Next Article in Journal
Cerebral Tissue Oxygen Saturation Is Enhanced in Patients following Transcatheter Aortic Valve Implantation: A Retrospective Study
Previous Article in Journal
Is Prostate Urethral Lift Effective in Patients with Multiple Comorbidities?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Investigation of Clofazimine Resistance and Genetic Mutations in Drug-Resistant Mycobacterium tuberculosis Isolates

1
Clinical Research Center, Masan National Tuberculosis Hospital, Changwon-si 51755, Korea
2
Department of Laboratory Medicine, Masan National Tuberculosis Hospital, Changwon-si 51755, Korea
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(7), 1927; https://doi.org/10.3390/jcm11071927
Submission received: 2 March 2022 / Revised: 25 March 2022 / Accepted: 26 March 2022 / Published: 30 March 2022
(This article belongs to the Topic Infectious Diseases)

Abstract

:
Recently, as clofazimine (CFZ) showed a good therapeutic effect in treating multi-drug-resistant tuberculosis (MDR-TB), the anti-tuberculosis activity and resistance were re-focused. Here, we investigated the CFZ resistance and genetic mutations of drug-resistant Mycobacterium tuberculosis (DR-Mtb) isolates to improve the diagnosis and treatment of drug-resistant TB patients. The minimal inhibitory concentration (MIC) of CFZ was examined by resazurin microtiter assay (REMA) with two reference strains and 122 clinical isolates from Korea. The cause of CFZ resistance was investigated in relation to the therapeutic history of patients. Mutations of Rv0678, Rv1979c and pepQ of CFZ resistant isolates were analyzed by PCR and DNA sequencing. The rate of CFZ resistance with MIC > 1 mg/L was 4.1% in drug-resistant Mtb isolates. The cause of CFZ resistance was not related to treatment with CFZ or bedaquiline. A CFZ susceptibility test should be conducted regardless of dugs use history. The four novel mutation sites were identified in the Rv0678 and pepQ genes related to CFZ resistance in this study.

1. Introduction

The CFZ, a fat-soluble riminophenazine dye-based antibiotic, has both antimycobacterial and anti-inflammatory activities [1,2]. The action of CFZ has suggested that the outer membrane is its primary action site, and the respiratory chain and ion transporters are the putative targets [3,4]. CFZ has shown activity against Mtb, including multidrug-resistant strains in vitro and in animal studies [5,6]. However, the inconsistency of therapeutic effects in animal infection experiments did not make them a choice for TB treatment [7,8]. In addition, CFZ has a relatively low drug concentration in plasma, skin, and gastrointestinal side effects and a long drug half-life [9,10]. It has not received attention as a treatment for TB as an oral drug. The interest in a CFZ-containing therapeutic regimen for TB has been re-focused after a study showed that a regimen containing CFZ and other drugs including high-dose fluoroquinolones was very effective against MDR in Bangladesh [11]. A systematic review presented observational studies and found that after treatment with CFZ, those with MDR and XDR TB experienced favorable treatment outcomes [12]. CFZ has recently attracted interest in DR-TB management, which is one of the major public health problems with TB treatment. According to the TB treatment guidelines that the WHO revised in 2016, CFZ, which was a group 5 drug, was newly included in the core drugs as it was classified as group C of the second drug [13]. A meta-analysis concluded that MDR-TB patients’ treatment outcome was significantly better when using linezolid, a later generation of fluoroquinolone, bedaquiline, CFZ and carbapenems, compared with standard treatment [14]. CFZ was added as a second-line drug and administered to MDR patients for more than nine months in multi-country clinical studies [12,13,14]. However, CFZ’s adverse effects of skin pigmentation occurred in most patients [12]. The study of the distribution of CFZ resistance to Mtb is needed for patients’ benefits and risks.
A breakpoint for CFZ susceptibility testing has not been defined by the Clinical & Laboratory Standards Institute (Wayne, PA, USA) or US Food and Drug Administration (Silver Spring, MD, USA), although the WHO has tentatively determined a critical concentration of 1 mg/L in MGIT based on small studies and unpublished data [15]. Previous studies have demonstrated that CFZ has MICs for Mtb, including multidrug-resistant strains, typically ranging from 0.125 to 2.0 mg/L [16,17,18]. Some studies have recommended CFZ breakpoints that range from 0.25 to 1 mg/L [19,20]. The resistance of CFZ supporting laboratory and clinical evidence is considered insufficient [21,22].
Resistance-associated variants (RAV) that lead to increased MICs of CFZ have been described in three genes of Mtb: Rv0678, Rv1979c and pepQ [23,24,25]. RAVs in Rv0678, a gene regulating the expression of the MmpS5-MmpL5 efflux pump, lead to 2- to 4-fold increases in CFZ MIC [23,26]. They have been isolated in vitro upon exposure to CFZ. However, the small number of clinical isolates related to CFZ resistance with MICs > 1 mg/L were reported in previous studies [24,27,28,29]. The mutations in Rv1979c, encoding a probable amino acid membrane transporter with permease activity, were reportedly associated with CFZ resistance in vitro [23] and in a clinical isolate with CFZ MICs > 1.2 mg/L [24]. The mutations in the putative proline aminopeptidase gene pepQ have been shown to confer low-level cross-resistance between bedaquiline and CFZ in vitro [25].
This study investigates 122 drug-resistant clinical isolates and two reference strains of Mtb of CFZ resistance. We determined the range of MIC to CFZ and investigated the genetic variability with CFZ resistance to improve for diagnosis and treatment of drug-resistant TB patients.

2. Materials and Methods

2.1. Clinical Isolates of Mtb

The 82 MDR and 40 XDR isolates were supplied from the Tuberculosis Specimen Bank of Masan National Hospital. The clinical isolates were collected from November 2009 to January 2016. TB that is resistant to both isoniazid and rifampin, which are representative primary anti-TB drugs, is defined as MDR. Among them is one resistant to one of the injectable drugs and fluoroquinolone drugs, which is defined as XDR [26]. The definition of XDR before 2021 was used for drug resistance criteria. The isolates used in this study did not have drug susceptibility test results for bedaquiline or linezolid. The clinical isolates of Mtb were sequentially cultured in Middlebrook 7H9 Broth (BBLTM MGIT TM Mycobacteria Growth Indicator Tube, Becton-Dickinson, Sparks, MD, USA) and Ogawa II agar (Asanpharm, Seoul, Korea). The bacterial colonies cultured 21 to 26 days on the agar were taken and applied to determine MIC, PCR, and DNA sequencing.

2.2. Determination of MIC

The CFZ MIC of Mtb was measured using the resazurin microtiter assay (REMA) because it is inexpensive, rapid, and simple to perform [30]. Resazurin is an oxidation–reduction indicator. A change from blue to pink indicates a reduction of resazurin and therefore bacterial growth. The REMA plate method was performed in 7H9-S broth containing Middlebrook broth, 0.1% Casitone, and 0.5% glycerol and supplemented with oleic acid, albumin, dextrose, and catalase (Becton-Dickinson, Sparks, MD, USA). CFZ (Sigma-Aldrich, St. Louis, MO, USA) was added to 7H9-S broth, with final concentrations ranging from 0.031 to 16 mg/L in 96-well plates for convenience of results reading. Growth controls containing no antibiotic and sterility controls without inoculation were also included. The Mtb H37Rv (ATCC 25618) and Mtb K (NCCP 15986) were used as reference strain validation of REMA. The inoculum was adjusted to McFarland 1.0 with DensiCHEKTM plus instrument (bioMereux, Marcy-l'Étoile, France) from fresh colonies on Ogawa II agar and further diluted 1:10 in 7H9-S broth, and 100 μL was used as an inoculum. The plates were covered, sealed in plastic bags, and incubated at 37 °C in the normal atmosphere. After 7 days of incubation, 30 μL of resazurin (Fisher scientific, Waltham, MA, USA) solution was added to each well, incubated for up to 48 h at 37 °C, and assessed for color development. The MIC was defined as the lowest drug concentration that prevented this color change. The resistance criterion for CFZ was read as >1 mg/L [20,23]. Drug susceptibility tests for the isolates showed MIC > 0.5 mg/L in CFZ and were repeated twice.

2.3. Statistical Analysis

The comparison of resistance of MDR and XDR isolates to CFZ were performed using the one-sample t-test. A cross-tabulation analysis was used to assess CFZ resistance between MDR and XDR isolates. The MICs were recorded as discrete ordinal values and were not continuous, and resistant numbers of isolates were small, so we used the nonparametric test. The paired-sample t-test was used to compare the MIC50 and MIC90 of MDR and XDR isolates to CFZ. A p-value of <0.05 was considered statistically significant. All statistical analyses were performed using SPSS Statistics version 20.0 (IBM, Armonk, NY, USA).

2.4. Clinical Data Analysis of CFZ-Resistant Isolates

The clinical information was invested by medical staff in electronical medical records to analyze the relationship of treatment history and progress to patients with the CFZ resistance acquisition of the Mtb isolates.

2.5. PCR and DNA Sequencing

Mtb DNA was extracted using a commercial kit, DNeasyTM UltraCleanTM Microbial Kit (Qiagen, Hilden, Germany) for PCR of the CFZ resistance-related genes. PCR amplification of the genes were performed using Maxime™ PCR premix i-StarTaq (iNtRON, Seongnam, Korea). The two reference strains and five isolates with a CFZ MIC of >1 were selected, and these DNA were extracted to invest the CFZ resistance gene mutation. The Rv0678, Rv1979c, and pepQ genes were amplified by PCR, respectively. The genomic DNA from CFZ-resistant mutants was isolated and reference strains were subjected to PCR amplification using Rv0678 primers rv0678F (5′-TGCCTTCGGAACCAAAGAA-3′) and rv0678R (5′-GACAACACGGTCACCTACAA-3′) as described previously [23] The Rv1979 gene was PCR-amplified using primers rv1979cF (5′-GCGGCGGAAATGAGTGT-3′) and re1979cR (5′-ATGCACGACGGCTTTATCA-3′) [23]. The gene was PCR-amplified using primers pepQF (5′-ATCAATGCCCCCTGGAAC-3′) and pepQR (5′-GCAGTTCTTCAACTTGGTG-3′) [25]. The PCR products were sequenced with the same primers used for amplification by Bioneer (Daejeon, Korea). For analysis of the sequences, ClustalW analysis of Mega 10.2.5 software was used to align and compare resistance-related gene sequences [31,32]. The gene polymorphisms were identified by aligning with the reference strain H37Rv (GenBank ID: NC_000962.3).

3. Results

3.1. CFZ MIC to Mtb

The CFZ’s MIC value of the H37Rv used as a reference strain was 0.25 to 0.5, and K was from 0.13 to 0.25, respectively. The distribution of Mtb isolates at the MIC of CFZ shown in Figure 1. Among the 122 isolates consisting of 82 MDR and 40 XDR, the number of bacteria showing MIC > 1 mg/L as determined by CFZ resistance was 4.1%. The CFZ resistance rate was 1.2% in MDR and 10.0% in XDR isolates, respectively. The CFZ resistance rate was higher in XDR than in MDR isolates (p = 0.001). CFZ’s MIC50 and MIC90 values, which mean 50% and 90% inhibition of the target bacteria, in all isolates were 0.13 and 0.25, respectively. The MDR CFZ’s MIC50 and MIC90 were 0.13 and 0.25, respectively. The XDR CFZ’s MIC50 and MIC90 were 0.25 and 0.5. The CFZ’s MIC50 values and MIC90 values were not statically significant (p = 0.09 and p = 0.11, respectively).

3.2. Clinical Data Analysis of CFZ Resistant Isolates

Drug resistance data of clinical Mtb isolates are shown Table 1. The five CFZ-resistant Mtb were isolated from four patients between 2010 and 2014. A patient was diagnosed with MDR TB, and the other three patients were diagnosed with XDR TB. The treatment history of patients and bacterial culture test of isolates are shown Figure 2. The P1 patient with MDR isolates was treatment-completed. The P2 was a chronic excretor, patients of sputum AFB smear-positive and culture-positive after treatment and who died from treatment failure. The P3 patient was a chronic excretor and was in treatment failure. The P3 patient had a history of arbitrarily stopping taking CFZ for about eight months and bedaquiline (BDQ) for six months during inpatient treatment. However, the two isolates were cultured from the patient before administering CFZ and BDQ. The P4 patient was loss to follow-up. The three patients had no history of taking CFZ and BDQ during hospitalization.

3.3. Sequencing of Genes Related to Resistance

The mutations of CFZ resistant isolates were observed in Rv0678 and pepQ in this study. The results showed that no mutation was observed in Rv1979c among all the isolates. The CFZ resistance-related gene mutation sites and related isolates are shown in Figure 3. The G138 insertion in the Rv0678 gene, known as an efflux pump Mmpl5 suppressor, was observed in CFZ-resistant isolate 9199. The G149A mutation in the Rv0678 gene, was observed in two CFZ-resistant isolates, 21873 and 22049. The mutation of G415T of pepQ known as cytoplasmic petidase was observed in isolate 22018. The mutation of G1010T was observed in isolate 9199. The G138 insertion of Rv0678 and G1010T of pepQ were observed from the same isolate 9199. The mutation sites of isolate 22033 were not detected in CFZ resistance-related genes. The 21873 strain is MDR. The 22033 and 22049 strains were all resistant to rifampin with XDR, and the sensitivity to bedaquiline could not be confirmed in this study. The 22018 and 22033 strains were isolated from the same patient. This patient had a history of arbitrarily stopping taking CFZ for about 6 months during inpatient treatment. However, as a result of administering CFZ after the bacteria was isolated, the association between drug discontinuation and Mtb mutation is expected to be low.

4. Discussion

One of the major problems with TB treatment is the emergence of MDR-TB and XDR-TB. Recently, the WHO has updated the definition of XDR-TB [33]. The new definition of pre-XDR is TB that meets MDR and rifampicin-resistant TB requirements and is resistant to fluoroquinolone. XDR meets MDR-TB and is resistant to fluoroquinolone and is resistant to one or more additional group A drugs, bedaquiline or linezolid. This leads to adequate access to treatment options for patients with resistant TB. There were no drug susceptibility test results for bedaquiline and linezolid in this study, so it is difficult to apply the latest accurate XDR bacteria. However, using the fluoroquinolone drug test results of isolates, most of the XDR isolates in this study correspond to pre-XDR. Through this study, CFZ resistance is suspected to be caused by drugs other than bedaquiline or CFZ, so it is necessary to conduct a drug susceptibility test before deciding to treat pre-XDR patients with CFZ for preventing adverse events from unnecessary drug administration.
A breakpoint concentration of 1 mg/L for CFZ susceptibility testing using the MGIT 960 method was proposed after the study of 26 multidrug-resistant clinical isolates revealed an MIC90 of 0.25 mg/L [15]. Previous studies have demonstrated that CFZ has MICs for Mtb H37Rv that typically range from 0.5 to 1.0 mg/L [18,20]. The in vitro resistance study surveyed CFZ MIC values for 90 clinical isolates mostly from patients with XDR-TB. The five (5.6%) isolates with MICs of >1.2 mg/L were determined as CFZ-resistant [20]. The five (5.6%) CFZ-resistant strains determined >1.2 mg/L isolated from 80 drug-resistant isolates and 10 drug-susceptible isolates [24]. The other previous study demonstrated that 27.7% of the 195 MDR-TB isolates were CFZ-resistant with MICs of >1 mg/L [34]. The serum peak of CFZ is 0.369 mg/L after a single 200 mg dose administered with anti-TB medicines and a high-fat meal and 1.0 mg/L average serum concentration for 300 mg daily dosing [35,36]. The MIC for CFZ of H37Rv observed in this study was similar when compared with previous research reports. The rate of CFZ resistance with MIC > 1 mg/L was 4.1% in DR-Mtb isolates in this study. The CFZ resistance rate was higher in XDR than in MDR isolates. The CFZ rates of MDR and XDR observed in this study were different when compared with previous research reports. The results of this study could be referred to as defining the breakpoint concentration for CFZ susceptibility testing.
The rv0678 gene, known as transcription repressors of the efflux pumps MmpL5 and MmpS5, has been reported to be related to drug resistance to rifampin, bedaquiline, and CFZ [20,23,24,26,29]. The CFZ-resistant isolates in this study did not have the same positions of mutation of Rv0678 gene’s previous reports, the sites of T2C, G193 deletion, G193 insertion, C466T, A202G, and C364 insertion. Although the site of resistance mutation is different from previous reports, mutations at different positions in the Rv0678 gene mean different amino acid expression, which is expected to be involved in CFZ resistance by increasing efflux pump expression.
The G265T, T157C, two frameshifts at codons 14 and 271 mutation sites of the pepQ gene, known as cytoplasmic petidase, presumed that resistance was mediated by a loss of function mutations [23,24,25]. The Leu to Ile at codon 145 of the pepQ gene mutations related to bedaquiline and CFZ resistance have been reported in clinical isolates [37]. We could not observe the same position of pepQ mutations of previous studies in the CFZ-resistant isolates in this study. However, mutations of G415T and G1010T were observed in our isolate. It is expected that inactivation of the protein expressed by the pepQ gene and changes in the amino acid structure will occur. The function of pepQ is to inhibit drug efflux, so it is presumed that the occurrence of mutations in this region increases the efflux pump function. Further studies such as efflux pump function and related drugs are needed to elucidate the mechanism of CFZ resistance.
Rv1979c is a putative permease that might be involved in amino acid transport [23,24]. Rv1979c might be involved in CFZ transport or uptake, either directly or indirectly, to alter the physiology of the bacteria to be less susceptible to the effect of CFZ. No mutation was detected in Rv1979c among CFZ resistant isolates in this study.
Although an isolate 22033 strain was resistant to CFZ, no mutation sites were found in the Rv0678, Rv1979c, and pepQ genes. It is necessary to elucidate the cause of resistance of this isolate through additional research such as whole-genome sequencing analysis.
Our study has too few CFZ-resistant strains and the retrospective nature of the analysis limits our conclusion of the molecular determinants of resistance to CFZ phenotype. The clinical CFZ-resistant strains have not been commonly observed in previous clinical studies [20,34]. The other studies have found some mutations potentially associated with resistance at lower MICs with >1 mg/L [25,29]. Testing of a larger number of strains from multicenter work is needed to conclusively identify a breakpoint value for CFZ with the REMA method and fully understand the association of CFZ resistant gene mutations with distinguished CFZ MICs.

Author Contributions

Conceptualization, S.P.; methodology, J.J.; formal analysis, J.K. and S.B.H.; writing—original draft preparation, S.P.; writing—review and editing, S.P.; supervision, S.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded an intramural grant (4631-304-210-13) from Masan National Tuberculosis Hospital, Korea.

Institutional Review Board Statement

The study was approved by institutional review board (IRB-398837-2018-E-26) and institutional biosafety committee (MTHIBC-18-11) at the Masan national tuberculosis hospital.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data that support the findings of this study are available on request from the corresponding author, Park S. The data are not publicly available due to their containing information that could compromise the privacy of research participants.

Acknowledgments

We thank the researchers of the clinical research center and the staff of the department of laboratory medicine at Masan National Tuberculosis Hospital.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results. References in this manuscript to any specific commercial products, process, service, manufacturer, or company do not constitute endorsement or recommendation by the Korean government or Masan National Tuberculosis Hospital. The findings and conclusions are solely those of the authors.

References

  1. Barry, V.C.; Belton, J.G.; Conalty, M.L.; Denneny, J.M.; Edward, D.W.; O’Sullivan, J.F.; Twomey, D.; Winder, F. A new series of phenazine (rimino-compounds) with high anti-tuberculosis activity. Nature 1957, 179, 1013–1015. [Google Scholar] [CrossRef] [PubMed]
  2. Van Rensburg, C.E.; Jooné, G.K.; O’Sullivan, J.F.; Anderson, R. Antimicrobial activities of clofazimine and B669 are mediated by lysophospholipids. Antimicrob. Agents Chemother. 1992, 36, 2729–2735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lechartier, B.; Cole, S.T. Mode of action of Clofazimine and combination therapy with benzothiazinones against Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 2015, 59, 4457–4463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Yano, T.; Kassovska-Bratinova, S.; Teh, J.S.; Winkler, J.; Sullivan, K.; Isaacs, A.; Schechter, N.M.; Rubin, H. Reduction of clofazimine by mycobacterial type 2 NADH: Quinone oxidoreductase: A pathway for the generation of bactericidal levels of reactive oxygen species. J. Biol. Chem. 2011, 286, 10276–10287. [Google Scholar] [CrossRef] [Green Version]
  5. Reddy, V.M.; O’Sullivan, J.F.; Gangadharam, P.R.J. Antimycobacterial activities of riminophenazines. J. Antimicrob. Chemother. 1999, 43, 615–623. [Google Scholar] [CrossRef]
  6. Van Rensburg, C.E.; Jooné, G.K.; Sirgel, F.A.; Matlola, N.M.; O’Sullivan, J.F. In vitro investigation of the antimicrobial activities of novel tetramethylpiperidine-substituted phenazines against Mycobacterium tuberculosis. Chemotherapy 2000, 46, 43–48. [Google Scholar] [CrossRef]
  7. Barry, V.C.; Buggle, K.; Byrne, J.; Conalty, M.L.; Winder, F. Absorption, distribution and retention of the riminocompounds in the experimental animal. Ir. J. Med. Sci. 1960, 416, 345–352. [Google Scholar] [CrossRef]
  8. Barry, V.C.; Conalty, M.L. The antimycobacterial activity of B663. Lepr. Rev. 1965, 36, 3–7. [Google Scholar]
  9. O’Connor, R.; O’Sullivan, J.F.; O’Kennedy, R. The pharmacology, metabolism and chemistry of clofazimine. Drug Metab. Rev. 1995, 27, 591–614. [Google Scholar] [CrossRef]
  10. Van Rensburg, C.E.; Anderson, R.; O’Sullivan, J.F. Riminophenazine compounds: Pharmacology and anti-neoplastic potential. Crit. Rev. Oncol. Hematol. 1997, 25, 55–67. [Google Scholar] [CrossRef]
  11. Van Deun, A.; Maug, A.K.J.; Salim, M.A.H.; Das, P.K.; Sarker, M.R.; Daru, P.; Rieder, H.L. Short, highly effective, and inexpensive standardized treatment of multidrug-resistant tuberculosis. Am. J. Respir. Crit. Care Med. 2010, 182, 684–692. [Google Scholar] [CrossRef] [PubMed]
  12. Gopal, M.; Padayatchi, N.; Metcalfe, J.Z.; O’Donnell, M.R. Systematic review of clofazimine for the treatment of drug-resistant tuberculosis. Int. J. Tuberc. Lung. Dis. 2013, 17, 1001–1007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. World Health Organization. WHO Treatment Guidelines for Drug-Resistant Tuberculosis, 2016 Update; WHO: Geneva, Switzerland, 2016; ISBN 9789241549639. [Google Scholar]
  14. Ahmad, N.; Ahuja, S.D.; Akkerman, O.W.; Alffenaar, J.-W.C.; Anderson, L.F.; Baghaei, P.; Bang, D.; Barry, P.M.; Bastos, M.L.; Behera, D.; et al. Treatment correlates of successful outcomes in pulmonary multidrug-resistant tuberculosis: An individual patient data meta- analysis. Lancet 2018, 392, 821–834. [Google Scholar] [CrossRef] [Green Version]
  15. World Health Organization. Technical Report on Critical Concentrations for Drug Susceptibility Testing of Medicines Used in the Treatment of Drug-Resistant Tuberculosis; WHO: Geneva, Switzerland, 2018; Available online: http://www.who.int/iris/handle/10665/260470 (accessed on 5 January 2022).
  16. Diacon, A.H.; Dawson, R.; von Groote-Bidlingmaier, F.; Symons, G.; Venter, A.; Donald, P.R.; van Niekerk, C.; Everitt, D.; Hutchings, J.; Burger, D.A.; et al. Bactericidal activity of pyrazinamide and clofazimine alone and in combinations with pretomanid and bedaquiline. Am. J. Respir. Crit. Care. Med. 2015, 191, 943–953. [Google Scholar] [CrossRef]
  17. Schön, T.; Jureen, P.; Chryssanthou, E.; Giske, C.G.; Sturegård, E.; Kahlmeter, G.; Hoffner, S.; Angeby, K.A. Wild-type distributions of seven oral second-line drugs against Mycobacterium tuberculosis. Int. J. Tuberc. Lung. Dis. 2011, 15, 502–509. [Google Scholar] [CrossRef]
  18. Cavanaugh, J.S.; Jou, R.; Wu, M.H.; Dalton, T.; Kurbatova, E.; Ershova, J.; Cegielski, J.P.; Global PETTS Investigators. Susceptibilities of MDR Mycobacterium tuberculosis isolates to unconventional drugs compared with their reported pharmacokinetic/pharmacodynamic parameters. J. Antimicrob. Chemother. 2017, 72, 1678–1687. [Google Scholar] [CrossRef]
  19. Jagannath, C.; Reddy, M.V.; Kailasam, S.; O’Sullivan, J.F.; Gangadharam, P.R. Chemotherapeutic activity of clofazimine and its analogues against Mycobacterium tuberculosis. In vitro, intracellular, and in vivo studies. Am. J. Respir. Crit. Care. Med. 1995, 151, 1083–1086. [Google Scholar] [CrossRef]
  20. Pang, Y.; Zong, Z.; Huo, F.; Jing, W.; Ma, Y.; Dong, L.; Li, Y.; Zhao, L.; Fu, Y.; Huang, H. In vitro drug susceptibility of bedaquiline, delamanid, linezolid, clofazimine, moxifloxacin, and gatifloxacin against extensively drug-resistant tuberculosis in Beijing, China. Antimicrob. Agents Chemother. 2017, 61, e00900–e00917. [Google Scholar] [CrossRef] [Green Version]
  21. Aung, K.J.; van Deun, A.; Declercq, E.; Sarker, M.R.; Das, P.K.; Hossain, M.A.; Rieder, H.L. Successful 9-month Bangladesh regimen’ for multidrug-resistant tuberculosis among over 500 consecutive patients. Int. J. Tuberc. Lung Dis. 2014, 18, 1180–1187. [Google Scholar] [CrossRef]
  22. Wang, Q.; Pang, Y.; Jing, W.; Liu, Y.; Wang, N.; Yin, H.; Zhang, Q.; Ye, Z.; Zhu, M.; Li, F.; et al. Clofazimine for treatment of extensively drug-resistant pulmonary tuberculosis in China. Antimicrob. Agents Chemother. 2018, 62, e02149. [Google Scholar] [CrossRef] [Green Version]
  23. Zhang, S.; Chen, J.; Cui, P.; Shi, W.; Zhang, W.; Zhang, Y. Identification of novel mutations associated with clofazimine resistance in Mycobacterium tuberculosis. J. Antimicrob. Chemother. 2015, 70, 2507–2510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Xu, J.; Wang, B.; Hu, M.; Huo, F.; Guo, S.; Jing, W.; Nuermberger, E.; Lu, Y. Primary clofazimine and bedaquiline resistance among isolates from patients with multidrug-resistant tuberculosis. Antimicrob. Agents Chemother. 2017, 61, e00239-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Almeida, D.; Ioerger, T.; Tyagi, S.; Li, S.Y.; Mdluli, K.; Andries, K.; Grosset, J.; Sacchettini, J.; Nuermberger, E. Mutations in pepQ Confer Low-Level Resistance to Bedaquiline and Clofazimine in Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 2016, 60, 4590–4599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Andries, K.; Villellas, C.; Coeck, N.; Thys, K.; Gevers, T.; Vranckx, L.; Lounis, N.; de Jong, B.C.; Koul, A. Acquired resistance of Mycobacterium tuberculosis to bedaquiline. PLoS ONE 2014, 9, e102135. [Google Scholar] [CrossRef] [Green Version]
  27. Bloemberg, G.V.; Keller, P.M.; Stucki, D.; Trauner, A.; Borrell, S.; Latshang, T.; Coscolla, M.; Rothe, T.; Hömke, R.; Ritter, C.; et al. Acquired resistance to bedaquiline and delamanid in therapy for tuberculosis. N. Engl. J. Med. 2015, 373, 1986–1988. [Google Scholar] [CrossRef] [Green Version]
  28. Hoffmann, H.; Kohl, T.A.; Hofmann-Thiel, S.; Merker, M.; Beckert, P.; Jaton, K.; Nedialkova, L.; Sahalchyk, E.; Rothe, T.; Keller, P.M.; et al. Delamanid and bedaquiline resistance in Mycobacterium tuberculosis ancestral Beijing genotype causing extensively drug-resistant tuberculosis in a Tibetan refugee. Am. J. Respir. Crit. Care Med. 2016, 193, 337–340. [Google Scholar] [CrossRef] [Green Version]
  29. Somoskovi, A.; Bruderer, V.; Homke, R.; Bloemberg, G.V.; Bottger, E.C. A mutation associated with clofazimine and bedaquiline cross-resistance in MDR-TB following bedaquiline treatment. Eur. Respir. J. 2015, 45, 554–557. [Google Scholar] [CrossRef] [Green Version]
  30. Martin, A.; Camacho, M.; Portaels, F.; Palomino, J.C. Resazurin microtiter assay plate testing of Mycobacterium tuberculosis susceptibilities to second-line drugs: Rapid, simple, and inexpensive method. Antimicrob. Agents Chemother. 2003, 7, 3616–3619. [Google Scholar] [CrossRef] [Green Version]
  31. Higgins, D.; Thompson, J.; Gibson, T.; Thompson, J.D.; Higgins, D.G.; Gibson, T.J. CLUSTAL W: Improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Res. 1994, 22, 4673–4680. [Google Scholar]
  32. Kumar, S.; Stecher, G.; Li, M.; Knyaz, C.; Tamura, K. MEGA X: Molecular Evolutionary Genetics Analysis across Computing Platforms. Mol. Biol. Evol. 2018, 35, 1547–1549. [Google Scholar] [CrossRef]
  33. World Health Organization. Meeting Report of the WHO Expert Consultation on the Definition of Extensively Drug-Resistant Tuberculosis; WHO: Geneva, Switzerland, 2021; ISBN 978-92-4-0001866-2. [Google Scholar]
  34. Zhang, Z.; Li, T.; Qu, G.; Pang, Y.; Zhao, Y. In vitro synergistic activity of clofazimine and other antituberculous drugs against multidrug-resistant Mycobacterium tuberculosis isolates. Int. J. Antimicrob. Agents. 2015, 45, 71–75. [Google Scholar] [CrossRef] [PubMed]
  35. Xu, J.; Lu, Y.; Fu, L.; Zhu, H.; Wang, B.; Mdluli, K.; Upton, A.M.; Jin, H.; Zheng, M.; Zhao, W.; et al. In vitro and in vivo activity of clofazimine against Mycobacterium tuberculosis persisters. Int. J. Tuberc. Lung Dis. 2012, 16, 1119–1125. [Google Scholar] [CrossRef] [PubMed]
  36. Steel, H.C.; Matlola, N.M.; Anderson, R. Inhibition of potassium transport and growth of mycobacteria exposed to clofazimine and B669 is associated with a calcium-independent increase in microbial phospholipase A2 activity. J. Antimicrob. Chemother. 1999, 44, 209–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Liu, Y.; Gao, J.; Du, J.; Shu, W.; Wang, L.; Wang, Y.; Xue, Z.; Li, L.; Xu, S.; Pang, Y. Acquisition of clofazimine resistance following bedaquiline treatment for multidrug-resistant tuberculosis. Int. J. Infect. Dis. 2021, 102, 392–963. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Distribution of minimum inhibitory concentration (MIC, mg/L) of CFZ for MDR (n = 82, gray bar) and XDR (n = 40, black bar). The resistance criterion for CFZ was read as >1 mg/L. The CFZ resistance rate was higher in XDR than in MDR isolates (p = 0.001).
Figure 1. Distribution of minimum inhibitory concentration (MIC, mg/L) of CFZ for MDR (n = 82, gray bar) and XDR (n = 40, black bar). The resistance criterion for CFZ was read as >1 mg/L. The CFZ resistance rate was higher in XDR than in MDR isolates (p = 0.001).
Jcm 11 01927 g001
Figure 2. The treatment history and isolates of patients. M, month; AMX/CLV, amoxicillin/clavulanate; BDQ, bedaquiline; CFZ, clofazimine; CLR, clarithromycin; CS, cycloserine; KM, kanamycin; LZD, linezolid; MFX, moxifloxacin; PAS, para-aminosalicylic acid; PTO, prothionamide; RFP, rifampicin; PZA, pyrazinamide.
Figure 2. The treatment history and isolates of patients. M, month; AMX/CLV, amoxicillin/clavulanate; BDQ, bedaquiline; CFZ, clofazimine; CLR, clarithromycin; CS, cycloserine; KM, kanamycin; LZD, linezolid; MFX, moxifloxacin; PAS, para-aminosalicylic acid; PTO, prothionamide; RFP, rifampicin; PZA, pyrazinamide.
Jcm 11 01927 g002
Figure 3. CFZ resistance-related gene mutation sites and related isolates and reference strains. (A) Rv0678 gene; (B) pepQ gene. The mutation site has been shown with white arrows and squares. The prediction of amino-acid transition has been shown on the gene mutation site. H37Rv and K were used as a reference for comparison.
Figure 3. CFZ resistance-related gene mutation sites and related isolates and reference strains. (A) Rv0678 gene; (B) pepQ gene. The mutation site has been shown with white arrows and squares. The prediction of amino-acid transition has been shown on the gene mutation site. H37Rv and K were used as a reference for comparison.
Jcm 11 01927 g003
Table 1. Drug resistance data of clinical Mtb isolates.
Table 1. Drug resistance data of clinical Mtb isolates.
PatientsIsolatesTypeDrug Resistance ProfileYear of Isolation
P121873MDRINH, RFP, EMB, RBU, PTO2014
P29199XDRINH, RFP, EMB, RBU, SM, KM, AMK, OFX, MFX, PAS, PTO, CS2010
P322018XDRINH, RFP, EMB, RBU, SM, KM, AMK, OFX, LEV, PTO, CS2014
22033XDRINH, RFP, EMB, RBU, SM, KM, AMK, OFX, LEV, PTO, CS2014
P422049XDRINH, RFP, EMB, RBU, SM, KM, AMK, OFX, LEV, PTO, CS2014
AMK, amikacin; CS, cycloserine; EMB, ethambutol; INH, isoniazid; KM, kanamycin; LEV, levofloxacin; MDR, Multi-Drug Resistant; MFX, moxifloxacin; OFX, ofloxacin; PAS, para-aminosalicylic acid; PTO, prothionamide; PZA, pyrazinamide; RBU, rifabutin; RFP, rifampicin; SM, streptomycin; XDR, Extensive-Drug Resistant.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Park, S.; Jung, J.; Kim, J.; Han, S.B.; Ryoo, S. Investigation of Clofazimine Resistance and Genetic Mutations in Drug-Resistant Mycobacterium tuberculosis Isolates. J. Clin. Med. 2022, 11, 1927. https://doi.org/10.3390/jcm11071927

AMA Style

Park S, Jung J, Kim J, Han SB, Ryoo S. Investigation of Clofazimine Resistance and Genetic Mutations in Drug-Resistant Mycobacterium tuberculosis Isolates. Journal of Clinical Medicine. 2022; 11(7):1927. https://doi.org/10.3390/jcm11071927

Chicago/Turabian Style

Park, Sanghee, Jihee Jung, Jiyeon Kim, Sang Bong Han, and Sungweon Ryoo. 2022. "Investigation of Clofazimine Resistance and Genetic Mutations in Drug-Resistant Mycobacterium tuberculosis Isolates" Journal of Clinical Medicine 11, no. 7: 1927. https://doi.org/10.3390/jcm11071927

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop