google Scholar]
- 57.Cambau E, Viveiros M, Machado D, et al. Revisiting susceptibility testing in MDR-TB by a standardized quantitative phenotypic assessment in a European multicentre study. J Antimicrob Chemother 2015; 70: 686–96. [DOI] [PubMed] [Google Scholar]
- 58.Schön T, Werngren J, Machado D, et al. Antimicrobial susceptibility testing of Mycobacterium tuberculosis complex isolates—the EUCAST broth microdilution reference method for MIC determination. Clin Microbiol Infect 2020; 26: 1488–92. [DOI] [PubMed] [Google Scholar]
- 59.European Committee on Antimicrobial Susceptibility Testing. Methods in AST of Mycobacterium species https://www.eucast.org/mycobacteria/methods_in_mycobacteria/ (accessed June 1, 2022). [Google Scholar]
- 60.Schön T, Köser CU, Werngren J, et al. What is the role of the EUCAST reference method for MIC testing of the Mycobacterium tuberculosis complex? Clin Microbiol Infect 2020; 26: 1453–55. [DOI] [PubMed] [Google Scholar]
- 61.Consortium CRyPTIC. Epidemiological cutoff values for a 96-well broth microdilution plate for high-throughput research antibiotic susceptibility testing of M tuberculosis. Eur Respir J 2022; 60: 2200239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.WHO. Optimized broth microdilution plate methodology for drug susceptibility testing of Mycobacterium tuberculosis complex. Geneva: World Health Organization, 2022. [Google Scholar]
- 63.Meehan CJ, Goig GA, Kohl TA, et al. Whole genome sequencing of Mycobacterium tuberculosis: current standards and open issues. Nat Rev Microbiol 2019; 17: 533–45. [DOI] [PubMed] [Google Scholar]
- 64.Ando H, Miyoshi-Akiyama T, Watanabe S, Kirikae T. A silent mutation in mabA confers isoniazid resistance on Mycobacterium tuberculosis. Mol Microbiol 2014; 91: 538–47. [DOI] [PubMed] [Google Scholar]
- 65.Feuerriegel S, Köser CU, Niemann S. Phylogenetic polymorphisms in antibiotic resistance genes of the Mycobacterium tuberculosis complex. J Antimicrob Chemother 2014; 69: 1205–10. [DOI] [PubMed] [Google Scholar]
- 66.Ajileye A, Alvarez N, Merker M, et al. Some synonymous and nonsynonymous gyrA mutations in Mycobacterium tuberculosis lead to systematic false-positive fluoroquinolone resistance results with the HAIN GenoType MTBD Rsl assays. Antimicrob Agents Chemother 2017; 61: e02169–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Miotto P, Tessema B, Tagliani E, et al. A standardised method for interpreting the association between mutations and phenotypic drug resistance in Mycobacterium tuberculosis. Eur Respir J 2017; 50: 1701354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Merker M, Kohl TA, Barilar I, et al. Phylogenetically informative mutations in genes implicated in antibiotic resistance in Mycobacterium tuberculosis complex. Genome Med 2020; 12: 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Köser CU, Cirillo DM, Miotto P. How to optimally combine genotypic and phenotypic drug susceptibility testing methods for pyrazinamide. Antimicrob Agents Chemother 2020; 64: e01003–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Beckert P, Sanchez-Padilla E, Merker M, et al. MDR M tuberculosis outbreak clone in Eswatini missed by Xpert has elevated bedaquiline resistance dated to the pre-treatment era. Genome Med 2020; 12: 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ektefaie Y, Dixit A, Freschi L, Farhat MR. Globally diverse Mycobacterium tuberculosis resistance acquisition: a retrospective geographical and temporal analysis of whole genome sequences. Lancet Microbe 2021; 2: e96–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Walker TM, Merker M, Knoblauch AM, et al. A cluster of multidrug-resistant Mycobacterium tuberculosis among patients arriving in Europe from the Horn of Africa: a molecular epidemiological study. Lancet Infect Dis 2018; 18: 431–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Briquet A, Vong R, Roseau JB, et al. Clinical features of Mycobacterium canettii infection: a retrospective study of 20 cases among French soldiers and relatives. Clin Infect Dis 2019; 69: 2003–10. [DOI] [PubMed] [Google Scholar]
- 74.WHO. WHO consolidated guidelines on drug-resistant tuberculosis treatment. Geneva: World Health Organization, 2019. [PubMed] [Google Scholar]
- 75.Miotto P, Piana E, Cirillo DM, Migliori GB. Genotype MTBDRplus: a further step toward rapid identification of drug-resistant Mycobacterium tuberculosis. J Clin Microbiol 2008; 46: 393–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Boehme CC, Nabeta P, Hillemann D, et al. Rapid molecular detection of tuberculosis and rifampin resistance. N Engl. J Med 2010; 363: 1005–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Tagliani E, Cabibbe AM, Miotto P, et al. diagnostic performance of the new version (v2.0) of GenoType MTBDRsl assay for detection of resistance to fluoroquinolones and second-line injectable drugs: a multicenter study. J Clin Microbiol 2015; 53: 2961–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Nathavitharana RR, Hillemann D, Schumacher SG, et al. Multicenter noninferiority evaluation of HAIN GenoType MTBDRplus version 2 and Nipro NTM+MDRTB line probe assays for detection of rifampin and isoniazid resistance. J Clin Microbiol 2016; 54:1624–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Chakravorty S, Simmons AM, Rowneki M, et al. The new Xpert MTB/RIF Ultra: improving detection of Mycobacterium tuberculosis and resistance to rifampin in an assay suitable for point-of-care testing. mBio 2017; 8: e00812–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Georghiou SB, Gomathi NS, Rajendran P, et al. Accuracy of the Truenat MTB-RIF Dx assay for detection of rifampicin resistance-associated mutations. Tuberculosis (Edinb) 2021; 127: 102064. [DOI] [PubMed] [Google Scholar]
- 81.WHO. WHO operational handbook on tuberculosis. Module 3: diagnosis—rapid diagnostics for tuberculosis detection. Geneva: World Health Organization, 2020. [PubMed] [Google Scholar]
- 82.WHO. WHO consolidated guidelines on tuberculosis. Module 3: diagnosis—rapid diagnostics for tuberculosis detection, 2021 update. Geneva: World Health Organization, 2021 [PubMed] [Google Scholar]
- 83.Cao Y, Parmar H, Gaur RL, et al. Xpert MTB/XDR: a 10-color reflex assay suitable for point-of-care settings to detect isoniazid, fluoroquinolone, and second-line-injectable-drug resistance directly from Mycobacterium tuberculosis-positive sputum. J Clin Microbiol 2021; 59: 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Driesen M, Kondo Y, de Jong BC, et al. Evaluation of a novel line probe assay to detect resistance to pyrazinamide, a key drug used for tuberculosis treatment. Clin Microbiol Infect 2018; 24: 60–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Willby MJ, Wijkander M, Havumaki J, et al. Detection of Mycobacterium tuberculosis pncA mutations by the Nipro Genoscholar PZA-TB II assay compared to conventional sequencing. Antimicrob Agents Chemother 2017; 62: e01871–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kadura S, King N, Nakhoul M, et al. Systematic review of mutations associated with resistance to the new and repurposed Mycobacterium tuberculosis drugs bedaquiline, clofazimine, linezolid, delamanid and pretomanid. J Antimicrob Chemother 2020; 75: 2031–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Rifat D, Li SY, Ioerger T, et al. Mutations in fbiD (Rv2983) as a novel determinant of resistance to pretomanid and delamanid in Mycobacterium tuberculosis. Antimicrob Agents Chemother 2020; 65: e01948–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Farooq HZ, Cirillo DM, Hillemann D, et al. Limited capability for testing Mycobacterium tuberculosis for susceptibility to new drugs. Emerg Infect Dis 2021; 27: 985–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Feuerriegel S, Kohl TA, Utpatel C, et al. Rapid genomic first- and second-line drug resistance prediction from clinical Mycobacterium tuberculosis specimens using Deeplex-MycTB. Eur Respir J 2021; 57: 2001796. [DOI] [PubMed] [Google Scholar]
- 90.Jouet A, Gaudin C, Badalato N, et al. Deep amplicon sequencing for culture-free prediction of susceptibility or resistance to 13 anti-tuberculous drugs. Eur Respir J 2021; 57: 2002338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Cabibbe AM, Spitaleri A, Battaglia S, et al. Application of targeted next-generation sequencing assay on a portable sequencing platform for culture-free detection of drug-resistant tuberculosis from clinical samples. J Clin Microbiol 2020; 58: e00632–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Colman RE, Anderson J, Lemmer D, et al. Rapid drug susceptibility testing of drug-resistant Mycobacterium tuberculosis isolates directly from clinical samples by use of amplicon sequencing: a proof-of-concept study. J Clin Microbiol 2016; 54:2058–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Park J, Shin SY, Kim K, Park K, Shin S, Ihm C. Determining genotypic drug resistance by ion semiconductor sequencing with the Ion AmpliSeq™ TB panel in multidrug-resistant Mycobacterium tuberculosis isolates. Ann Lab Med 2018; 38: 316–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Heyckendorf J, Andres S, Köser CU, et al. What is resistance? Impact of phenotypic versus molecular drug resistance testing on therapy for multi- and extensively drug-resistant tuberculosis. Antimicrob Agents Chemother 2018; 62: e01550–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Gröschel MI, Walker TM, van der Werf TS, Lange C, Niemann S, Merker M. Pathogen-based precision medicine for drug-resistant tuberculosis. PLoS Pathog 2018; 14: el007297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Colman RE, Mace A, Seifert M, et al. Whole-genome and targeted sequencing of drug-resistant Mycobacterium tuberculosis on the iSeql00 and MiSeq: a performance, ease-of-use, and cost evaluation. PLoS Med 2019; 16: el002794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Friedrich SO, Rachow A, Saathoff E, et al. Assessment of the sensitivity and specificity of Xpert MTB/RIF assay as an early sputum biomarker of response to tuberculosis treatment. Lancet Respir Med 2013; 1: 462–70. [DOI] [PubMed] [Google Scholar]
- 98.Rigouts L, Miotto P, Schats M, et al. Fluoroquinolone heteroresistance in Mycobacterium tuberculosis: detection by genotypic and phenotypic assays in experimentally mixed populations. Sci Rep 2019; 9: 11760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ng KCS, Supply P, Cobelens FGJ, et al. How well do routine molecular diagnostics detect rifampin heteroresistance in Mycobacterium tuberculosis? J Clin Microbiol 2019; 57: e00717–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Metcalfe JZ, Streicher E, Theron G, et al. Cryptic microheteroresistance explains Mycobacterium tuberculosis phenotypic resistance. Am J Respir Crit Care Med 2017; 196: 1191–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.de Vos M, Ley SD, Wiggins KB, et al. Bedaquiline microheteroresistance after cessation of tuberculosis treatment. N Engl, J Med 2019; 380: 2178–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Pankhurst LJ, Del Ojo Elias C, Votintseva AA, et al. Rapid, comprehensive, and affordable mycobacterial diagnosis with whole-genome sequencing: a prospective study. Lancet Respir Med 2016; 4: 49–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Mugwagwa T, Abubakar I, White PJ. Using molecular testing and whole-genome sequencing for tuberculosis diagnosis in a low-burden setting: a cost-effectiveness analysis using transmission-dynamic modelling. Thorax 2021; 76: 281–91. [DOI] [PubMed] [Google Scholar]
- 104.Ramachandran R, Muniyandi M. Rapid molecular diagnostics for multi-drug resistant tuberculosis in India. Expert Rev Anti Infect Ther 2018; 16:197–204. [DOI] [PubMed] [Google Scholar]
- 105.Cates L, Codreanu A, Ciobanu N, et al. Budget impact of next-generation sequencing for diagnosis of TB drug resistance in Moldova. Int J Tuberc Lung Dis 2022; 26: 963–69. [DOI] [PubMed] [Google Scholar]
- 106.Gunther G, Guglielmetti L, Leu C, Lange C, van Leth F. Availability and costs of medicines for the treatment of tuberculosis in Europe. Clin Microbiol Infect 2022; 29:77–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Canetti G, Rist N, Grosset J. Measurement of sensitivity of the tuberculous bacillus to antibacillary drugs by the method of proportions. Methodology, resistance criteria, results and interpretation. Rev Tuberc Pneumol (Paris) 1963; 27: 217–72. [PubMed] [Google Scholar]
- 108.Telenti A, Imboden P, Marchesi F, et al. Detection of rifampicin-resistance mutations in Mycobacterium tuberculosis. Lancet 1993; 341: 647–50. [DOI] [PubMed] [Google Scholar]
- 109.Ismail N, Omar SV, Ismail NA, Peters RPH. Collated data of mutation frequencies and associated genetic variants of bedaquiline, clofazimine and linezolid resistance in Mycobacterium tuberculosis. Data Brief 2018; 20:1975–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.WHO. The use of next-generation sequencing technologies for the detection of mutations associated with drug resistance in Mycobacterium tuberculosis complex: technical guide. Geneva: World Health Organization, 2018. [Google Scholar]
- 111.WHO. Catalogue of mutations in Mycobacterium tuberculosis complex and their association with drug resistance. Geneva: World Health Organization, 2021. [Google Scholar]
- 112.Van Deun A, Barrera L, Bastian I, et al. Mycobacterium tuberculosis strains with highly discordant rifampin susceptibility test results. J Gin Microbiol 2009; 47: 3501–06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Gurbanova E, Mehdiyev R, Blondal K, et al. Mitigation of discordant rifampidn-susceptibiHty results obtained by Xpert Mycobacterium tuberculosis/rifampicin and mycobacterium growth indicator tube. Microb Drug Resist 2017; 23: 1045–52. [DOI] [PubMed] [Google Scholar]
- 114.Hu P, Zhang H, Fleming J, et al. Retrospective analysis of false-positive and disputed rifampin resistance Xpert MTB/RIF assay results in clinical samples from a referral hospital in Hunan, China. J Gin Microbiol 2019; 57: e01707–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Abate D, Tedla Y, Meressa D, Ameni G. Isoniazid and rifampicin resistance mutations and their effect on second-line anti-tuberculosis treatment. Int J Tuberc Lung Dis 2014; 18: 946–51. [DOI] [PubMed] [Google Scholar]
- 116.Tolani MP, D’souza DT, Mistry NF. Drug resistance mutations and heteroresistance detected using the GenoType MTBDRplus assay and their implication for treatment outcomes in patients from Mumbai, India. BMC Infect Dis 2012; 12: 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Huyen MN, Cobelens FG, Buu TN, et al. Epidemiology of isoniazid resistance mutations and their effect on tuberculosis treatment outcomes. Antimicrob Agents Chemother 2013; 57: 3620–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Dooley KE, Miyahara S, von Groote-Bidlingmaier F, et al. Early bactericidal activity of different isoniazid doses for drug resistant TB (INHindsight): a randomized open-label clinical trial. Am J Respir Crit Care Med 2020; 201: 1416–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.He G, Li Y, Chen X, Chen J, Zhang W. Prediction of treatment outcomes for multidrug-resistant tuberculosis by whole-genome sequencing. Int J Infect Dis 2020; 96: 68–72. [DOI] [PubMed] [Google Scholar]
- 120.Lempens P, Decroo T, Aung KJM, et al. Initial resistance to companion drugs should not be considered an exclusion criterion for the shorter multidrug-resistant tuberculosis treatment regimen. Int J Infect Dis 2020; 100: 357–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Ismail NA, Omar SV, Joseph L, et al. Defining bedaquiline susceptibility, resistance, cross-resistance and associated genetic determinants: a retrospective cohort study. EBioMedicine 2018; 28: 136–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Liu Y, Gao M, Du J, et al. Reduced susceptibility of Mycobacterium tuberculosis to bedaquiline during antituberculosis treatment and its correlation with clinical outcomes in China. Clin Infect Dis 2021; 73: e3391–97 [DOI] [PubMed] [Google Scholar]
- 123.Ismail NA, Omar SV, Moultrie H, et al. Assessment of epidemiological and genetic characteristics and clinical outcomes of resistance to bedaquiline in patients treated for rifampicin-resistant tuberculosis: a cross-sectional and longitudinal study. Lancet Infect Dis 2022; 22: 496–506. [DOI] [PubMed] [Google Scholar]
- 124.Ghodousi A, Hussain Rizvi A, Khanzada FM, et al. In vivo microevolution of Mycobacterium tuberculosis and transient emergence of atpE_Ala63Pro mutation during treatment in a pre-XDR TB patient. Eur Respir J 2022; 59: 2102102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Le Ray LF, Aubry A, Sougakoff W, et al. atpE mutation in Mycobacterium tuberculosis not always predictive of bedaquiline treatment failure. Emerg Infect Dis 2022; 28: 1062–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Leung KL, Yip CW, Yeung YL, et al. Usefulness of resistant gene markers for predicting treatment outcome on second-line anti-tuberculosis drugs. J Appl Microbiol 2010; 109: 2087–94. [DOI] [PubMed] [Google Scholar]
- 127.Rigouts L, Coeck N, Gumusboga M, et al. Specific gyrA gene mutations predict poor treatment outcome in MDR-TB. J Antimicrob Chemother 2016; 71: 314–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Chien JY, Chien ST, Chiu WY, Yu CJ, Hsueh PR. Moxifloxacin improves treatment outcomes in patients with ofloxacin-resistant multidrug-resistant tuberculosis. Antimicrob Agents Chemother 2016; 60: 4708–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Zheng X, Ning Z, Drobniewski F, et al. pncA mutations are associated with slower sputum conversion during standard treatment of multidrug-resistant tuberculosis. Int J Antimicrob Agents 2017; 49: 183–88. [DOI] [PubMed] [Google Scholar]
- 130.Farhat MR, Jacobson KR, Franke MF, Kaur D, Murray M, Mitnick CD. Fluoroquinolone resistance mutation detection is equivalent to culture-based drug sensitivity testing for predicting multidrug-resistant tuberculosis treatment outcome: a retrospective cohort study. Clin Infect Dis 2017; 65: 1364–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Maitre T, Petitjean G, Chauffour A, et al. Are moxifloxacin and levofloxacin equally effective to treat XDR tuberculosis? J Antimicrob Chemother 2017; 72: 2326–33. [DOI] [PubMed] [Google Scholar]
- 132.Lee M, Lee J, Carroll MW, et al. Linezolid for treatment of chronic extensively drug-resistant tuberculosis. N Engl J Med 2012; 367: 1508–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Wasserman S, Louw G, Ramangoaela L, et al. Linezolid resistance in patients with drug-resistant TB and treatment failure in South Africa. J Antimicrob Chemother 2019; 74: 2377–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Via LE, Cho SN, Hwang S, et al. Polymorphisms associated with resistance and cross-resistance to aminoglycosides and capreomycin in Mycobacterium tuberculosis isolates from South Korean patients with drug-resistant tuberculosis. J Clin Microbiol 2010; 48: 402–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kuhlin J, Davies Forsman L, Mansjö M, et al. Genotypic resistance of pyrazinamide but not minimum inhibitory concentration is associated with longer time to sputum culture conversion in patients with multidrug-resistant tuberculosis. Clin Infect Dis 2021; 73: e3511–17. [DOI] [PubMed] [Google Scholar]
- 136.Ahmad N, Ahuja SD, Akkerman OW, et al. Treatment correlates of successful outcomes in pulmonary multidrug-resistant tuberculosis: an individual patient data meta-analysis. Lancet 2018; 392: 821–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Fregonese F, Ahuja SD, Akkerman OW, et al. Comparison of different treatments for isoniazid-resistant tuberculosis: an individual patient data meta-analysis. Lancet Respir Med 2018; 6: 265–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.WHO. WHO consolidated guidelines on tuberculosis. Module 4: treatment—drug-resistant tuberculosis treatment. Geneva: World Health Organization, 2020. [PubMed] [Google Scholar]
- 139.Katiyar SK, Bihari S, Prakash S, Mamtani M, Kulkarni H. A randomised controlled trial of high-dose isoniazid adjuvant therapy for multidrug-resistant tuberculosis. Int J Tuberc Lung Dis 2008; 12: 139–15. [PubMed] [Google Scholar]
- 140.Tack I, Dumicho A, Ohler L, et al. Safety and effectiveness of an all-oral, bedaquiline-based, shorter treatment regimen for rifampicin-resistant tuberculosis in high human immunodeficiency virus (HIV) burden rural South Africa: a retrospective cohort analysis. Clin Infect Dis 2021; 73: e3563–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Ndjeka N, Campbell JR, Meintjes G, et al. Treatment outcomes 24 months after initiating short, all-oral bedaquiline-containing or injectable-containing rifampicin-resistant tuberculosis treatment regimens in South Africa: a retrospective cohort study. Lancet Infect Dis 2022; 22:1042–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.WHO. Rapid communication: key changes to the treatment of drug-resistant tuberculosis 2022. Geneva: World Health Organization, 2022. [Google Scholar]
- 143.Lange C, Aarnoutse RE, Alffenaar JWC, et al. Management of patients with multidrug-resistant tuberculosis. Int J Tuberc Lung Dis 2019; 23: 645–62. [DOI] [PubMed] [Google Scholar]
- 144.Lange C, Dheda K, Chesov D, Mandalakas AM, Udwadia Z, Horsburgh CR Jr. Management of drug-resistant tuberculosis. Lancet 2019; 394: 953–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Conradie F, Bagdasaryan TR, Borisov S, et al. Bedaquiline–pretomanid–linezolid regimens for drug-resistant tuberculosis. N Engl J Med 2022; 387: 810–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Roelens M, Battista Migliori G, Rozanova L, et al. Evidence-based definition for extensively drug-resistant tuberculosis. Am J Respir Crit Care Med 2021; 204: 713–22. [DOI] [PubMed] [Google Scholar]
- 147.Schnippel K, Ndjeka N, Maartens G, et al. Effect of bedaquiline on mortality in South African patients with drug-resistant tuberculosis: a retrospective cohort study. Lancet Respir Med 2018; 6: 699–706. [DOI] [PubMed] [Google Scholar]
- 148.Ismail N, Rivière E, Limberis J, et al. Genetic variants and their association with phenotypic resistance to bedaquiline in Mycobacterium tuberculosis: a systematic review and individual isolate data analysis. Lancet Microbe 2021; 2: e604–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Trisakul K, Nonghanphithak D, Chaiyachat P, et al. High clustering rate and genotypic drug-susceptibility screening for the newly recommended anti-tuberculosis drugs among global extensively drug-resistant Mycobacterium tuberculosis isolates. Emerg Microbes Infect 2022; 11: 1857–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.WHO. WHO position statement on the use of delamanid for MDR-TB. Geneva: World Health Organization, 2018. [Google Scholar]
- 151.Khoshnood S, Taki E, Sadeghifard N, et al. Mechanism of action, resistance, synergism, and clinical implications of delamanid against multidrug-resistant Mycobacterium tuberculosis. Front Microbiol 2021; 12: 717045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Cegielski JP, Chan PC, Lan Z, et al. Aminoglycosides and capreomycin in the treatment of multidrug-resistant tuberculosis: individual patient data meta-analysis of 12 030 patients from 25 countries, 2009–2016. Clin Infect Dis 2021; 73: e3929–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.The CRyPTIC Consortium. A data compendium associating the genomes of 12,289 Mycobacterium tuberculosis isolates with quantitative resistance phenotypes to 13 antibiotics. PLoS Biol 2022; 20: e3001721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Walker TM, Miotto P, Köser CU, et al. The 2021 WHO catalogue of Mycobacterium tuberculosis complex mutations associated with drug resistance: a genotypic analysis. Lancet Microbe 2022; 3: e265–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Sibandze DB, Kay A, Dreyer V, et al. Rapid molecular diagnostics of tuberculosis resistance by targeted stool sequencing. Genome Med 2022; 14: 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.WHO. Target product profile for next-generation drug-susceptibility testing at peripheral centres. Geneva: World Health Organization; 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Lazarchik A, Nyaruhirira AU, Chiang CY, Wares F, Horsburgh CR. Global availability of susceptibility testing for second-line anti-tuberculosis agents. Int J Tuberc Lung Dis 2022; 26: 524–28. [DOI] [PubMed] [Google Scholar]