JCDR - Register at Journal of Clinical and Diagnostic Research
Journal of Clinical and Diagnostic Research, ISSN - 0973 - 709X
Microbiology Section DOI : 10.7860/JCDR/2020/43876.13896
Year : 2020 | Month : Aug | Volume : 14 | Issue : 08 Full Version Page : DC01 - DC05

Identification of Non-tuberculous Mycobacteria by Partial Gene Sequencing and Matrix Assisted Laser Desorption Ionisation Time of Flight at a Tertiary Care Hospital in Telangana, India

Neelima Angaali1, Rajashekhar Kadasu2, Madhusudhan Apparao Patil3, Vijay Dharma Teja4

1 Assistant Professor, Department of Microbiology, Nizam’s Institute of Medical Sciences, Hyderabad, Telangana, India.
2 Ph.D. Scholar, Department of Microbiology, Nizam’s Institute of Medical Sciences, Hyderabad, Telangana, India.
3 Additional Professor, Department of Microbiology, Nizam’s Institute of Medical Sciences, Hyderabad, Telangana, India.
4 Professor and Head, Department of Microbiology, Nizam’s Institute of Medical Sciences, Hyderabad, Telangana, India.


NAME, ADDRESS, E-MAIL ID OF THE CORRESPONDING AUTHOR: Dr. Neelima Angaali, Raja Residency, Flat-201, Street No. 7, Himayat Nagar, Hyderabad-500029, Telangana, India.
E-mail: neelimasudharshan@gmail.com
Abstract

Introduction

Non-tuberculous Mycobacteria (NTM) pulmonary disease is often unrecognised or misdiagnosed as Mycobacterium tuberculosis (MTB), Multi-Drug Resistant Tuberculosis (MDRTB), because of similar clinical presentation in counties with high burden of Tuberculosis (TB) including India. In India due to lack of awareness among clinicians and lack of laboratory facilities to diagnose these infections, its prevalence is largely unknown.

Aim

To evaluate the efficacy of identification of NTM species by Matrix Assisted Laser Desorption Ionisation Time of Flight (MALDI-TOF) and Heat Shock Protein (hSP65) gene sequencing and to determine their Antimicrobial Susceptibility Testing (AST).

Materials and Methods

All the clinical specimens from pulmonary and extra-pulmonary TB suspects at Nizam’s Institute of Medical Sciences, Hyderabad, Telangana, India, over a period of one year i.e., from June 2017 to May 2018 were included in the study. The specimens were subjected to microscopy, culture and GeneXpert. The NTMs isolated in the culture were further characterised genotypically by MALDI-TOF and hsp65 gene sequencing. The identified NTM isolates were subjected to AST. All the methods were followed as per the standard protocols. Data was analysed using SPSS 25.

Results

A total of 1085 samples were processed out of which Mycobacteria was detected in 201 cases (18.5%). Among the culture positives, MTB complex was detected in 146 cases (13.5%) and NTM in 55 (5.06%). Mycobacterium abscessus was the predominant isolate. The most common co-morbidities were bronchiectasis and Chronic Obstructive Pulmonary Disease (COPD). Linezolid, clarithromycin, moxifloxacin and amikacin showed high sensitivity.

Conclusion

Molecular assay helps in rapid identification which can lead to targeted therapy and can thus combat antimicrobial resistance. The MALDI-TOF and hsp65 gene sequencing also offers quick results at a low cost and is easy to perform hence it can be considered as an alternate diagnostic tool for identification.

Keywords

Introduction

NTM are mycobacteria which do not cause tuberculsosis. Also, known as atypical Mycobacteria or Mycobacteria other than TB. NTM species are diverse and widespread in the environment, more than 140 NTM species have been identified [1]. In the past, majority of the NTM species were considered as colonisers or environmental contaminants but now increasingly recognised as important pulmonary pathogens in both immunocompromised and immunocompetent population [2]. NTM are now considered as an important cause of lung disease in various parts of the world causing morbidity and mortality [3-5]. NTM often produce infections in the presence of predisposing factors/underlying diseases. The most common being HIV infection, cystic fibrosis, underlying chronic lung disease, previous TB and work in the mining industry [2].

Depending on population and geographic location, NTM infection rates vary widely. Most reports are from developed countries that have low rates of TB [6]. NTM pulmonary disease is often unrecognised or misdiagnosed as MTB and MDR-TB, because of similar clinical presentation in counties with high burden of TB including India. In India due to lack of awareness among clinicians and lack of laboratory facilities to diagnose these infections, its prevalence is largely unknown [6]. Among few reports available in India, NTM isolation rates range from 0.5-8.6% [7]. A recent study from central India reported increasing prevalence from 1.0-3.5% in 2005-2008 [7].

As incidence of antibiotic resistance in pathogens from environment and nosocomial habitats has been increasing, these under documented microorganism needs attention because India is still fighting to overcome the scenario of TB. Most of the clinical studies showed NTM’s to be remarkably resistant or only partially susceptible to the anti-tubercular drugs such as isoniazid, rifampicin, ethambutol and pyrazinamide. Treatment for one species is not effective for others [8]. In India, accurate identification of NTM to the species level and AST is not part of routine laboratory practice, so most of them are treated empirically with anti-TB drugs. The MDR of NTM has now become a globally challenging health issue.

The present study aimed to accurately identify all isolated NTM using different molecular methods and to determine their clinical significance in a TB-endemic region (Telangana) and to know its AST.

Materials and Methods

A prospective study was conducted at Nizam’s Institute of Medical Sciences, Hyderabad, Telangana, India over a period of one year i.e., from June 2017 to May 2018. All the clinical specimens collected from pulmonary and extra-pulmonary TB suspects were included in the study. Institutional Ethics Committee approved the study. The assigned number from ethics committee was SRC/AC4/365, an informed consent was taken from all the patients included in the study. The demographic details like age, sex, occupation, address etc and clinical details such as presenting symptoms, onset of symptoms, previous TB history and co-morbidities were obtained from the subjects. A total of 1085 subjects were included in the study. Sample size was calculated using open Epi web based software using frequency of 8.5% in Indian population.

The clinical specimens were obtained as per the standard protocol [9]. Specimens, which contain normal commensal bacterial flora, were decontaminated with 4% Sodium hydroxide (NaOH) method [9]. All the specimens were subjected to microscopy, culture and geneXpert. The smear microscopy was done using the auramine-rhodamine staining method, culture by BacT/ALERT three-Dimensional (3D) system (BioMerieux, Marcyl’Etoile, France) and Lowenstein-Jensen (LJ) media [10]. The positive flagged bottles were subjected to Zeihl Neelsen (ZN) stain. Samples that failed to show any growth after six weeks of incubation was treated as negative for Mycobacteria. Cultures with positive growth on the BacT/ALERT 3D and presence of Acid-Fast Bacilli (AFB) by ZN stain were tested with a rapid TB antigen assay MPT64 (SD-Bioline Ag MPT64 Rapid TM assay; Standard Diagnostics, Kyonggi-do, Korea) which identifies antigens specific to Mycobacteria Tuberculosis Complex (MTBC). This test was done as per the manufacturer’s instructions [11,12]. The cultures which tested negative for MPT 64 were further characterised by MALDI-TOF and Gene sequencing.

MALDITOF-MS (Matrix Assisted Laser Desorption Ionization Time Of Flight- Mass Spectroscopy)

Loopful of NTM isolates grown on solid medium were taken for each organism. Add 500 μL of 70% ethanol in a pre-labeled 2 mL micro centrifuge vial containing 200 μL of 0.5 mm glass beads and capped securely. A bead beater was used for mechanical disruption of the cells for 5 minutes (or) vortex the tube for 15 minutes and incubated at room temperature for 10 minutes and centrifuged to create a pellet (e.g., 10,000 g for 2 minutes). The pellet was resuspended by adding 10 μL of 70% formic acid to the pellet and repeated aspiration/dispensation was done until the pellet was uniformly dispersed. Add 10 μL of acetonitrile to the vial and mix by repeated aspiration/dispensation, centrifuge and 1 μL of the extract supernatant for each organism was transferred to the designated spots on the target slide. Allow each spot to dry completely. One μL (1 μL) of VITEK MS-CHCA Matrix were transferred to each spot. Allow each spot to completely dry. The slide was run in the MALDI-TOF (BIOMERIEUX VITEC-MS) instrument to obtain the identification [13].

hsp65 Gene Sequencing

Deoxyribonucleic Acid (DNA) from NTM isolates was extracted as described by Van Soolingen D et al., [14]. In short, a loop-full of NTM growth was suspended in 500 μl of 1X Tris-EDTA (TE) buffer (10 mM Tris-HC1, 1 mMEDTA, pH 8.0) and heated at 90°C for 30 minutes in order to lyse the cells. Add 50 μl of lysozyme and incubated for 1 hour at 37°C, add 70 μL of 10% sodium dodecyl sulfate (W/V) and 6 μL of proteinase K (at a 10 mg/mL concentration) the mixture was incubated for 10 minutes at 65°C, followed by phenol-chloroform method used to extract the DNA. The extracted DNA samples were stored at -20°C prior to the next steps. A hsp65 gene sequencing and Polymerase Chain Reaction (PCR) amplification was done to identify the NTM species. hsp65 gene primer sets used were Tb-11 F- ACCAACGATGGTGTCTCCAT and Tb-12R- CTTGTCGAACCGCATACCCT 439 base pairs [15]. PCR reactions were performed using10 mMTris-HCl (pH 8.3), 50 mM KCl, 2 mM MgCl2, 0.2 mMdNTP mix, 0.1 U μl_1 Taq polymerase, 0.5 μM ofeach of the primers, DNA template and nuclease free water.

PCR cycle conditions for amplification of the genes hsp65 were as follows: 94°C for 4 minutes followed by 40 cycles of 94°C for 30 seconds, 60°C for 30 seconds, and 72°C for 30 seconds, and then final extension at 72°C for 10 minutes. Analysis of 3 μL of the PCR products (amplicon) was done by electrophoresis on 1.5% gel agarose. GelRed™ DNA stain was used in staining the gel and the fragments were visualised in the gel documentation system (Gel Doc, ATP Co) under UV light. QIA quick PCR purification kit (QIAGEN, Germany) was used to purify PCR products and analysed by Sanger sequencing (3130 xl-3730 xl genetic analyser, applied biosystem 16/48 capillaries of 50 cm leanth). DNASTAR Laser gene Software (version 7.1) was used to analyse the sequencing results of hsp65 genes. The sequences were compared with similar sequences of the organisms in Gene Bank using the NCBI BLAST online software of the National Center for Biotechnology Information (http://hsp65blast.phsa.ca/blast/blast.html).

Anti-Microbial Susceptibility

All isolates identified by gene sequencing and MALDI-TOF were subjected to antimicrobial susceptibility testing by Broth microdilution as per CLSI guidelines [16]. The following antimicrobials were tested-isoniazid, rifamycin, streptomycin, ethambutol, levofloxacin, moxifloxacin, linezolid, imipenem, co-trimoxazole, clarithromycin, doxycycline and amikacin.

Statistical Analysis

Descriptive statistics included frequencies and prevalence. Continous variables were expressed as median. Categorical variables were expressed as frequencies and percentage. The statistical analysis of the data was done using the microsoft excel and Statistical Package for the Social Sciences (SPSS) version 25.

Results

About 1085 samples were processed. A male preponderance was observed (33/55, 60%) while females were 22/55 (40%). The median of age was 40.5 years. The age group 41-50 years was highly affected (41%). Pulmonary samples were predominant 54 out of 55 (98.2%) and one extra pulmonary sample (1.8%). Among the 1085 samples, bronchial wash (27.8%) were maximum followed by followed by tissue, pleural fluid and Cerebrospinal fluid (CSF) [Table/Fig-1]. About 43 patients were associated with co-morbidities. The predominant co-morbidities were bronchiectasis (27.9%) and COPD (25.5%) [Table/Fig-2]. About 15.2% were direct smear positive [Table/Fig-3]. Mycobacterium was detected in 201 cases (18.5%), MTB complex in 146 cases (13.5%) and NTM in 55 (5%) [Table/Fig-4]. Among the 55 NTM isolates, 53 were identified as rapid growers (RGM) 4.8% (M.abscessus) and 2 as slow growers (SGM) 0.18% (M.simiae). MALDI-TOF identified 49 (89%) out of which 48 were M.abscessus and 1- M.simiae [Table/Fig-5].

Sample-wise distribution.

SampleNumber (n=1085)Percentage
Bronchial wash30227.8
Sputum1009.2
Lymph node211.93
Peritoneal fluid40.36
Pleural fluid12011
Synovial fluid544.97
Pus413.77
Urine847.74
Tissue15414
Gastric aspirate60.55
Ascitic fluid322.94
CSF10910
CAPD151.3
BMA171.56
Aspirated fluid151.38
FNAC70.64
Pericardial fluid40.36

CSF: Cerebrospinal fluid; CAPD: Continous ambulatory peritoneal dialysis; BMA: Bone marrow aspirate; FNAC: Fine Needle aspiration cytology


Frequency of concomitant co-morbidities in patients (n=43).

Co-morbiditiesNumber (n=43)Percentage
Bronchiectasis1227.9
COPD1125.5
Previous history of TB716.27
SLE511.6
Malignancy49.3
Sarcoidosis24.65
ILD12.32
Post renal transplant12.32

COPD: Chronic obstructive pulmonary disease; SLE: Systemic lupus erythematosis; ILD: Interstitial lung disease; TB: Tuberculosis


Auramine rhodamine flourescence stain.

The image shows golden yellow fluorescent bacilli

Growth on bact alert, LJ Media and Chocolate agar.

MALDITOF identification of M.abscessus and M.simiae.

Gene sequencing-partial gene sequencing of hsp65 gene [Table/Fig-6] identified all the 55 (100%) isolates out of which 53 (89%) were M.abscessus and 2 (11%) M.simiae. There was 89% concordance among both the molecular methods [Table/Fig-7]. Subspecies identification of M.abscessus was also done through sequencing where all of them were identified as M.abscessus subspecies abscessus.

Partial hsp65 gene sequencing.

NTM identification by MALDITOF-MS and hsp65 gene sequencing.

Total no. of isolateshsp65 gene sequencing identificationMALDITOF-MS identification% of similarity
5555 (100%)49 (89%)89.09%
Type strains identified
M. abscessus53 (96.36)48 (97.95)90.56%
M. simiae2 (3.64)1 (2.05)50%

All the identified isolates were subjected to antimicrobial susceptibility testing. M.abscessus showed 100% sensitivity to linezolid followed by clarithromycin, (95.6%), moxifloxacin, (93.4%), amikacin (93.4%). A 100% resistance was observed to rifampicin, isoniazid, ethambutol [Table/Fig-8]. M.simiae showed 100% sensitivity to clarithromycin, amikacin, levofloxacin, moxifloxacin, co-trimoxazole and 100% resistance to the first line drugs [Table/Fig-9].

AST pattern of M.abscessus.

AST Pattern of M.simiae.

Discussion

Improper identification of NTM can cause clinical and diagnostic dilemmas; just positive microscopy cannot differentiate M.tuberculosis complex from NTM. Management of patients with M.tuberculosis complex and NTM is entirely different therefore prompt isolation, detection and differentiation are necessary for proper and accurate treatment [17,18]. NTM prevalence varies across the globe [Table/Fig-10] [19]. Karak K et al., from Kolkata, have reported a prevalence of 17.4% which was comparatively higher than the other reports [20]. Chakrabarthi A et al., from Chandigarh documented NTM isolation rate of 3.4% from various clinical specimens [21]. Paramasivan CN et al., from Chennai, South India reported 8.6% [22]. Das BK et al., reported isolation of 8.3% NTM from Delhi and Kasauli [23]. The prevalence of NTM was 5% in this study which was closer to the study conducted by Jesudason MV et al., who reported 3.9% [24] [Table/Fig-11].

Worldwide prevalence of NTM [19].

RegionsBacteria
USAMAC, M.kanasasii
S.E.AmericaM.abscessus
EuropeMAC
N.EuropeM.xenopi
UK, GreeceRGM
AsiaM.fortuitum
E.asiaM.abscessus, M.fortuitum, M.intracellulare
AustraliaMAC
S.australiaRGM (M.abscessus)
AfricaM.abscessus, avium, fortuitum

USA: United States of America; S.E. America: South East America; N.Europe: North Europe; UK: United Kingdom; E.Asia: East Asia; S.Australia: South Australia; MAC: Mycobacterium avium complex; RGM: Rapid grower mycobacteria


Prevalence of NTM-Various studies table [20-22,24-26].

StudyPrevalenceSpecimensCommonest isolate
Karak K et al., [20]17.4%SputumM.fortuitum
Chakrabarthi A et al., [21]3.4%Various specimensM.fortuitum
Paramasivan CN et al., [22]8.6%SputumM.avium/intracellulare
Jesudason MV and Gladstone P [24]3.9%Various specimensM.abscessus, fortuitum
Umrao J et al., [25]29%Various specimensM.abscessus
Jain S et al., [26]9.9%Various specimensM.kanasasii
Current study5%Various specimensM.abscessus

In this study, most of the NTM’s were isolated from pulmonary samples, bronchial wash being the predominant sample (95.5%), in Jesudason MV and Gladstone P study recorded largest yield of NTM from pus and biopsy specimens followed by the respiratory specimens [24]. Umrao J et al., study yielded 79.4% from pulmonary sites and 18.2% from extra pulmonary sites, Shenai S et al., study yielded 81% from pulmonary sites [25,27]. The median age group in this study was 40.5 years while Jesudason MV et al., reported 28 years, Umrao J et al., reported 48 years [24,25]. In this study, proportion of males were higher (60%) compared to females which was similar to Umrao J et al., study (60.4%) while Jesudason MV et al., reported 56% males [24,25].

The common co-morbidities in this study were bronchiectasis, COPD and previous history of TB. Jain S et al. also reported main underlying risk factors as COPD (23.1%) and previous TB (30.8%); Shenai S et al., stated COPD 12%, bronchiectasis (9%), previous pulmonary TB (40%) and Huang HL et al., reported previous pulmonary TB (22.1%) and COPD (19.8%) as common co-morbities [26-28]. The direct smear positivity was 15.2% in this study while it was 20% in Shenai S et al., study and 25.6% in the study by Jain S et al., [27,26]. In this study, the culture positivity was 18.5%. Of these positives, 72.6% were identified as MTBC and 27.4% as NTM. Jain S et al., reported 30.7% culture positivity out of which, MTBC constituted (90.1%) and NTM (9.9%) [26]. In Umrao J et al., study, the culture positivity was 19.6%, of which 29.0% were confirmed as NTM and 71.0% MTBC [25]. RGM prevalence was 4.8% while SGM 0.18% in the present study. RGM are a major cause of pulmonary NTM disease in some regions of Asia. This is in contrast with studies of NTM in other parts of the world [2]. The Netherlands study reported only 3% RGM while United States, 5% [29,30]. The high rates in Asia could be due to environmental exposure, isolation frequency and Ethnic factors [28]. Increase in latitude had a direct affect on NTM isolation rates while RGM isolates exhibited a reverse trend [28].

In this study, M.abscessus was the major isolate (95.6%) while Jesudason MV et al., reported M.chelonae-fortuitum as the predominant isolate (87%) and Maurya AK et al., reported M.fortuitum (27.4%) and M.abscessus (14.4%) as the predominant isolates [24,11]. Present study was in agreement with the previous studies carried out in China, Saudi Arabia and Taiwan in which M. abscessus was the predominant NTM species [28]. The MALDI-TOF role in identification of bacteria is already been proved but the challenge is the identification of fungi (yeast and filamentous fungi) and Mycobacteria which are usually difficult with the conventional methods. Molecular methods have shown great improvement in species identification. In this study MALDI-TOF identified 89% of NTM isolates of which M.abscessus is the predominant isolate which is in accordance with Shetye S et al., study 86.67% [31]. In this study, gene sequencing identified all the 55 isolates. MALDI-TOF and gene sequencing identification agreement was 89% which was close to the study conducted by Shetye S et al., 86.67% [31].

The treatment of NTM infections caused by M. abscessus was most difficult [32,33]. In this study 100% sensitivity was observed for linezolid followed by clarithromycin (95.6%), moxifloxacin and amikacin (93.4%). Jesudason MV et al., showed highest susceptibility to amikacin (99.2%) [24]. Ahmed I et al., study RGM isolates showed maximum response to amikacin and clarithromycin, followed by Linezolid, a variable response to Quinolone (45%-ciprofloxacin, 75%- moxifloxacin) and poor sensitivity to Imipenem (20%) [34].

In this study, all the M. abscessus were characterised as subspecies abscessus. Mycobacterium abscessus complex is subdivided into M. abscessus subsp. abscessus, M.abscessus subsp. massiliense, M.abscessus subsp. Bolletii. Inducible macrolide resistance conferred by erythrosomal ribosomal methyl transferase erm (41) gene was observed in M. abscessus and M. bolletii sub species. When M. fortuitum or M. abscessus subsp abscessus isolate is exposed to macrolide, the erm gene activity is induced the organism appear to be susceptible in vitro but will not respond to the macrolide in vivo. This is termed as ‘cryptic resistance which can be ruled out by incubating with macrolide for atleast 14 days [35]. In this study, M.simiae showed 100% resistance to isoniazid, rifampicin, ethambutol, streptomycin and doxycycline and 100% sensitivity to cotrimoxazole, amikacin, clarithromycin, linezolid, moxifloxacin, levofloxacin and 50% resistance to imipenem. Coolen-Allou N et al., study reported maximum susceptibility to amikacin, moxifloxacin, ciprofloxacin and clarithromycin (96%, 92%, 87% and 100% of tested patients, respectively) and resistance to rifampicin, ethambutol and isoniazid were 85%, 89% and 92% respectively [36]. Both slow and rapid growers in this study were susceptible to linezolid, clarithromycin, amikacin and were resistant to first line TB drugs i.e., isoniazid, rifampicin, ethambutol and streptomycin hence AST of NTM’s should be performed as a routine.

Limitation(s)

Study was done for a short duration and follow-up of the patients was not done in the study. Epidemiology and predictors of NTM infections and environmental distribution of NTM need to be studied.

Conclusion(s)

NTM species level identification is done in only specialised laboratories. NTM species shows variation in clinical spectrum and AST. Hence, there is a need to identify NTM isolates to the species level as misdiagnosis can lead to inappropriate treatment. Molecular assay helps in rapid identification which can lead to targeted therapy and can thus combat antimicrobial resistance. The MALDI-TOF also offers quick results at a low cost and is easy to perform, hence it can be considered as an alternate diagnostic tool for identification.

CSF: Cerebrospinal fluid; CAPD: Continous ambulatory peritoneal dialysis; BMA: Bone marrow aspirate; FNAC: Fine Needle aspiration cytologyCOPD: Chronic obstructive pulmonary disease; SLE: Systemic lupus erythematosis; ILD: Interstitial lung disease; TB: TuberculosisUSA: United States of America; S.E. America: South East America; N.Europe: North Europe; UK: United Kingdom; E.Asia: East Asia; S.Australia: South Australia; MAC: Mycobacterium avium complex; RGM: Rapid grower mycobacteria

References

[1]Griffith DE, Aksamit T, Brown-Elliott BA, Catanzaro A, Daley C, Gordin F, An official ATS/IDSA statement: Diagnosis, treatment, and prevention of non tuberculous mycobacterial diseases Am J Respir Crit Care Med 2007 175(4):367-416.10.1164/rccm.200604-571ST17277290  [Google Scholar]  [CrossRef]  [PubMed]

[2]Marras TK, Daley CL, Epidemiology of human pulmonary infection with non-tuberculous mycobacteria Clin Chest Med 2002 23(3):553-67.10.1016/S0272-5231(02)00019-9  [Google Scholar]  [CrossRef]

[3]Khan K, Wang J, Marras TK, Nontuberculous mycobacterial sensitization in the United States: National trends over three decades Am J Respir Crit Care Med 2007 176(3):306-13.10.1164/rccm.200702-201OC17507546  [Google Scholar]  [CrossRef]  [PubMed]

[4]Billinger ME, Olivier KN, Viboud C, de Oca RM, Steiner C, Holland SM, Nontuberculous mycobacteria-associated lung disease in hospitalized persons, United States, 1998-2005 Emerg Infect Dis 2009 15(10):1562-69.10.3201/eid1510.09019619861046  [Google Scholar]  [CrossRef]  [PubMed]

[5]Horsburgh CR Jr, Epidemiology of disease caused by nontuberculous mycobacteria Semin Respir Infect 1996 11(4):244-51.  [Google Scholar]

[6]Gopinath K, Singh S, Non-tuberculous Mycobacteria in TB endemic countries: Are we neglecting the danger? PLoS Negl Trop Dis 2010 4(4):e61510.1371/journal.pntd.000061520436962  [Google Scholar]  [CrossRef]  [PubMed]

[7]Jani MN, Rodrigues C, Mehta AP, The neglected and often ignored: Nontuberculous Mycobacteria J Glob Infect Dis 2011 3(1):9410.4103/0974-777X.7730521572618  [Google Scholar]  [CrossRef]  [PubMed]

[8]Wallace RJ, Glassroth J, Griffith DE, Olivier KN, Cook JL, Gordin F, Diagnosis and treatment of disease caused by nontuberculous mycobacteria Am J Respir Crit Care Med 197 156(Suppl):S1-25.10.1164/ajrccm.156.2.atsstatement  [Google Scholar]  [CrossRef]

[9]Koneman EW, Allen SD, Janda WM, Color Atlas and Textbook of Diagnostic Microbiology 2006 6th editionPhiladelphia, Pa, USALippincott Williams & Wilkins  [Google Scholar]

[10]Truant JP, Brett WA, Thomas W Jr, Flourescence microscopy of tubercle bacilli stained with auramine and rhodamine Henry Ford Hosp Med Bull 1962 10:287-96.  [Google Scholar]

[11]Maurya AK, Nag VL, Kant S, Kushwaha RA, Kumar M, Mishra V, Evaluation of an immunochromatographic test for discrimination between Mycobacterium tuberculosis complex & non tuberculous mycobacteria in clinical isolates from extra-pulmonary tuberculosis Indian J Med Res 2012 135(6):901-06.  [Google Scholar]

[12]Lee AS, Jelfs P, Sintchenko V, Gilbert GL, Identification of non-tuberculous mycobacteria: Utility of the GenoType Mycobacterium CM/AS assay compared with HPLC and 16SrRNA gene sequencing Journal of Medical Microbiology 2009 58(7):900-04.10.1099/jmm.0.007484-019502366  [Google Scholar]  [CrossRef]  [CrossRef]

[13]Huang TS, Lee CC, Tu HZ, Lee SSJ, Rapid identification of Mycobacteria from positive MGIT broths of primary cultures by MALDIT-TOF mass spectrometry PLOS ONE 2018 13(2):e019229110.1371/journal.pone.019229129394275  [Google Scholar]  [CrossRef]  [PubMed]

[14]Van Soolingen D, Hermans PW, De Haas PE, Soll DR, Van Embden JD, Occurrence and stability of insertion sequences in Mycobacterium tuberculosis complex strains: Evaluation of an insertion sequence-dependent DNA polymorphism as a tool in the epidemiology of tuberculosis J Clin Microbiol 1991 29(11):2578-86.10.1128/JCM.29.11.2578-2586.19911685494  [Google Scholar]  [CrossRef]  [PubMed]

[15]Kim SH, Shin JH, Identification of nontuberculous mycobacteria using multilocous sequence analysis of 16S rRNA, hsp65, and rpoB J Clin Lab Anal 2018 32(1):e2218410.1002/jcla.2218428230286  [Google Scholar]  [CrossRef]  [PubMed]

[16]Susceptibility testing of Mycobacteria, Nocardia and other aerobic Actinomycetes: Approved standard-second edition. CLSI document M24-A2. 2011  [Google Scholar]

[17]Singh AK, Maurya AK, Umrao J, Kant S, Kushwaha RA, Nag VL, Role of genotype((R)) mycobacterium common mycobacteria/additional species assay for rapid differentiation between Mycobacterium tuberculosis complex and different species of Non-Tuberculous Mycobacteria J Lab Physicians 2013 5(2):83-89.10.4103/0974-2727.11984724701099  [Google Scholar]  [CrossRef]  [PubMed]

[18]Hasegawa N, Miura T, Ishii K, Yamaguchi K, Lindner TH, Merritt S, New simple and rapid test for culture confirmation of Mycobacterium tuberculosis complex: A multicenter study J Clin Microbiol 2002 40(3):908-12.0.1128/JCM.40.3.908-912.200211880414  [Google Scholar]  [CrossRef]  [PubMed]

[19]Honda JR, Virdi R, Chan ED, Global environmental non tuberculous mycobacteria and their contemporaneous man made and natural niches Frontiers in Microbiology 2018 9:202910.3389/fmicb.2018.0202930214436  [Google Scholar]  [CrossRef]  [PubMed]

[20]Karak K, Bhattacharyya S, Majumdar S, De PK, Pulmonary infections caused by mycobacteria other than M.tuberculosis inand around Calcutta Indian J Pathol Microbiol 1996 39:131-34.  [Google Scholar]

[21]Chakrabarthi A, Sharma M, Dubey ML, Isolation rates of different mycobacterial species from Chandigarh Indian J Med Res 1990 91:111-14.  [Google Scholar]

[22]Paramasivan CN, Govindan D, Prabhakar R, Somsundaran PR, Subbammal S, Tripathy SP, Species level identification of Non Tuberculous Mycobacteria from South Indian BCG trial area during 1981 Tubercle 1985 66:09-15.10.1016/0041-3879(85)90048-0  [Google Scholar]  [CrossRef]

[23]Das BK, Sharma VK, Bhanu LN, Saxena SN, Bhardwaj BK, Characterisation of mycobacterial strains from clinical specimens Indian J Pathol Micobiol 1982 25:19  [Google Scholar]

[24]Jesudason MV, Gladstone P, Non Tuberculous Mycobacteria isolated from clinical specimens at a tertiary care hospital in South India Indian J Med Microbi 2005 23(3):172-75.10.1016/S0255-0857(21)02588-3  [Google Scholar]  [CrossRef]

[25]Umrao J, Singh D, Zia A, Saxena S, Sarsaiya S, Singh S, Prevalence and species spectrum of both pulmonary and extrapulmonary nontuberculous mycobacteria isolates at a tertiary care center Int J Mycobacteriol 2016 5(3):288-93.10.1016/j.ijmyco.2016.06.00827847012  [Google Scholar]  [CrossRef]  [PubMed]

[26]Jain S, Sankar MM, Sharma N, Singh S, High prevalence of non-tuberculous mycobacterial disease among non-HIV infected individuals in a TB endemic country- Experience from a tertiary center in Delhi, India Pathog Glob Health 2014 108(2):118-22.10.1179/2047773214Y.000000013324649868  [Google Scholar]  [CrossRef]  [PubMed]

[27]Shenai S, Rodrigues C, Mehta A, Time to identify and define non-tuberculous mycobacteria in a tuberculosis-endemic region Int J Tuberc Lung Dis 2010 14(8):1001-08.  [Google Scholar]

[28]Huang HL, Cheng M, Lu P, Shu C, Wang J, Wang JT, Epidemiology and predictors of NTM pulmonary infection in Taiwan-A retrospective, five-year multicenter study Scientific Reports 2017 7:1630010.1038/s41598-017-16559-z29176633  [Google Scholar]  [CrossRef]  [PubMed]

[29]Van Ingen J, Bendien SA, de Lange WC, Hoefsloot W, Dekhuijzen PN, Boeree MJ, Clinical relevance of non-tuberculous mycobacteria isolated in the Nijmgen-Arnhem region, the Netherlands Thorax 2009 64:502-06.10.1136/thx.2008.11095719213773  [Google Scholar]  [CrossRef]  [PubMed]

[30]O’ Brian RJ, Geiter LJ, Snider DE, The epidemiology of NonTuberculous Mycobacteria disease in the United States: Results from a national survey Am Rev Respir Dis 1987 135:1007-14.  [Google Scholar]

[31]Shetye S, Chheda P, Walke D, Matkar S, Mukundan U, Malditof MS for rapid identification of Mycobacteria species in liquid culture media Journal of Bacteriology and Mycology 2018 6(2):105-07.10.15406/jbmoa.2018.06.00185  [Google Scholar]  [CrossRef]

[32]Falkinham JO, Epidemiology of infection by nontuberculous mycobacteria Clin Microbiol Rev 1996 9(2):177-215.10.1128/CMR.9.2.1778964035  [Google Scholar]  [CrossRef]  [PubMed]

[33]Jarzembowski JA, Young MB, Nontuberculous mycobacterial infections Arch Pathol Lab Med 2008 132(8):1333-41.10.5858/2008-132-1333-NMI18684037  [Google Scholar]  [CrossRef]  [PubMed]

[34]Ahmed I, Jabeen K, Hasan R, Identification of non-tuberculous mycobacteria isolated from clinical specimens at a tertiary carehospital: A cross-sectional study BMC Infectious Diseases 2013 13:49310.1186/1471-2334-13-49324148198  [Google Scholar]  [CrossRef]  [PubMed]

[35]Nash KA, A novel gene erm (41) confers inducible macrolides resistance to clinical isolates of M.abscessus but is absent from M. chelonae Antimicrob. Agents Chemother 2009 53:1367-76.10.1128/AAC.01275-0819171799  [Google Scholar]  [CrossRef]  [PubMed]

[36]Coolen-Allou N, Touron T, Belmonte O, Gazaille V, Andre M, Allyn J, Clinical, radiological, and microbiological characteristics of mycobacterium simiae. Infection in 97 patients Antimicrob Agents Chemother 2018 62(7):e00395-1830.10.1128/AAC.00395-1829760130  [Google Scholar]  [CrossRef]  [PubMed]