Skip to main content

Current trends and intricacies in the management of HIV-associated pulmonary tuberculosis

Abstract

Human immunodeficiency virus (HIV) epidemic has undoubtedly increased the incidence of tuberculosis (TB) globally, posing a formidable global health challenge affecting 1.2 million cases. Pulmonary TB assumes utmost significance in the programmatic perspective as it is readily transmissible as well as easily diagnosable. HIV complicates every aspect of pulmonary tuberculosis from diagnosis to treatment, demanding a different approach to effectively tackle both the diseases. In order to control these converging epidemics, it is important to diagnose early, initiate appropriate therapy for both infections, prevent transmission and administer preventive therapy. Liquid culture methods and nucleic acid amplification tests for TB confirmation have replaced conventional solid media, enabling quicker and simultaneous detection of mycobacterium and its drug sensitivity profile Unique problems posed by the syndemic include Acquired rifampicin resistance, drug–drug interactions, malabsorption of drugs and immune reconstitution inflammatory syndrome or paradoxical reaction that complicate dual and concomitant therapy. While the antiretroviral therapy armamentarium is constantly reinforced by discovery of newer and safer drugs every year, only a few drugs for anti tuberculosis treatment have successfully emerged. These include bedaquiline, delamanid and pretomanid which have entered phase III B trials and are also available through conditional access national programmes. The current guidelines by WHO to start Antiretroviral therapy irrespective of CD4+ cell count based on benefits cited by recent trials could go a long way in preventing various complications caused by the deadly duo. This review provides a consolidated gist of the advancements, concepts and updates that have emerged in the management of HIV-associated pulmonary TB for maximizing efficacy, offering latest solutions for tackling drug–drug interactions and remedial measures for immune reconstitution inflammatory syndrome.

Background

HIV and tuberculosis (TB) have always been faithful comrades facilitating each other in spreading across the globe. According to recent World Health Organization (WHO) estimates, 9.6 million cases of tuberculosis (TB) occurred all over the world with 12 % (1.2 million) being co-infected with human immunodeficiency virus (HIV); with 37 % of these new TB cases going undiagnosed [1]. In the year 2014, an estimated 1.5 million had died due to TB with a quarter of them caused by HIV. Pulmonary TB is the commonest form of TB even in HIV even though it is more frequently associated with dissemination locally and systemically, when the two infections coexist. Therefore, early diagnosis and prevention of pulmonary TB, with suitable chemoprophylaxis have become key components towards achieving the end TB strategy [1].

Despite wide spread scale up of antiretroviral therapy (ART), only about one-third (392,000 or 77 % of the notified TB cases known to be HIV infected) were put on antiretroviral therapy (ART). The aim should be to combine the ideal anti-tuberculosis treatment (ATT) with mutually compatible highly active antiretroviral therapy (HAART) combinations to maximize efficacy, avoiding drug–drug interaction. When ART is initiated in HIV infected subjects, a major complication that could arise is the onset of ART related immune reconstitution inflammatory syndrome or IRIS, requiring, early detection and prompt treatment. These important aspects pertaining to diagnosis and treatment of this deadly duo is being elaborated in the following paragraphs.

Diagnosis of tuberculosis in HIV infected

Latent TB infection

HIV infection is one important factor for progression to TB disease, mandating meticulous screening and treatment for latent M. tuberculosis infection especially in TB prevalent countries.

Diagnosed of latent TB infection

1. Tuberculin skin test (TST) Targeted tuberculin testing for LTBI forms a strategic component of TB control identifying high risk population prone for developing TB [2]. Studies have shown that TST-positive patients benefit more from IPT than those who are TST negative [3]. Anergy, improper cold chain maintenance can give rise to false negative results in HIV [2]. Considering these limitations in resource limited set-ups, World Health Organization’s Guidelines Group strongly recommends IPT irrespective of TST for people living with HIV [3].

2. TB MPB-64 skin patch test MPB-64 is a specific mycobacterial antigen secreted by M. tuberculosis, M. bovis and some strains of M. bovis used in this patch. This test is simple, non-invasive, does not require a laboratory or highly skilled personnel, unaffected by anergy in HIV-infected individuals and becomes positive in 3–4 days after patch application on skin, and induration on skin lasts for a week. In a study in Manila, Philippines the sensitivity of the transdermal Patch was 87.8 %, with an efficacy of 92.9 % and a specificity of 100 % [4].

3. IFN-γ release assays (IGRA’s) These in vitro blood assays based on IFN production from sensitized T cells TB antigens like early secretory antigenic target 6 (ESAT 6) and culture filtrate protein 10 (CFP 10), are commercially available as QuantiFERON-TB QFT), Enhanced QuantiFERON-TB Gold assay and ELISPOT format, T-SPOT-TB assay] [5].

A study of asymptomatic adults from South Africa, a country with a high prevalence of co-infection found that the rates of positive ELISPOT and ELISA results did not vary significantly by HIV status compared to TST [6]. Due to the requirement of a good laboratory infrastructure and costs, the WHO’s Guidelines Group does not recommend IGRA to screen people living with HIV for eligibility to receive IPT, as IGRA does not spell out who will benefit most from IPT [3].

4. Blood biomarkers Studies are underway using host RNA gene expression from whole blood that allows for identification of prospective high risk individuals who can potentially progress to active tuberculosis disease [7].

Diagnosis of active TB disease

High clinical suspicion is required in diagnosing early TB disease especially in the context of advanced HIV due to paucity of classical symptoms. WHO guidelines on systematic screening for active pulmonary TB using syndromic evaluation, with active case finding serves a dual purpose, channelizing individuals for either chemoprophylaxis or for prompt initiation of treatment [810]. This simplified syndromic questionnaire of three symptoms namely cough, fever and night sweats had been effectively used to diagnose or rule out TB in a study from South East Asia [11].

a. Imaging techniques

Adding a chest X-ray or a CT scan to symptom screening not only increases the detection rate but the cost as well. The spectrum of radiographic manifestation of pulmonary TB is dependent on the relative level of HIV-related immunodeficiency and varies from normal chest X-ray (CXR) to full blown miliary TB [12, 13] (Fig. 1). A study to evaluate the utility of initial CXR in the diagnostic algorithm for symptomatic HIV-infected patients with negative sputum smears, found that basing a diagnosis of pulmonary TB on initial CXR leads to over diagnosis of TB. CXR had a sensitivity and specificity of 72 and 57 %, respectively, with positive predictive value of 21 % and negative predictive value of 93 % in diagnosing PTB [14]. In addition routine abdominal ultrasonography, followed by abdominal CT scan in inconclusive cases, significantly increased the detection of abdominal TB in patients with advanced immunodeficiency [15].

Fig. 1
figure 1

Varying manifestations of TB in HIV. a Apparently normal CXR. b Caeseating mediastinal adenitis typical of TB. c Miliary TB

b) Microbiology

1. Sputum Smear microscopy The most frequent and conventional method of pulmonary TB detection in HIV co-infected persons involves examination of at least two sputum specimens, including one early-morning specimen, for acid-fast bacilli (AFB) [16]. Though the sensitivity of sputum microscopy in HIV infection is low (43–51 %), it has the advantage of being inexpensive, relatively rapid to perform, and specific in most settings. Fluorescence microscopy, light emitting diode microscopy, alternative specimen processing methods, such as concentration, bleach sedimentation and same-day sputum collection (front loading) strategies increase the sensitivity of sputum microscopy from 59 to 93 % in detecting pulmonary TB in HIV co-infected persons [1719].

2. Culture of Mycobacterium tuberculosis (M.Tb) Mycobacterial culture still remains the gold standard for TB diagnosis. The inherent weaknesses of smear microscopy to determine viability, drug resistance and species identification has made culture test indispensable. HIV co-infection does not affect the yield of mycobacterial culture, but its growth on traditional solid medium like the Middlebrooks or Lowenstein-Jenson medium is rather slow requiring up to 6–8 weeks. Repeated sputum examinations for culture up to three specimens increases the yield by 10–17 % [16].

With increased sensitivity and reduced delays, automated liquid culture systems have replaced solid cultures, increasing case detection by 10 % with quicker TB diagnosis (detecting mycobacterial growth within 1–2 weeks), resulting in prompt treatment initiation. Methods currently in practice include Mycobacteria Growth Indicator Tube (MGIT) 960 [Becton–Dickinson Diagnostic Instruments Systems] using fluorescent sensors, MB/BacT system (Organon Teknika) using colorimetric sensors, ESP culture system II (Difco Laboratories, USA) using pressure sensors, or redox reagents, such as Alamar blue [2022]. Among HIV-TB co-infected patients, MGIT was found to be more sensitive than culture and microscopy [23]. Other available techniques include bacteriophage based assays (FASTPlaqueTB), Luciferase reporter phage based test and Microscopic observation drug susceptibility (MODS) assays. The latter is a low cost non-commercial method for early detection of micro colonies and drug resistance [24, 25].

3. Molecular techniques Nucleic acid amplification tests (NAAT), are designed to amplify nucleic acid regions specific for M. tuberculosis complex and can be used directly on the clinical samples. NAAT yield rapid results, are highly specific with improved sensitivity compared to smear microscopy. These techniques can detect specific mutations, thus providing information on drug sensitivity as well.

Line probe assays (LPA), endorsed by the WHO in 2008 for molecular detection of drug resistance uses a PCR/hybridization technique to distinguish members of the M.Tb complex and simultaneously identifies drug-resistant strains by detecting the most common single nucleotide polymorphisms associated with resistance [25]. LPAs are highly sensitive (≥97 %) and specific (≥99 %) for the detection of rifampicin resistance, alone or in combination with isoniazid (sensitivity ≥90 %; specificity ≥99 %), on isolates of M. tuberculosis but it use is restricted to smear-positive sputum specimens only [26].

Xpert-MTB rif (GeneXpert) has currently taken the center stage, providing results within 2 h, with an increase in case detection rate of 45 % compared to smear microscopy and can be used in smear negative patients also [27]. It is a TB-specific automated, cartridge-based nucleic acid amplification assay, endorsed by WHO, for the rapid diagnosis of TB as well as early detection of rifampicin resistance among HIV-infected individuals among presumptive TB patients [27]. Xpert—Ultra is an improved version that has been equated to a liquid culture in its ability to detect Tuberculosis with an improvised specificity to detect MT. TB as well as rifampicin resistance even among smear negative patients with HIV where the conventional Xpert—MTB rif has a lesser yield [28]. Loop-mediated isothermal amplification and Fluorescence in situ hybridization using peptide nucleic acid probes, are other rapid and simplified molecular techniques using NAAT platform used to diagnose Mycobacterial infection, with high sensitivity and specificity [29, 30].

4. Serological diagnosis of tuberculosis Serological tests have no role to play for detecting active TB disease either pulmonary or extra pulmonary TB, and have been banned by WHO [31].

Other diagnostic techniques

Capture ELISA test is available to detect lipoarabinomannan (LAM) in urine specimens. Among hospitalized HIV-infected patients with suspected TB in Zimbabwe, the sensitivity of LAM was found to be significantly higher than that of Sputum smear microscopy, especially in advanced HIV with lower CD4 counts [32]. Bedside LAM-guided initiation of ATT for HIV-infected hospitalized patients with suspected TB was associated with a reduced 8-week mortality rate in a pragmatic, randomized, multi-centric trial in Africa [33]. WHO has now recommended the detection of LAM, in urine specimens in immunosuppressed HIV-infected patients as an adjunctive means of rapidly diagnosing active TB disease [34].

In-spite of all these newer TB diagnostic technologies, including Urinary LAM and Gene Xpert, clinical suspicion and thorough screening of TB in HIV still plays a vital role.

Preventive therapy for TB in HIV infection

As the risk of TB developing in HIV infected individuals is 5–10 % every year, current WHO guidelines recommend screening of all HIV-infected individuals for TB (intensified case finding), and if found to be un-infected, receive isoniazid preventive therapy (IPT) for a 6 months, irrespective of TST status [35, 36].

Various randomized controlled clinical trials on different durations and multiple drug regimens have yielded varying results (Table 1).

Table 1 Summary of characteristics and conclusion of randomised controlled clinical trials using various regimens for TB prophylaxis in HIV

A Cochrane systematic review of 12 trials, published in 2010 among 8578 patients demonstrated that IPT reduced the risk of active TB by 64 % among TST positive HIV-infected participants, but only by 14 % among TST negative individuals [37]. The key message from the review was that efficacy was similar regardless of drug type, regimen or duration compared to INH monotherapy [37]. CDC also recommended a 12-weekly regimen of INH and rifapentine given as DOT as an equal alternative to 9-months of daily self-supervised INH for treating LTBI in otherwise healthy patients aged ≥12 years and advised against use of rifampicin and pyrazinamide for 2 months as prophylaxis [38, 39]. Despite evidences in favour of preventive therapy, adoption of TB preventive therapy in clinical practice has been pretty slow, primarily due to the difficulty in ruling out active TB in HIV infection, apart from reluctance on part of policy makers and program implementers. Getahun et al. [40] suggested IPT be taken over by the HIV intervention programme for an effective implementation after thorough screening for TB.

Patient selection for IPT

In 2011, the WHO released simplified guidelines for IPT, using the clinical algorithm of any cough, night sweats, weight loss and/or fever. Similarly, house hold contacts of sputum smear positive pulmonary TB cases, especially children, are a good entry point for screening for TB and delivery of IPT [36]. A community wide survey among South African mine workers found that majority (approximately 80 %) were eligible for IPT [41]. Due to low specificity of symptom screening, C-reactive protein (CRP) that has a higher specificity for active TB has been incorporated into the diagnostic algorithm. A point-of-care CRP could facilitate rapid initiation of IPT in HIV patients without active TB [42, 43].

IPT with antiretroviral therapy (ART)

Mortality within the first 6 months after initiating ART has been attributed to TB in most resource-limited settings. Protection against TB is further optimized when IPT is combined with ART. Synergistic protection, with greater than 50 % reduction in TB rates, was found in patients who received both IPT and ART than protection afforded by either treatment alone [44, 45]. However, empirical TB treatment in situations where it is difficult to diagnose subclinical TB like advanced HIV disease may not be of much benefit in reducing mortality [46].

Cost-effectiveness of IPT

A randomized trial of HIV infected adults from India showed that IPT, 6EH increased life expectancy by 0.8 months incurring $100 per individual while 36 months of INH extended life expectancy by 0.2 months with an additional per person cost of $55 [47]. In addition, antepartum IPT for HIV-infected women, irrespective of CD4 count and TST status, was shown to have greater health benefits and more cost-effective than no IPT or TST-driven IPT in a study from India [48].

Brazil demonstrated that training health care workers to screen HIV-infected adults with positive TST and providing IPT to those with latent TB infection was cost-effective relative to the Brazilian GDP per capita [49]. Similarly, the trial from United States using 12-dose of weekly rifapentine plus isoniazid administered as directly observed treatment, also proved to be a cost-effective alternative to 9 months of daily self-administered isoniazid [50]. A decision-analysis model to study the cost-effectiveness of IPT in Botswana revealed that treating PLHIV who have positive TST with 36-month IPT was more cost effective for reducing both TB and death compared with providing IPT without a TST, providing only 6-month IPT, or expanding ART eligibility without IPT [51].

Drug resistance and IPT

A systematic review, assessing the effect of IPT on the risk of INH resistant TB, reported that IPT increases the risk of INH resistance by 1.45 times, whilst it was not significant (Relative risk 1.45; 95 % confidence interval 0.85–2.47) [52]. Though the study had a small sample size and analysis was restricted by incomplete testing of isolates, the relative risk showed a minimal increase in INH resistant TB. However, clinical trials of IPT in HIV-infected patients in Botswana, India and South Africa with more defined cohorts did not show any increased risk of INH resistance amongst patients given IPT [5356].

Summary

Latent TB is still a difficult diagnosis to make in the wake of HIV coinfection. Advances in molecular technology have partially replaced conventional cultures helping in prompt diagnosis of active TB yielding sensitivity results at the earliest. TB should be meticulously screened and INH preventive therapy for 6 months or any other equivalent regimen should be started for TB-free HIV-infected individuals irrespective of Tuberculin Skin Testing. There is a need to update the knowledge and awareness of INH Preventive Therapy among health care workers and policy makers for rapid implementation and reap the benefits as it can be easily coupled and monitored with ART initiation.

Treatment for HIV-associated pulmonary TB

ATT drugs to be used

Management of pulmonary TB in HIV demands meticulous monitoring as it is complicated by smear negativity and atypical presentation, emergence of drug resistance, Immune reconstitution inflammatory syndrome, drug–drug interaction and increased pill burden [57]. The Standards of TB care in India (STCI) recommend a four drug regimen consisting of Isoniazid (INH), rifampicin, pyrazinamide and ethambutol given during in the intensive phase of 2 months followed by INH and rifampicin with/without ethambutol (optional) in the next 4 months for treatment naive patients along with high dose pyridoxine and cotrimaxazole in HIV-TB coinfection [58]. HIV infection favours mycobacteremia and tissue invasion resulting in abundance of intracellular and intermittently dividing bacilli, making rifampicin indispensable in HIV associated TB [59]. This is despite therapeutic interactions with other concomitant medications notably HAART. Non-rifampicin regimens in HIV have been associated with inferior outcomes coupled with longer duration of therapy [60, 61]. The addition of quinolones in the intensive phase does not increase cure rate any further [62]. Interestingly, the sputum conversion rate is faster in HIV-PTB coinfection with effective ATT [63]. Studies comparing TB treatment outcomes between HIV infected and uninfected individuals have obviously shown better results in the latter group [64].

Empirical ATT even in advanced HIV has no definitive role to play. The A5274 study clearly demonstrated that even in a high TB burden population of 1368 participants with advanced HIV (CD4 < 50 cells/mm3), there was no benefit of empiric TB therapy over IPT when mortality rates at 48 weeks was compared [65].

Duration of TB therapy

TB treatment duration is not influenced or confounded by HIV infection currently being 6 months for Pulmonary and extended in severe forms of extra pulmonary TB like bone and neurological TB. Centre for Disease Control, Atlanta recommends extension of ATT beyond 6 months in HIV-coinfected pulmonary TB patients in specific instances like delayed sputum conversion or poor clinical improvement with/without evidence of dissemination, low CD4 count at nadir and presence of cavitation [66]. Swaminathan et al. [67] compared a 6 months standard intermittent therapy with a 9 months regimen (an extended continuation phase of 3 months) in the pre-HAART era among HIV infected pulmonary TB patients, and found that the favourable outcomes, failures at the end of treatment, adverse events and mortality (during treatment and follow-up of 24 months) to TB were similar. However, there was a significant reduction in bacteriologically confirmed recurrences when a longer regimen was used. The meta-analysis by Menzies et al. [68] demonstrated that extended treatment beyond 6 months (to 8 months) did not appreciably increase the efficacy further, justifying the current duration of 6 months. The same meta-analysis found only a single study having a higher risk of relapse with extensive cavitary disease demanding 8 months treatment. Hence, the authors, concluded that high risk identification would not be easy in the public health perspective that makes generalized extension of regimen in all cases unnecessary [68].

Dosing schedule

The legacy of intermittent regimens in India dates back to 1964 when Tuberculosis Research Centre used streptomycin and INH administered twice weekly, yielding a cure rate of 94 % and a relapse rate of 8 % [69]. The popular attributes of intermittent regimens were operational feasibility, lesser cost, fewer side effects and ease of supervision. The scientific basis for intermittent usage of ATT is based on post antibiotic effect of ATT or lag period [70]. Mycobacterium undergoes a lag phase in its growth pattern due to the influence of the post antibiotic effect, when bacilli fail to grow for a period of a few days even after removal of the exposed drug i.e. ATT [70]. However, newer hypotheses challenge this approach. It has been postulated that the ratio of peak concentration to minimum inhibitory concentration better correlates with the post antibiotic effect and contributes to suppression of resistance than lag phase. The authors further hypothesized that asynchrony between the metabolism of these intermittently growing bacilli and the day of drug administration could facilitate emergence of drug resistant mutants [71].

Swaminathan et al. [67] study showed a higher incidence of acquired rifampicin resistance (ARR) among failures, using thrice weekly rifampicin in HIV-PTB, which was unaltered by the duration of the regimen used. Interestingly, the meta-analysis by Menzies et al. [68] did not find intermittent ATT dosing to be a cause of poorer outcomes unless the frequency of dosing went down to below thrice a week, but had only 2 % of patient in the meta-analysis being co-infected. However, the meta-analysis by Faiz khan et al. showed that the use of intermittent regimen in HIV, especially in the intensive phase, increased the risk of failures, relapses and emergence of drug resistance which further intensifies in the wake of baseline H-resistance [72]. The study by Li et al. [73] showed that a daily intensive phase of ATT followed by an intermittent continuation phase served as a useful alternative to a fully daily regimen. The updated review by Faiz khan et al. [74] recommended use of a daily regimen throughout and extension of TB regimen to 8 months, but suggested further evaluation of data to make categorical conclusions. Recently, there has been a paradigm shift in approach to TB treatment among HIV in India, with the STCI recommending daily regimen for all HIV co-infected patients with ethambutol reinforced in the continuation phase (2EHRZ7/4HRE7) [58]. The only randomized trial of head to head comparison of daily vs thrice weekly ATT regimen in HIV patients with newly diagnosed rifampicin sensitive pulmonary TB, started on timely ART, showed a higher cure rate with daily compared to thrice weekly ATT (90 vs 76 %) but at the expense of higher incidence of hepatoxicity [75].

Acquired rifamycin resistance (ARR)—a unique phenomenon complicating HIV-associated pulmonary TB

The greater percentage of persistors and bacillary mutants in an immunocompromised environment (of HIV coinfection) facilitates and favours emergence of drug resistance to ATT notably rifampicin [76]. Acquired rifamycin resistance (ARR) is the emergence of resistance (defined as MIC > 128 µg/ml) to rifamycin among patients whose pretreatment isolates were sensitive. ARR is a rarity in HIV seronegative individuals with PTB. In a cohort of 1435 HIV sero-negative patients with drug susceptible PTB enrolled in various trials at the Tuberculosis Research Centre, Chennai, only 4 patients developed rifampicin resistance irrespective of the dosing schedule [77].

On the contrary, ARR in the HIV co-infected had been reported with all types of rifamycin regimens used intermittently, be it once weekly rifapentine [78], twice weekly rifabutin [79, 80] or thrice weekly rifampicin [67]. Advanced stage of HIV, absence of HAART, extensive and/or disseminated TB, initial H-resistance, and suboptimal drug concentrations due to malabsorption are contributing factors for ARR. Increased tissue bacillary load in HIV, coupled with defective clearance attributed to subdued immune apparatus leads to selection of genomic mutants resistant to rifampicin, which is much more pronounced in the face of baseline INH resistance [8083]. The study by Narendran et al. [84] showed that HIV and INH resistance at baseline were significant risk factors associated with ARR and HAART only reduced the frequency but did not offset the trend. The NCT 00933790 trial evaluating the incidence of failures and ARR among co-infected with different dosing schedules of ATT and timely HAART showed that ARR occurred only with intermittent dosing [75].

Recurrences

TB recurrences can be either due to endogenous re-activation or exogenous re-infection, the relative proportions depending on the background incidence of TB, level of immune suppression, length of rifampicin-containing ATT and adherence to treatment [79, 85]. The proportion of recurrences due to re-infection is more frequent in HIV positive individuals especially in countries with a higher TB burden than HIV-seronegative individuals with TB who have a true relapse [86].

Persistence of smear positivity in HIV—are they true failures?

The decision making based on sputum smears alone can often be misleading and one such situation is the phenomenon of persistent sputum positivity despite regular treatment. A practical approach requires segregation of cases into patients with clinical improvement but smear positive or those who are truly failing therapy with persistence/recrudescence of clinical symptoms or both currently the national programme in many countries has scaled up to detect early drug resistance using line probe assay and Xpert-MTB Rif at the start of TB treatment that can simplify the problem [87]. Causes of true failures in HIV associated TB include emergence of ATT drug resistance, virological failure to ART, immunological discordance (lower CD4 with undetectable viral load) and malabsorption of drugs leading to cryptic non-adherence [79, 80, 82]. All of them may influence smear and culture conversion with the risk of subsequent failures or recurrences and transmission. Mal-absorption of ATT drugs may cause persistence of symptoms with delayed clearance of organisms, providing a favourable nidus for emergence of drug resistant mutants [8183].

The second group of apparent failures comprises of patients with clinical improvement but still have sputum smears positive for AFB or have apparent deterioration. The differential diagnosis of the latter includes IRIS [88], transient resistance or “bacillary escape” [89] and non-tuberculous mycobacteria mimicking TB [90]. Atypical mycobacteria including M. avium intracellulare, M. kansasi, M. fortitium have been isolated more frequently in advanced stages of HIV disease when the CD4 is <100 cells/mm3 [90]. Nocardiosis may be misdiagnosed as TB due to its close resemblance to the latter both microscopically as well as clinically [91].

Shortening TB treatment—a myth or a reality?

Whether a shorter TB regimen can work in the face of HIV coinfection still remains a mystery. The RIFAQUIN trial tested a combination of moxifloxacin replacing Isoniazid in the intensive phase along with rifampicin, pyrazinamide and ethambutol given daily in the 2 months intensive phase followed by high dose rifapentine (900/1200) for 2 months twice weekly or 4 months once weekly with moxifloxacin in the continuation phase and compared to a standard 6 months. The study failed to demonstrate non-inferiority of the 4 months regimen over the standard 6 months regimen. However, the 6 months regimen administered once weekly during the continuation phase showed comparable efficacy with better compliance and feasibility of implementation [92]. An important limitation in this study was that it enrolled only a quarter of HIV patients, with a relatively higher level of CD4 (above 200 cells/mm3) that limited its generalisability to advanced stages of HIV. The OFLOTUB study, that used gatifloxacin replacing ethambutol, also could not prove the 4 months regimen to be non-inferior to the 6 months regimen. These findings were more evident and obvious in the South African site which had half of their recruited patients being co-infected with HIV [93]. A regimen of PA_824 along with moxifloxacin and pyrazinamide, the STAND trial was being evaluated as a non-rifampicin, non-INH alternative that could prove useful in drug sensitive and MDR-TB, without significant drug interaction with ART. The study would resume enrolment shortly after being suspended temporarily [94]. A recent model had been developed that could predict efficacy of new regimens of varying durations, taking into account culture positive rate at 2 months and could spell out the recurrence rates to help investigators formulate shorter regimens with better efficacy [95].

Summary

A standard four drug rifampicin containing regimen given daily for 6 months is still the ideal regimen for pulmonary TB in HIV despite frantic efforts globally to shorten TB treatment. Ethambutol can be added in the continuation phase. Empirical Antituberculosis therapy has no role to play. Acquired rifamycin resistance is a unique complication in HIV-TB. Patients failing therapy should not only be evaluate for ATT resistance but also efforts taken to rule out virological failure, malabsorption and screened simultaneously for non-tuberculous mycobacteria.

Multidrug-resistant (MDR) and extensively drug-resistant (XDR) tuberculosis

Globally in 2014, an estimated 480,000 people developed multidrug-resistant TB (MDR-TB) with an estimated 39.5 % deaths and 9.7 % harboring extensively drug resistant TB (XDR-TB) [1]. India, China and Russia contribute about 60 % of global burden. While MDR-TB appears not to cause infection or disease more readily than drug susceptible TB in the HIV infected population, delayed diagnosis, inadequate initial treatment, and prolonged infectiousness contribute to increased attack rates among contacts, leading to high case fatality rates among patients [96].

Treatment for drug resistant-TB consists of at least 4–5 effective drugs. This includes a fluoroquinolone, a second line injectable agent (capreomycin, kanamycin or amikacin) and at least 2 agents from the remaining second line anti-TB drug classes [cycloserine, thionamides (ethionamide or prothionamide), linezolid, clofazimine] along with add on drugs like pyrazinamide, ethambutol, high dose INH, bedaquiline, delamanid, Amoxycillin clavulanate, para-amino salicylic acid selected preferentially in the order described above [97]. The intensive phase can be up to 8 months with a continuation phase without injectable up to 20 months depending on the therapeutic response. WHO had also come out with a shortened 9–12 month regimen based on the findings conducted by the International Union against Tuberculosis and Lung Diseases and Medical Research Council, United Kingdom, that consisted of moxifloxacin, clofazimine, prothionamide and pyrazinamide for 9 months reinforced with kanamycin and INH high dose in the first 4–5 months of intensive phase. This regimen, however, is approved for use in second line ATT naïve patients (<1 month of treatment) with DST pattern showing sensitivity to both quinolone and second line aminoglycoside at baseline, without extra-pulmonary or disseminated form of TB or pregnancy complicating TB [98].

Evidence for this regimen originated from the Bangladesh observational cohort study by Van Deun among MDR-TB patients which showed a relapse free cure rate of 84.5 % among 515 patients, but in a virtually HIV-free population [99]. The same regimen, tested in the francophone African countries, among 408 patients (that included HIV positive −22 %) showed a relapse free survival rate of 82.1 %. Although treatment success rates did not differ by HIV status among those who survived, the death rate was higher among HIV co-infected 18 % died, compared to 5 % in HIV-seronegative patients [100].

Prognosis of MDR-TB in HIV continues to be grave with a death rate of over 50 % once again highlighting the ardent need to reconstitute the fallen immunity by timely ART initiation [101]. Two additional factors which hinder TB control among drug resistant cases include nosocomial transmission and increased incidence of drug toxicity with frequent drug interruptions that could worsen the prognosis [102, 103]. Both the conditions can lead to amplification of drug resistance and need to be addressed urgently. Stringent airborne infection control measures need to be in place as an effective strategy and overcrowding in hospitals minimized [102]. One study, surprisingly, reported a toxicity of only 7 % among 2000 patients in a study in South Africa elucidating the fact that it is comparable to HIV seronegative group [104].

XDR-TB or extensively drug resistant defined as MDR-TB (resistance to INH and rifampicin) plus resistance to any fluoroquinolone and one of the second line anti-TB injectable agents (kanamycin, amikacin or capreomycin) constitutes a formidable medical challenge. Although the absence of rifampicin brings down the occurrence of drug–drug interactions significantly, therapy for XDR-TB is confounded by other adverse reactions like QTC prolongation, anemia, psychiatric effects, nephrotoxicity and gastrointestinal intolerance in addition to increased pill burden [103]. Treatment options are extremely limited and challenging with higher frequencies of adverse events and death [96]. In a recent study on long term follow-up of XDR-TB patients from South Africa, independent predictors of probability of net sputum culture conversion were no previous history of multidrug-resistant tuberculosis (p = 0.0007), use of clofazamine (p = 0.0069) with survival depending on culture conversion (p < 0.0001), treatment with clofazamine (p = 0.021) and ART in HIV (p = 0.003) [105].

Newer TB drugs in the pipeline

Bedaquiline (BDQ), one of the newer drugs conditionally approved by the FDA recently, has been a useful adjunct in drug resistant-TB therapy, especially when choice is limited. It is a diarylquinoline derivative that inhibits the mycobacterial ATP synthetase [106]. It inhibits both actively replicating and non-replicating mycobacteria with no cross resistance to any of the first line drugs and quinolones. It takes 3–5 days for perception of its bactericidal effect and has an extremely long half-life of 4–5 months and has to be taken along with food. It is administered 400 mg daily for the first 2 weeks followed by 200 mg thrice weekly up to a total of 24 weeks. CYP3A4 is the major cytochrome isoenzyme that metabolizes bedaquiline to M2, that is mainly removed in the stools, with only 1–4 % excreted in urine [107]. Drug-drug interactions occur with CYP3A4 inducers (e.g., rifampicin reduced bedaquiline exposure by approximately 50 %, obviating co-administration). The most frequent suspected adverse reactions (>20.0 % of patients) during treatment with bedaquiline in the controlled trials (C208 stages 1 and 2) were nausea (35.3 %), arthralgia (29.4 %), headache (23.5 %), hyperuricemia (22.5 %), and vomiting (20.6 %) [106]. The magnitude of QTcF prolongation, one of the potential side effects of BDQ was greater in patients treated concomitantly with clofazimine but none of them ever developed torsade de pointes [108]. One novel way that is being evaluated in preclinical studies to counteract the effect of Bedaquiline causing QTC prolongation is by adding verapamil [109]. Delamanid (OPC67687) 100 mg, has also entered phase IIb and III trials. Follow up data from trial 204 which enrolled 481 MDR-TB patients showed a 74.5 % favourable response among patients using delamanid for 6 months or more, compared to 55 % with use of delamanid for less than 2 months [110]. PA-824, a nitroimidazole (along with moxifloxacin and pyrazinamide) proved efficacious and safe in the phase II b study showing a greater bactericidal activity compared to standard ATT and is used in the STAND trial [94, 111].

Newer TB drugs and ART and drug–drug interactions and therapeutic implications

Bedaquiline concentration is increased with co-administered lopinavir/ritonavir by 22 % due to reduced clearance, while BDQ concentration is reduced by 50 % with efavirenz and these combinations are better avoided. Nevirapine has no interaction with BDQ and can be given safely [112]. Delamanid is expected not to have any clinically significant interaction with EFV, NVP or boosted PI and can be used concomitantly, as it neither induces nor inhibits the CYP 450 system. PA-824 concentrations are reduced by both EFV and lopinavir/ritonavir being 35 and 17 % respectively [113]. Newer derivatives of Linezolid in the pipeline are sutezolid, AZD 5487, radezolid and tedizolid, which are devoid of the myelosuppressive side effects of the linezolid, their predecessor [114].

Antiretroviral treatment (ART) in HIV-PTB

The influence of HIV and ART on TB outcomes

In the pre-HAART era, a number of studies have shown considerably lower cure rates, higher mortality and recurrence rates of TB after standard ATT in HIV co-infected patients compared to sero-negative individuals [115117]. The study in Spain showed an efficacy of 43 % in HIV infected Vs 70 % in the HIV negative TB [115]. Nevertheless, the study by Chaisson et al. did not reveal the outcomes to be different based on HIV status (87 % in the uninfected vs 81 % among co-infected). The median CD4 count of the study cohort was 475 cells/mm3 suggesting that preservation of adequate immunity could potentially improve TB outcomes in HIV emphasizing the need for early ART initiation [116]. Treatment of TB alone in HIV-TB co-infected patients did not substantially increase CD4 count nor reduced the viral load [117]. This had prompted both the WHO [118] and the National AIDS Control Organisation, (NACO) India [119] to recommend ART initiation irrespective of CD4 in HIV-TB infection. The updated systematic review on TB treatment in HIV by Faiz khan et al. of which the authors had also contributed, showed substantial improvement in TB treatment outcomes, reduction in relapse rates, mortality and acquisition of drug resistance with early ART initiation [74]. Timely ART initiation undoubtedly improved TB outcomes. Comparing HIV-TB studies conducted at NIRT in the pre- and post-ART era, the TB outcomes were significantly better with HAART co-administration (favourable response of 93 vs. 83 %) with a reduction in all-cause mortality from 17 to 5 % [67, 120]. Pulmonary TB patients initiating ART in the intensive phase had a better TB outcome (adjusted odds ratio 1.83 (95 % CI 1.29–2.60) compared those not on ART from a study from Malawi [121]. A study from Thailand also reflected the same trends with 43 % mortality in the absence of ART compared to 7 % when co-administered with ART [122].

The ideal ART regimen for TB co-infected patients and newer options available

Tenofovir, emtricitabine/lamivudine along with efavirenz as a single pill once a day is most ideal [123]. That efavirenz can still maintain an adequate plasma despite co-administration with rifampicin makes it suitable for co-administration with rifampicin in HIV-PTB [124, 125]. While efavirenz dose (600 vs 800 mg) and concurrent rifampicin administration had less clinical impact [126], a polymorphism in the CYP2B6 gene (G–T mutation) resulted in significantly higher blood levels of the drug resulting in increased risk of neuro-psychiatric toxicity [127]. Trials have shown lower dose of EFZ (400 mg) to be therapeutically useful, with effective virological suppression but with a lesser toxicity (39 vs 48 %) at the same time compared to the standard 600 mg [128]. Use of nevirapine (available as generic fixed-drug combination) is not recommended routinely with rifampicin, unless there is a contraindication to efavirenz like pregnancy or psychiatric illness. However, nevirapine initiation should be without a “lead in” period, starting with 200 mg twice daily to maximize efficacy in the presence of rifampicin [120, 129]. Caution needs to be exercised in patients with a higher CD4 (>400 in males and above 250 in females) who are prone for fulminant hepatitis while using this strategy [123]. Triple NRTI regimens containing abacavir can be used alternatively in patients negative for HLA B57:01, but virological suppression is inferior especially when the viral load is high [130]. A quadruple NRTI regimen including tenofovir has been found to be as effective as EFV based regimen in trials of a smaller number [131]. Rifabutin offers greater flexibility than rifampicin and the modifications that are required in concomitant therapy is depicted in Table 2.

Table 2 Therapeutic modifications that are required while administering rifamycins and ART concomitantly [103, 177]

Recently discovered drugs in the pipeline include tenofovir alafenamide (GS 7340), a prodrug of tenofovir, that gets converted at the site of lymphoid involvement and liver making it more potent, with greater tissue infiltration at a dosage ten times less than the conventional tenofovir and is currently evaluated in phase III trials in a fixed dose combination. There was significant improvement in estimated glomerular filtration rate, bone density and reduced proteinuria compared to conventional tenofovir disoproxil fumarate [132]. Dolutegravir has been shown to have a superior tolerability compared to darunavir in the Flamingo study [133]. The dosage of dolutegravir has to be doubled if concomitantly administered with rifampicin to 50 mg BD instead of the OD dosage (given with rifabutin) especially if given along with EFV [134]. Alternatively, dolutegravir 50 mg BD can also be combined with lamivudine and abacavir [123].

When to start ART after ATT initiation

Evidences from the START [135] and the TEMPRANO study [136] has made WHO recommend early initiation of ART, even in asymptomatic HIV individuals irrespective of CD4 so that Immune restoration is near complete with reduction in mortality due to both infective and non-infective causes with comparable toxicities. The START study which had 62 % of events attributed to TB showed that six participants in the immediate arm and 20 in the deferred arm of ART had broken down with TB favouring immediate ART initiation [135]. Early initiation of ART not only reduces mortality and morbidity due to HIV and TB with faster sputum conversion but also prevents IRIS secondarily [79]. The SAPIT trial observed 56 % lower mortality among patients who concomitantly started ART during TB treatment compared to those who subsequently started ART after ATT completion [137]. The important finding that can be deduced from the results of both the STRIDE [138] and SAPIT [139] trials is that while HIV-TB patients with a CD4 > 50 cells/mm3 need to start ART at the earliest with counselling on early recognition of signs and symptoms of IRIS and toxicity, a permissible delay of ART initiation among patients with higher CD4 up to 12 weeks after starting ATT helps in reducing drug interactions between rifampicin and NNRTIs, cumulative drug toxicities (especially hepatotoxicity), pill burden and the incidence of immune reconstitution inflammatory syndrome without any compromise on survival or predisposition to newer opportunistic infections [140].

The CAMELIA study enrolling 661 patients with a median follow-up of 25 months reflected similar findings that were deduced from patients in the lower range of CD4 in the SAPIT and STRIDES study. Early ART initiation by 2 weeks post ATT reduced mortality significantly compared to late initiation of ART at 8 weeks post ATT (18 vs 27 %, p = 0.0006). However, the risk of IRIS was 2.5 times more in the early ART group [141].

Current WHO and ART guidelines in India also recommend ART between 2 and 8 weeks of starting ATT on similar principles [119, 142].

Adverse reactions occur more often among HIV-infected patients with TB on concurrent medication than among TB without HIV (serious ADR—27 vs 13 %), occurring mostly in the first 2 months of treatment [143, 144]. Hepatoxicity is common due to shared metabolic pathways of anti-TB and antiretroviral drugs, alcoholism, co-infection with Hepatitis B and C, IRIS hepatitis and obstruction by nodes at the porta hepatitis all predispose to liver injury [88]. Most cases present with transaminitis which resolves when drugs are withheld. In drug sensitive TB, non-hepatotoxic drugs like streptomycin, ethambutol and ofloxacin should be substituted for TB treatment till liver functions return to normal, when all drugs can be re-introduced. Other adverse reactions include cutaneous and gastrointestinal symptoms and peripheral neuropathy which can occasionally be disabling [88]. Lactic Acidosis can present in various ways and needs periodic estimation [145].

Isolated hyperbilirubinemia is very common with use of atazanavir but is more of a cosmetic disfigurement that may stigmatise the patient. Zinc sulphate supplementation may be useful in management of ATV-related HBR in selected patients [146].

Summary

The ideal co-administered ART regimen with ATT is a single pill of tenofovir (300 mg) along with emtricitabine/lamivudine (300 mg) and efavirenz (600 mg) initiated between 2 and 12 weeks after ATT initiation, provided the patients’ CD4 cell count is above 50 cells/mm3 with compromise on survival, (when there is no clinical compulsion). However, among patients with a CD4 of less than or equal to 50 cells/mm, immediate initiation of ART is the rule with proper counselling for IRIS and close monitoring for cumulative/additive toxicity that will help to maintain adherence

Tuberculosis immune reconstitution inflammatory syndrome (TB-IRIS)

Tuberculosis immune reconstitution inflammatory syndrome (TB-IRIS) is the paradoxical worsening of symptoms and signs of TB after starting ART (rarely with ATT itself), despite a favorable immunological recovery and effective virological suppression [147]. Incidence ranges from 8–43 % [148]. The frequency and severity of IRIS depends on the degree of CD4 lymphocytopenia at nadir, the presence of other opportunistic infections and also the strategic location of the lesion. Figure 2 shows a pulmonary TB patient with HIV who presented with IRIS lesions in the brain after initiating ART [149, 150]. TB-IRIS is of two types (1) Paradoxical TB-IRIS that occurs in HIV-TB co-infected patients started on ATT and subsequently started on ART. Paradoxical IRIS is relatively easy to diagnose because of its biphasic pattern of initial improvement with ATT followed by a latter phase “paradoxical” deterioration after ART initiation. (2) Unmasking TB-IRIS or ANTIRETROVIRAL therapy (ART) associated TB occurs in an asymptomatic individual without a prior diagnosis of TB who starts developing symptoms with ART initiation. This could either be subclinical or undiagnosed TB [147, 151].

Fig. 2
figure 2

Pontine abscess as a manifestation of paradoxical TB-IRIS in a patients with HIV (& hemiparesis)

Predictors of IRIS—risk factors

The most consistent risk factors are very low levels of CD4 cell count, CD4/CD8 ratio, hemoglobin, weight, presence of disseminated disease, shorter ATT-ART time interval, extra pulmonary foci and other opportunistic infections at the time of ART initiation [150152]. Interleukin-6 and CRP were found to be reliable predictors of IRIS in a pure cohort of culture positive TB in HIV prospectively followed for IRIS occurrence in India. Evaluation of predictors from the CADRIS trial showed that baseline levels of vitamin D and higher D-dimer, Interferon gamma (IFN-γ), and sCD14 were independently associated with risk of IRIS [153]. TNFA-308*2 and IL-6174G gene polymorphisms have been implicated in TB-IRIS causation [154].

Pathogenesis of TB-IRIS

Functional restoration of immunocompetent cells (CD4) causing a cytokine outburst with an overriding Th1 over Th2 response, apart from increase in number and their redistribution to the site of the lesion, is the primary mechanism of IRIS causation [155]. The majority of paradoxical IRIS cases occurs within the first 3 months after starting ART [156]. The T-regulatory cells or committed suppressors of the immune system are normally restored post ART but are functionally defective, tilting the tide towards an inflammatory reaction [157]. Recently, MMP-7, a tissue matrix metallo-proteinase, has also been implicated in TB-IRIS [158].

Clinical features of TB IRIS

Fever with rigor or chills (resembling malaria) is the commonest and consistent symptom of TB-IRIS, with lymph node enlargement being the commonest manifestation [88]. Symptoms vary in severity from localized superficial lymphadenopathy and subcutaneous abscesses to severe forms like adult respiratory distress syndrome, meningitis, enlarging space occupying lesions like tuberculomas and viscus perforation, which can end fatally [151, 159]. Compressive features due to lymphadenopathy include stridor due to tracheal narrowing and superior vena caval (SVC) obstruction [160]. Patients with abdominal TB may present with pain and diarrhea. Other abdominal manifestations include hepatosplenomegaly, psoas abscesses, splenic micro abscesses, splenic rupture, epididymo-orchitis, uretric compression, and acute renal failure [159]. Osteomyelitis, sub-cutaneous abscesses and thromboembolic episodes have been reported [160]. Radiological worsening in pulmonary TB without symptoms or “cryptic IRIS” has been reported among TB patients alone after starting ATT in the pre HIV era also [161].

Diagnosis of IRIS

It is important for physicians to remember that the onset of this syndrome is linked temporally to ART initiation, ART substitution (from I line to II line suppressing viremia) and ART interruption followed by re-initiation [88]. A strong suspicion and awareness of IRIS supplemented by the immunological tools of CD4 T cell count and viral load, after ruling out toxicity and drug resistance, will help clinch the diagnosis [162]. Initial work up of febrile episodes for infections endemic to a particular place like malaria, urinary tract infection, typhoid needs to be performed. Radiological deterioration in Chest skiagram is a usual accompaniment in almost all cases of IRIS [153]. In some instances, increase in CD4 cell count may not be evident [156]. A decline of viral load less than 0.5 log10 copies/ml compared to baseline has a high negative predictive value in ruling out IRIS and is a mandatory criterion to differentiate it from HIV disease progression [159, 163]. The International network for the study of HIV associated IRIS (INSHI) criteria can be used for diagnosing unmasking IRIS and paradoxical IRIS [147]. Lipoarabinomannan (LAM) detection serves as a useful index of systemic antigen burden and has proved to be useful as a predictive marker [164].

When IRIS occurs at an extrapulmonary location, appropriate investigation can be performed and tissue specimen obtained wherever possible, and sent for bacteriological staining and culture in addition to routine histopathology. A negative culture that succeeds a pre-existing or baseline positive culture (that was sensitive to specific drug therapy) confirms the diagnosis of IRIS straightaway. However, this criteria of culture negativity cannot be applied for diagnosing unmasking IRIS or in paradoxical IRIS when the ATT-IRIS interval is too short. Hypercalcemia is a unique accompaniment of TB-IRIS, attributed to the calcium deposition through increased secretions of 1.25 dihydoxy cholecalciferol by activated macrophages and CD4 T cells [165].

Differential diagnosis of IRIS

Drug resistant TB is the closest mimic that requires to be ruled out before steroids are administered as they form the cornerstone of IRIS therapy currently [166]. The phenomenon of “fall and rise” exhibited by the acid fast bacilli (described by Toman) typically mimics the temporal sequence of paradoxical IRIS [70]. Steroid administration in the face of MDR-TB could spell disaster, ending fatally. Zidovudine induced anemia also mimics IRIS presenting with fever, rigor that settles after appropriate modification of ART [88]. Late onset IRIS (occurring after 3 months of ART initiation) is not rare but re-estimation of viral load is compulsory in such patients, to rule out ART failure and HIV progression [167]. Lymphoma of the Non-Hodgkins type which could occur or co-exist with TB in HIV may flare up after ART and steroid administration could confuse the hispathology findings [168].

Management

Preventive strategy

Intensive screening for TB and other opportunistic infections along with INH and cotrimaxozole prophylaxis reduces IRIS incidence [149, 167169]. ART initiation before CD4 goes down considerably could protect against opportunistic infections and subsequent IRIS [148]. If a patient with TB has a CD4 > 50 cells/mm3, the initiation of ART can be delayed up to 8 weeks with close monitoring that reduces toxicity and IRIS but does not compromise on survival [169].

Treatment

Anti-inflammatory drugs especially steroids form the backbone therapy for TB-IRIS, even though non-steroidal anti-inflammatory agents could prove to be a useful initial therapy for milder and localized cases of IRIS [153, 170]. A dose of 0.5–2 mg/kg body weight is usually used and tapered in a 4–8 weeks period depending on the site and severity of inflammation. Premature withdrawal of steroids can cause recrudescence of symptoms [153]. Severe forms may require parenteral steroids initially followed by switch to oral steroids. Thalidomide in steroid dependent IRIS has shown good results. [171] Maraviroc, monteleukast, pentoxifylline have not been proved to be effective in IRIS treatment [172]. Experimental drugs for IRIS treatment in the pipeline include CXCR-3 antagonists, the main receptor for CXCL-10 [173], IL-18 binding protein and IL-6 inhibitors [174, 175], anti-CD28 therapy [176] and drugs modulating MMP-7 activity [158].

Conclusions

HIV-associated pulmonary TB mandates a committed approach that encompasses both effective as well as enduring therapy originating from newer drug combinations, evolving ideas and emerging concepts from clinical trials globally, which if implemented in a proper and coordinated manner could not only save millions of lives but also offer a better quality of life to patients suffering from this coinfection.

Abbreviations

HIV:

human immune deficiency virus infection

TB:

tuberculosis

PTB:

pulmonary TB

ART:

antiretroviral therapy

ATT:

anti-tuberculosis therapy

LTBI:

latent TB infection

TST:

tuberculin sensitivity test

IGRA:

interferon gamma releases assay

CXR:

chest X-ray

AFB:

acid fast bacillus

M.Tb:

mycobacterium tuberculosis

MGIT:

mycobacterial growth indicator tube

NAAT:

nucleic acid amplification test

VEGF:

vascular endothelial growth factor

ADA:

adenosine deaminase

LAM:

lipoarabinomannan

LPA:

line probe assay

INH:

isoniazid

IPT:

INH preventive therapy

STCI:

standards of TB care in India

DOT:

directly observed therapy

ARR:

acquired rifampicin resistance

NIRT:

National Institute for Research in tuberculosis

CD:

cluster of differentiation

MDR:

multidrug resistant

XDR:

extensively drug resistance

IRIS:

immune reconstitution inflammatory syndrome

MMP:

matrix metallo-proteinases

INSHI:

international network for the study of HIV associated IRIS

References

  1. World Health Organization. Global tuberculosis report 2015. 2015. www.who.int/tb/publications/global_report/gtbr2015_executive_summary.pdf. Accessed Jun 2016.

  2. American Thoracic Society and Centers for Disease Control and Prevention. Targeted tuberculosis testing and treatment of latent tuberculosis infection. Am J Respir Crit Care Med. 2000;161:S221–47.

    Article  Google Scholar 

  3. Guidelines for intensified tuberculosis case finding and isoniazid preventive therapy for people living with HIV in resource constrained settings. STOP TB Department, World Health Organisation, Geneva. http://whqlibdoc.who.int/publications/…/9789241500708_eng.pdf.

  4. Nakamura RM, Einck L, Velmonte MA, et al. Detection of active tuberculosis by an MPB-64 transdermal patch: a field study. Scan J Infect Dis. 2001;33(6):405–7.

    Article  CAS  Google Scholar 

  5. Lalvani A. Diagnosing tuberculosis infection in the 21st century: new tools to tackle an old enemy. Chest. 2007;131:1898–906.

    Article  PubMed  Google Scholar 

  6. Rangaka MX, Wilkinson KA, Seldon R, et al. The effect of HIV-1 infection on T-cell-based and skin test detection of tuberculosis infection. Am J Respir Crit Care Med. 2007;175:514–20.

    Article  PubMed  Google Scholar 

  7. Zak DE, Penn-Nicholson A, Scriba TJ, et al. A blood RNA signature for tuberculosis disease risk: a prospective cohort study. Lancet. 2016;387(10035):2312–22.

    Article  CAS  PubMed  Google Scholar 

  8. World Health Organisation. Systematic screening for active tuberculosis. Principle and recommendations. World Health Organisation. http://apps.who.int/iris/bitstream/10665/84971/1/9789241548601_eng.pdf. Accessed 3 May 2016.

  9. Koole O, Thai S, Khun KE, et al. Evaluation of the 2007 WHO guideline to improve the diagnosis of tuberculosis in ambulatory HIV-positive adults. PLoS ONE. 2011;6(4):e18502. doi:10.1371/journal.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Getahun H, Kittikraisak W, Heiling CM, et al. Development of a standardized screening rule for tuberculosis in people living with HIV in resource constrained settings: individual participant data meta-analysis of observational studies. PLoS Med. 2011;8(1):e1000391.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Cain KP, McCarthy KD, Heilig CM, et al. An algorithm for tuberculosis screening and diagnsosi in people with HIV. N Engl J Med. 2010;362:707–16.

    Article  CAS  PubMed  Google Scholar 

  12. Swaminathan S, Narendran G, Menon PA, et al. Impact of HIV infection on radiographic features in patients with pulmonary tuberculosis. Indian J Chest Dis Allied Sci. 2007;49:133–6.

    Google Scholar 

  13. Akinbami AA, Adegboyega AO, Oshinaike OO, et al. Chest X-ray findings in HIV patients in relation to the CD4 count. Nig Q J Hosp Med. 2011;21(4):306–11.

    CAS  PubMed  Google Scholar 

  14. Padmapriyadarsini C, Tripathy S, Sekar L, et al. Evaluation of a diagnostic algorithm for sputum smear-negative pulmonary tuberculosis in HIV-Infected adults. J Acquir Immune Defic Syndr. 2013;63(3):331–8.

    Article  PubMed  Google Scholar 

  15. Heller T, Goblirsch S, Bahlas S, et al. Diagnostic value of FASH ultrasound and chest X-ray in HIV co-infected patients with abdominal tuberculosis. Int J Tuberc Lung Dis. 2013;17(3):342–4.

    Article  CAS  PubMed  Google Scholar 

  16. Monkongdee P, McCarthy KD, Cain KP, et al. Yield of acid-fast smear and mycobacterial culture for tuberculosis diagnosis in people with human immunodeficiency virus. Am J Respir Crit Care Med. 2009;180(9):903–8.

    Article  PubMed  Google Scholar 

  17. Cattamanchi A, Davis JL, Worodria W, et al. Sensitivity and specificity of fluorescence microscopy for diagnosing pulmonary tuberculosis in a high HIV prevalence setting. Int J Tuberc Lung Dis. 2009;13(9):1130–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Cattamanchi A, Davis JL, Pai M, et al. Does bleach processing increase the accuracy of sputum smear microscopy for diagnosing pulmonary tuberculosis? J Clin Microbiol. 2010;48:2433–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Thammavong C, Paboriboune P, Bouchard B, et al. Bleach treatment of sputum samples aids pulmonary tuberculosis screening among HIV-infected patients in Laos. Int J Tuberc Lung Dis. 2011;15(10):1353–8.

    Article  CAS  PubMed  Google Scholar 

  20. Gil-Setas A, Torroba L, Fernandez JL, et al. Evaluation of the MB/BacT system compared with Middlebrook 7H11 and Lowenstein–Jensen media for detection and recovery of mycobacteria from clinical specimens. Clin Microbiol Infect. 2004;10:224–8.

    Article  CAS  PubMed  Google Scholar 

  21. Lee JJ, Suo J, Lin CB, et al. Comparative evaluation of the BACTEC MGIT 960 system with solid medium for isolation of mycobacteria. Int J Tuberc Lung Dis. 2003;7:569–74.

    CAS  PubMed  Google Scholar 

  22. Farnia P, Mohammadi F, Mirsaedi M, et al. Application of oxidation reduction assay for monitoring treatment of patients with pulmonary tuberculosis. J Clin Microbiol. 2004;42:3324–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Brum CB, Ramos DF, Abilleira F, et al. The BACTEC MGIT(tm) 320 system as a laboratory tool to diagnose tuberculosis in a Brazilian hospital with a high prevalence of HIV infection. Rev Soc Bras Med Trop. 2016;49(1):112–4.

    Article  PubMed  Google Scholar 

  24. Kalantri S, Pai M, Pascopella L, et al. Bacteriophage based tests for the detection of Mycobacterium tuberculosis in clinical specimens: a systematic review and meta-analysis. BMC Infect Dis. 2005;5:59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Leung E, Minion J, Benedetti A, et al. Microcolony culture techniques for tuberculosis diagnosis: a systematic review. Int J Tuberc Lung Dis. 2012;16(1):16–23.

    Article  CAS  PubMed  Google Scholar 

  26. Molecular line probe assays for rapid screening of patients at risk of multidrug-resistant tuberculosis. WHO policy statement 2008. http://www.who.int/tb/features_archive/policy_statement.pdf. Accessed 7 Jan 2014.

  27. World Health Organization. Automated real-time nucleic acid amplification technology for rapid and simultaneous detection of tuberculosis and rifampicin resistance: Xpert MTB/RIF system: policy statement. Geneva: World Health Organization; 2011.

    Google Scholar 

  28. Alland D, Rowneki M, Smith L et al. Xpert MTB/RIF Ultra: a new near-patient TB test with sensitivity equal to culture abstract 91 CROI 2015, Seattle.

  29. Boehme CC, Nabeta P, Henostroza G, et al. Operational feasibility of using loop-mediated isothermal amplification for diagnosis of pulmonary tuberculosis in microscopy centers of developing countries. J Clin Micrbiol. 2007;45:1936–40.

    Article  CAS  Google Scholar 

  30. Hongmanee P, Stender H, Rasmussen OF. Evaluation of a fluorescence in situ hybridisation assay for differentiation between tuberculous and nontuberculous Mycobacteria species in smears of Lowenstein–Jensen and Mycobacteria Growth Indicator Tube cultures using peptide nucleic acid probes. J Clin Microbiol. 2001;39:1032–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. WHO. Tuberculosis serodiagnostic tests policy statement 2011. http://www.who.int/tb/features_archive/factsheet_serodiagnostic_test.pdf?ua=1 Accessed 290616.

  32. Zijenah LS, Kadzirange G, Bandason T, et al. Comparative performance characteristics of the urine lipoarabinomannan strip test and sputum smear microscopy in hospitalized HIV-infected patients with suspected tuberculosis in Harare, Zimbabwe. BMC Infect Dis. 2016;16:20.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Peter JG, Zijenah LS, Chanda D, et al. Effect on mortality of point-of-care, urine-based lipoarabinomannan testing to guide tuberculosis treatment initiation in HIV-positive hospital inpatients: a pragmatic, parallel-group, multicountry, open-label, randomised controlled trial. Lancet. 2016;387(10024):1187–97.

    Article  PubMed  Google Scholar 

  34. World Health Organization (WHO). The use of lateral flow urine lipoarabinomannan assay (LF-LAM) for the diagnosis and screening of active tuberculosis in people living with HIV. http://www.who.int/tb/publications/useof-lf-lam-tb-hiv/en/. Accessed 10 Jun 2016.

  35. Kaplan JE, Benson C, Holmes KH, et al. Guidelines for prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: recommendations from CDC, the National Institutes of Health, and the HIV Medicine Association of the Infectious Diseases Society of America. MMWR Recomm Rep. 2009;58(RR-4):1–207.

    Google Scholar 

  36. World Health Organization. Guidelines for intensified tuberculosis case-finding and isoniazid preventive therapy for people living with HIV in resource-constrained settings. Geneva: World Health Organization. 2011. (http://whqlibdoc.who.int/publications/2011/9789241500708_eng.pdf).

  37. Akolo C, Adetifa I, Shepperd S, et al. Treatment of latent tuberculosis infection in HIV infected persons. Cochrane Database Syst Rev. 2010;1:CD000171.

    PubMed  Google Scholar 

  38. Recommendations for use of an isoniazid-rifapentine regimen with direct observation to treat latent Mycobacterium tuberculosis infection. Morb Mortal Wkly Rep (MMWR). 2011;60(48):1650–3.

  39. Sterling TR, Villarino ME, Borisov AS, et al. Three months of weekly rifapentine and isoniazid for latent tuberculosis infection. NEJM. 2011;365:2155–66.

    Article  CAS  PubMed  Google Scholar 

  40. Getahun H, Granich R, Sculier D, et al. Implementation of isoniazid preventive therapy for people living with HIV worldwide: barriers and solutions. AIDS. 2010;24(suppl 5):S57–65.

    Article  PubMed  Google Scholar 

  41. Lewis JJ, Fielding KL, Grant AD, et al. Eligibility for Isoniazid Preventive Therapy in South African Gold Mines. PLoS ONE. 2013;8(11):e81376. doi:10.1371/journal.pone.0081376.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lawn SD, Kerkhoff AD, Vogt M, et al. Diagnostic and prognostic value of serum C-reactive protein for screening for HIV-associated tuberculosis. Int J Tuberc Lung Dis. 2013;17(5):636–43.

    Article  CAS  PubMed  Google Scholar 

  43. Yoon C, Davis JL, Huang L, et al. Point-of-care C-reactive protein testing to facilitate implementation of isoniazid preventive therapy for people living with HIV. J Acquir Immune Defic Syndr. 2014;65(5):551–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Suthar AB, Lawn SD, del Amo J, et al. Antiretroviral therapy for prevention of tuberculosis in adults with HIV: a systematic review and meta-analysis. PLoS Med. 2012;9(7):e1001270.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Rangaka MX, Boulle A, Wilkinson RJ, et al. Randomized controlled trial of isoniazid preventive therapy in HIV-infected persons on antiretroviral therapy. Clin Infect Dis. 2012;55(12):1698–706.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hosseinipour MC, Bisson GP, Miyahara S, et al. Empirical tuberculosis therapy versus isoniazid in adult outpatients with advanced HIV initiating antiretroviral therapy (REMEMBER): a multicountry open-label randomised controlled trial. Lancet. 2016;387(10024):1198–209.

    Article  CAS  PubMed  Google Scholar 

  47. Pho MT, Swaminathan S, Kumarasamy N, et al. The cost-effectiveness of tuberculosis preventive therapy for HIV-infected individuals in southern India: a trial-based analysis. PLoS ONE. 2012;7(4):e36001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kapoor S, Gupta A, Shah M. Cost-effectiveness of isoniazid preventive therapy for HIV-infected pregnant women in India. Int J Tuberc Lung Dis. 2016;20(1):85–92.

    Article  CAS  PubMed  Google Scholar 

  49. Azadi M, Bishai DM, Dowdy DW, et al. Cost-effectiveness of tuberculosis screening and isoniazid treatment in the TB/HIV in Rio (THRio) Study. Int J Tuberc Lung Dis. 2014;18(12):1443–8.

    Article  CAS  PubMed  Google Scholar 

  50. Shepardson D, Marks SM, Chesson H, et al. Cost-effectiveness of a 12-dose regimen for treating latent Tuberculous infection in the United States. Int J Tuberc Lung Dis. 2013;17(12):1531–7.

    Article  CAS  PubMed  Google Scholar 

  51. Smith T, Samandari T, Abimbola T, et al. Implementation and operational research: cost-effectiveness of antiretroviral therapy and isoniazid prophylaxis to reduce tuberculosis and death in people living with HIV in Botswana. J Acquir Immune Defic Syndr. 2015;70(3):e84–93.

    Article  CAS  PubMed  Google Scholar 

  52. Balcells ME, Thomas SL, Godfrey-Faussett P, et al. Isoniazid preventive therapy and risk for resistant tuberculosis. Emerg Infect Dis. 2006;12(5):744–51. doi:10.3201/eid1205.050681.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Samandari T, Agizew TB, Nyirenda S, et al. 6-Month versus 36-month isoniazid preventive treatment of tuberculosis in adults with HIV infection in Botswana: a randomised, double-blind, placebo-controlled trial. Lancet. 2011;377(9777):1588–98.

    Article  CAS  PubMed  Google Scholar 

  54. Swaminathan S, Menon PA, Gopalan N, et al. Efficacy of a six-month versus a 36-month regimen for prevention of tuberculosis in HIV-infected persons in India: a randomized clinical trial. PLoS ONE. 2012;7(12):e47400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Martinson NA, Barnes GL, Moulton LH, et al. New regimens to prevent tuberculosis in adults with HIV infection. N Engl J Med. 2011;365(1):11–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sterling TR, Scott NA, Miro JM, et al. Three months of weekly rifapentine and isoniazid for treatment of Mycobacterium tuberculosis infection in HIV co-infected persons. AIDS. 2016;30(10):1607–15.

    Article  CAS  PubMed  Google Scholar 

  57. Burman WJ, Jones B. Treatment of HIV-related tuberculosis in the era of effective antiretroviral therapy. AJRCC. 2001;164(1):7–12.

    CAS  Google Scholar 

  58. WHO Standards for TB care in India. www.searo.who.int/india/meidacentre/events/2014/stci_book.pdf.

  59. Mitchinson DA. Role of individual drugs in the chemotherapy of tuberculosis. Int J Tuberc Lung Dis. 2000;4(9):796–806.

    Google Scholar 

  60. Jindani A, Nunn AJ, Enarson DA. Two 8-month regimens of chemotherapy for treatment of newly diagnosed pulmonary tuberculosis: international multicentre randomised trial. Lancet. 2004;364(9441):1244–51.

    Article  CAS  PubMed  Google Scholar 

  61. O’Donnel MM, Souza Carvalho S, et al. Poor response to tuberculosis treatment with regimens without rifampicin in immunosuppressed AIDS patients. Braz J Infect Dis. 2002;6(6):272–5.

    PubMed  Google Scholar 

  62. el-Sadr WM, Perlman DC, Matts JP, et al. Evaluation of an intensive intermittent-induction regimen and duration of short-course treatment for human immunodeficiency virus-related pulmonary tuberculosis. Terry Beirn Community Programs for Clinical Research on AIDS (CPCRA) and the AIDS Clinical Trials Group (ACTG). Clin Infect Dis. 1998;26(5):1148–58.

    Article  CAS  PubMed  Google Scholar 

  63. Brindle RJ, Nunn PP, Githui W, et al. Quantitative bacillary response to treatment in HIV-associated pulmonary tuberculosis. Am Rev Respir Dis. 1993;147(4):958–61.

    Article  CAS  PubMed  Google Scholar 

  64. Ackah AN, Coulibaly D, Digbeu H, Greenberg AE, De Cock KM, et al. Response to treatment, mortality, and CD4 lymphocyte counts in HIV-infected persons with tuberculosis in Abidjan. Cote d’Ivoire. Lancet. 1995;345(8950):607–10.

    Article  CAS  PubMed  Google Scholar 

  65. Kumwenda J et al. Empirical therapy versus IPT in hiv-Infected persons initiating ART (ACTG A5274 48w). CROI 20166 abstract 745. Boston.

  66. Centre for Disease Control. Guidelines for prevention and treatment of opportunistic infections in HIV-infected adults and adolescents. Am Thorac Soc MMWR. 2009;58:1–198.

    Google Scholar 

  67. Swaminathan S, Narendran G, Venkatesan P, et al. Efficacy of a 6-month versus 9-month intermittent treatment regimen in HIV-infected patients with tuberculosis: a randomized clinical trial. Am J RespirCrit Care Med. 2010;181(7):743–51.

    Article  Google Scholar 

  68. Menzies D, Benedetti A, Paydar A, et al. Effect of duration and intermittency of rifampin on tuberculosis treatment outcomes: a systematic review and meta-analysis. PLoS Med. 2009;6(9):e1000146.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Tuberculosis Chemotherapy Centre. Intermittent treatment of pulmonary tuberculosis. A concurrent comparison of twice-weekly isoniazid plus streptomycin and daily isoniazid plus p-Aminosalicylic acid in domiciliary treatment. Lancet. 1963;1078–80.

  70. Toman K. What is the ‘fall and rise phenomenon’. Tuberculosis case finding and chemotherapy, questions and answers. Geneva: WHO; 1979.

    Google Scholar 

  71. Chang KC, Leung CC, Grosset J, et al. Treatment of tuberculosis and optimal dosing schedules. Thorax. 2011;66(11):997–1007.

    Article  PubMed  Google Scholar 

  72. Khan FA, Minion J, Pai M, et al. Treatment of active tuberculosis in HIV-coinfected patients: a systematic review and meta-analysis. Clin Infect Dis. 2010;50(9):1288–99.

    Article  PubMed  Google Scholar 

  73. Li J, Munsiff SS, Driver CR, Sackoff J. Relapse and acquired rifampin resistance in HIV-infected patients with tuberculosis treated with rifampin- or rifabutin-based regimens in New York City, 1997–2000. Clin Infect Dis. 2005;41(1):83–91.

    Article  CAS  PubMed  Google Scholar 

  74. Ahmad Khan F, Minion J, Al-Motairi A, et al. An updated systematic review and meta-analysis on the treatment of active tuberculosis in patients with HIV infection. Clin Infect Dis. 2012;55(8):1154–63.

    Article  CAS  PubMed  Google Scholar 

  75. Narendran G, Ramesh Kumar S, Menon PA, et al. Daily is better than thrice-weekly ATT in HIV patients with culture confirmed pulmonary TB,- a RCT from South India (CTRI-476/09, NCT00933790) WEAB0201 presented at AIDS 2016 conference, Durban.

  76. Bifani P, Mathema B, Kurepina N, et al. The evolution of drug resistance in Mycobacterium tuberculosis: from a mono-rifampin-resistant cluster into increasingly multidrug-resistant variants in an HIV-seropositive population. J Infect Dis. 2008;198(1):90–4.

    Article  CAS  PubMed  Google Scholar 

  77. Tuberculosis Research Centre. Low rate of emergence of drug resistance in sputum positive patients treated with short course chemotherapy. Int J Tuberc Lung Dis. 2001;5:40–5.

    Google Scholar 

  78. Vernon A, Burman W, Benator D, et al. Acquired rifamycinmonoresistance in patients with HIV-related tuberculosis treated with once-weekly rifapentine and isoniazid. Tuberculosis trials consortium. Lancet. 1999;353(9167):1843–7.

    Article  CAS  PubMed  Google Scholar 

  79. Nahid P, Gonzalez LC, Rudoy I, et al. Treatment outcomes of patients with HIV and tuberculosis. Am J RespirCrit Care Med. 2007;175(11):1199–206.

    Article  Google Scholar 

  80. Burman W, Benator D, Vernon A, et al. Acquired rifamycin resistance with twice-weekly treatment of HIV-related tuberculosis. Am J Respir Crit Care Med. 2006;173:350–6.

    Article  CAS  PubMed  Google Scholar 

  81. Gurumurthy P, Ramachandran G, Hemanth Kumar AK, et al. Decreased bioavailability of rifampin and other antituberculosis drugs in patients with advanced human immunodeficiency virus disease. Antimicrob Agents Chemother. 2004;48(11):4473–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Perlman DC, Segal Y, Rosenkranz S, et al. The clinical pharmacokinetics of rifampin and ethambutol in HIV-infected persons with tuberculosis. Clin Infect Dis. 2005;41(11):1638–47.

    Article  CAS  PubMed  Google Scholar 

  83. Hemanth Kumar AK, Narendran G, Kumar RS, et al. Rifampicin exposure is lower in HIV-infected TB patients receiving intermittent than daily anti-TB treatment. RMP exposure is lower in HIV-infected TB patients receiving intermittent than daily anti-tuberculosis treatment. IJTLD. 2015;19(7):805–7.

    Article  CAS  Google Scholar 

  84. Narendran G, Menon PA, Venkatesan P, et al. Acquired rifampicin resistance in thrice-weekly TB therapy: impact of HIV and antiretroviral therapy. Clinical Infectious diseases. Clin Infect Dis. 2014;59(12):1798–804.

    Article  PubMed  Google Scholar 

  85. Korenromp EL, Scano F, Williams BG, et al. Effects of human immunodeficiency virus infection on recurrence of tuberculosis after rifampin-based treatment: an analytical review. Clin Infect Dis. 2003;37(1):101–12.

    Article  PubMed  Google Scholar 

  86. Narayanan S, Swaminathan S, Supply P, et al. Impact of HIV infection on the recurrence of tuberculosis in South India. J Infect Dis. 2010;201(5):691–703.

    Article  PubMed  Google Scholar 

  87. Guidelines for PMDT in India 2012. http://tbcindia.gov.in/WriteReadData/l892s/8320929355Guidelines%20for%20PMDT%20in%20India%20-%20May%202012.pdf. Accessed 25 Jun 2016.

  88. Narendran G, Swaminathan S. Tuberculosis immune reconstitution inflammatory syndrome: profile of an enigmatic condition. Curr Sci. 2013;105(5):657–65.

    CAS  Google Scholar 

  89. Caminero JA. Guidelines for clinical and operational management of drug-resistant tuberculosis 2013. http://www.theunion.org/what-we-do/publications/technical/english/mdr-tbguide_6-19-13_web.pdf. Accessed 25 Jun 2016.

  90. Gopinath K, Singh S. Non-tuberculous mycobacteria in TB-endemic countries: are we neglecting the danger? PLoS Negl Trop Dis. 2010;4(4):e615. doi:10.1371/journal.pntd.0000615.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Helal ZH, Khan MI, Ashour MSE-D, et al. Detection and characterization of nocardia from patients diagnosed as tuberculosis in Egypt. Int J Biomed Sci. 2008;4(3):179–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Jindani A, Harrison TS, Nunn AJ, et al. High-dose rifapentine with moxifloxacin for pulmonary tuberculosis. N Engl J Med. 2014;371:1599–608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Merle CS, Sismanidis C, Sow OB, et al. A pivotal registration phase III, multicenter, randomized tuberculosis controlled trial: design issues and lessons learnt from the Gatifloxacin for TB (OFLOTUB) project. Trials. 2012;13:61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Dawson R, Diacon AH, Everitt D, et al. Efficiency and safety of the combination of moxifloxacin, pretomanid (PA-824), and pyrazinamide during the first 8 weeks of antituberculosis treatment: a phase 2b, open-label, partly randomised trial in patients with drug-susceptible or drug-resistant pulmonary tuberculosis. Lancet. 2015;385(9979):1738–47.

    Article  CAS  PubMed  Google Scholar 

  95. Wallis RS, Wang C, Meyer D, Thomas N. Month 2 culture status and treatment duration as predictors of tuberculosis relapse risk in a meta-regression model. PLoS ONE. 2013;8(8):e71116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Wells CD, Cegielski P, Nelson LJ, et al. HIV infection and multi drug resistant tuberculosis: the perfect storm. J Infect Dis. 2007;196:S86–107.

    Article  PubMed  Google Scholar 

  97. WHO treatment guidelines for drug-resistant tuberculosis: 2016. http://www.who.int/tb/MDRTBguidelines2016.pdf. Accessed 28 Jun 2016.

  98. World Health Organization. The shorter MDR-TB regimen. http://www.who.int/tb/Short_MDR_regimen_factsheet.pdf. Accessed 28 Jun 2016.

  99. Aung KJM, Van Deun A, Declercq E, et al. Successful ‘9-month Bangladesh regimen’ for multidrugresistant tuberculosis among over 500 consecutive patients. Int J Tuberc Lung Dis. 2014;18(10):1180–7.

    Article  CAS  PubMed  Google Scholar 

  100. Kuaban, et al. West African Cohort study. Late breaker session. 46th Union World Conference on Lung Health, Cape Town.

  101. Palacios E, Franke M, Muñoz M, et al. HIV-positive patients treated for multidrug-resistant tuberculosis: clinical outcomes in the HAART era. Int J Tuberc Lung Dis. 2012;16(3):348–54.

    Article  CAS  PubMed  Google Scholar 

  102. Menon S. Preventing nosocomial MDR TB transmission in sub Saharan Africa: where are we at? Glob J Health Sci. 2013;5(4):200–10. doi:10.5539/gjhs.v5n4p200.

    PubMed  PubMed Central  Google Scholar 

  103. CDC. Managing drug interactions in the treatment of HIV-related tuberculosis. 2013. http://www.cdc.gov/tb/TB_HIV_Drugs/default.htm.

  104. Van der Walt M, Lancaster J, Odendaal R, et al. Serious treatment related adverse drug reactions amongst anti-retroviral Naïve MDR-TB patients. PLoS ONE. 2013;8(4):e58817. doi:10.1371/journal.pone.0058817.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Pietersen E, Ignatius E, Streicher EM, et al. Long-term outcomes of patients with extensively drug-resistant tuberculosis in South Africa: a cohort study. Lancet. 2014;383:1230–9.

    Article  PubMed  Google Scholar 

  106. Fox GJ, Menzies D. A review of the evidence for using bedaquiline (TMC207) to treat multi-drug resistant tuberculosis. Infect Dis Ther. 2013;2:123–44.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Andries K, Verhasselt P, Guillemont J, et al. A diarylquinoline drug active on the ATP synthase of Mycobacterium tuberculosis. Science. 2005;307:223–7.

    Article  CAS  PubMed  Google Scholar 

  108. Mase S, Chorba T, Lobue P, Castro K. Provisional CDC guidelines for the use and safety monitoring of bedaquiline fumarate (Sirturo) for the treatment of multidrug-resistant tuberculosis. MMWR. 2013;62(9):1.

    Google Scholar 

  109. Srikrishna G, Gupta S, Dooley KE, Bishai WR. Can the addition of verapamil to Bedaquiline containing regimens improve tuberculosis treatment outcomes? A novel approach to optimising TB treatment. Future microbiology 2015;10(8): 1257-6014 B].

  110. Skripconoka V, Danilovits M, Pehme L, et al. Delamanid improves outcomes and reduces mortality in multidrug-resistant tuberculosis. Eur Respir J. 2013;41(6):1393–400.

    Article  PubMed  Google Scholar 

  111. Shortening treatment by advancing novel drugs (STAND). NCT02342886 https://clinicaltrials.gov/ct2/show/NCT02342886?term=NCT02342886&rank=1. Accessed 28 Jun 2016.

  112. Van Heeswijk RPG, Dannemann B, Hoetelmans RMW. Bedaquiline: a review of human pharmacokinetics and drug–drug interactions. J Antimicrob Chemother. 2014;. doi:10.1093/jac/dku171.

    PubMed  Google Scholar 

  113. Dooley K. Drug–drug interactions in TB-diabetes and HIV-TB. “Developing a comprehensive therapeutic research strategy for the converging epidemics of TB, T2DM, and HIV” Workshop NIAID, NIDDK, Office of AIDS Research 11th May 2016.

  114. Zumla A, Gillepsie SH, Hoelscher M, et al. New antituberculosis drugs, regimens and adjuvant therapies: needs, advances and future prospects. Lancet. 2014;14:237–340.

    Google Scholar 

  115. Ruiz-Navarro MD, Hernández Espinosa JA, Bleda Hernández MJ, et al. Effects of HIV status and other variables on the outcome of tuberculosis treatment in Spain. Arch Bronconeumol. 2005;41(7):363–70.

    PubMed  Google Scholar 

  116. Chaisson RE, Clermont HC, Holt EA, et al. Six-month supervised intermittent tuberculosis therapy in Haitian patients with and without HIV. Am J Respir Crit Care Med. 1996;154:1034–8.

    Article  CAS  PubMed  Google Scholar 

  117. Morris L, Martin DJ, Bredell H, et al. HIV-1 RNA levels and CD4 lymphocyte counts during treatment for active tuberculosis in South African patients. J Infect Dis. 2003;187:1967–71.

    Article  PubMed  Google Scholar 

  118. World Health Organization (WHO). Rapid advice: antiretroviral therapy for HIV infection in adults and adolescents. 2009. http://www.searo.who.int/LinkFiles/HIV-AIDS_Rapid_Advice_Adult_ART_Guidelines(web).pdf. Accessed 30 Jan 2013.

  119. Revised NACO guidelines for ART initiation in adults. Office memorandum T-11020/36/2005 dated 4 November 2011.

  120. Swaminathan S, Padmapriyadarsini C, Venkatesan P, et al. Efficacy and safety of once-daily nevirapine-or efavirenz-based antiretroviral therapy in HIV associated tuberculosis: a randomized clinical trial. Clin Infect Dis. 2011;53(7):716–24.

    Article  CAS  PubMed  Google Scholar 

  121. Tweya Tweya H, Feldacker C, Phiri S, et al. Comparison of treatment outcomes of new smear-positive pulmonary tuberculosis patients by HIV and antiretroviral status in a TB/HIV clinic, Malawi. PLoS ONE. 2013;8(2):e56248. doi:10.1371/journal.pone.0056248.

    Article  CAS  PubMed  Google Scholar 

  122. Akksilp S, Karnkawinpong O, Wattanaamornkiat W, et al. Antiretroviral therapy during tuberculosis treatment and marked reduction in death rate of HIV-infected patients, Thailand. Emerg Infect Dis. 2007;13(7):1001–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Panel on antiretroviral guidelines for adults and adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. Department of Health and Human Services. http://www.aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf. Accessed 27 Jun 2016.

  124. Patel A, Patel K, Patel J, et al. Safety and antiretroviral effectiveness of concomitant use of rifampicin and efavirenz for antiretroviral-naive patients in India who are coinfected with tuberculosis and HIV-1. J Acquir Immune Defic Syndr. 2004;37:1166–9.

    Article  CAS  PubMed  Google Scholar 

  125. Pedral-Sampaio DB, Alves CR, Netto EM, et al. Efficacy and safety of efavirenz in HIV patients on rifampin for tuberculosis. Braz J Infect Dis. 2004;8:211–6.

    Article  CAS  PubMed  Google Scholar 

  126. Manosuthi W, Kiertiburanakul S, Sungkanuparph S, et al. Efavirenz 600 mg/day versus efavirenz 800 mg/day in HIV-infected patients with tuberculosis receiving rifampicin: 48 weeks results. AIDS. 2006;20:131–2.

    Article  CAS  PubMed  Google Scholar 

  127. Ramachandran G, Hemanth Kumar AK, Rajasekharan S, et al. CYP2B6 G516T polymorphism but not rifampicin coadminstration influences steady state pharmacokinetics of efavirenz in human immunodeficiency virus infected patients in South India. Antimicrob Agents Chemother. 2009;53:863–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Carey D, Puls R, Amin J, et al. Efficacy and safety of efavirenz 400 mg daily versus 600 mg daily: 96-week data from the randomised, double-blind, placebo-controlled, non-inferiority ENCORE1 study. Lancet Infect Dis. 2015;15(7):793–802.

    Article  CAS  PubMed  Google Scholar 

  129. Manosuthi W, Tantanathip P, Chimsuntorn S, et al. Treatment outcomes of patients co-infected with HIV and tuberculosis who received a nevirapine-based antiretroviral regimen: a four-year prospective study. Int J Infect Dis. 2010;14:e1013–7.

    Article  PubMed  Google Scholar 

  130. Munderi P, Walker AS, Kityo C, et al. Nevirapine/zidovudine/lamivudine has superior immunological and virological responses not reflected in clinical outcomes in a 48-week randomized comparison with abacavir/zidovudine/lamivudine in HIV-infected Ugandan adults with low CD4 cell counts. HIV Med. 2010;11:334–44.

    Article  CAS  PubMed  Google Scholar 

  131. Moyle G, Higgs C, Teague A, et al. An open-label, randomized comparative pilot study of a single-class quadruple therapy regimen versus a 2-class triple therapy regimen for individuals initiating antiretroviral therapy. Antivir Ther. 2006;11:73–8.

    CAS  PubMed  Google Scholar 

  132. Gallant JE, et al. Switching tenofovir DF to tenofovir alafenamide in virologically suppressed adults. CROI 2016. Abstract 29. Boston.

  133. Molina JM, Clotet B, van Lunzen J, et al. Once-daily dolutegravir versus darunavir plus ritonavir for treatment-naive adults with HIV-1 infection (FLAMINGO): 96 week results from a randomised, open-label, phase 3b study. Lancet HIV. 2015;2(4):e127–36.

    Article  PubMed  Google Scholar 

  134. Dooley KE, Sayre P, Borland J, et al. Safety, tolerability, and pharmacokinetics of the HIV integrase inhibitor dolutegravir given twice daily with rifampin or once daily with rifabutin: results of a phase 1 study among healthy subjects. J Acquir Immune Defic Synr. 2013;62(1):21–7.

    Article  CAS  Google Scholar 

  135. The INSIGHT START Study Group. Initiation of antiretroviral therapy in early asymptomatic HIV infection. N Engl J Med. 2015;373:795–807.

    Article  CAS  PubMed Central  Google Scholar 

  136. The TEMPRANO ANRS 12136 Study Group. A trial of early antiretrovirals and isoniazid preventive therapy in Africa. N Engl J Med. 2015;373:808–22.

    Article  CAS  Google Scholar 

  137. Abdool Karim SS, Naidoo K, Grobler A, et al. Timing of initiation of antiretroviral drugs during tuberculosis therapy. N Engl J Med. 2010;362(8):697–706.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Havlir DV, Kendall MA, Ive P, et al. Timing of antiretroviral therapy for HIV-1 infection and tuberculosis. N Engl J Med. 2011;365(16):1482–91 (stride).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Abdool Karim SS, Naidoo K, Grobler A, et al. Integration of antiretroviral therapy with tuberculosis treatment. N Engl J Med. 2011;365(16):1492–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Swaminathan S, Padmapriyadarsini C, Narendran G. HIV associated tuberculosis—clinical update. Clin Infect Dis. 2010;50(10):1377–86.

    Article  CAS  PubMed  Google Scholar 

  141. Blanc FX, Sok T, Laureillard D, et al. Earlier versus later start of antiretroviral therapy in HIV-infected adults with tuberculosis. N Engl J Med. 2011;365:1471–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. WHO. Consolidated guidelines on the use of antiretroviral drugs for treating and preventing HIV infection. http://apps.who.int/iris/bitstream/10665/85321/1/9789241505727_eng.pdf?ua=1. Accessed 08 May 2014.

  143. Yimer G, Aderaye G, Amogne W, et al. Anti-tuberculosis therapy-induced hepatotoxicity among Ethiopian HIV-positive and negative patients. PLoS ONE. 2008;3(3):e1809. doi:10.1371/journal.pone.0001809.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Pukenyte E, Lescure FX, Rey D, et al. Incidence of and risk factors for severe liver toxicity in HIV-infected patients on anti-tuberculosis treatment. Int J Tuberc Lung Dis. 2007;11:78–84.

    CAS  PubMed  Google Scholar 

  145. Imhof A, Ledergerber B, Huldrych F, The Swiss HIV Cohort Study A, et al. Risk factors for and outcome of hyperlactatemia in HIV-infected persons: is there a need for routine lactate monitoring? Clin Infect Dis. 2005;41:721–8.

    Article  PubMed  Google Scholar 

  146. Moyle G, Else L, Jackson A, et al. Coadministration of atazanavir–ritonavir and zinc sulfate: impact on hyperbilirubinemia and pharmacokinetics. Antimicrob Agents Chemother. 2013;57(8):3640–4. doi:10.1128/AAC.00357-13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Meintjes G, Lawn SD, Scano F, et al. Tuberculosis-associated immune reconstitution inflammatory syndrome: case definitions for use in resource-limited settings. Lancet Infect Dis. 2008;8(8):516–23.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Breton G, Duval X, Estellat C, et al. Determinants of immune reconstitution inflammatory syndrome in HIV type 1-infected patients with tuberculosis after initiation of antiretroviral therapy. Clin Infect Dis. 2004;39(11):1709–12.

    Article  PubMed  Google Scholar 

  149. Haddow LJ, Moosa MY, Mosam A, et al. Incidence, clinical spectrum, risk factors and impact of HIV-associated immune reconstitution inflammatory syndrome in South Africa. PLoS ONE. 2012;7(11):e40623.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Gopalan N, Andrade BB, Swaminathan S. Tuberculosis-immune reconstitution inflammatory syndrome in HIV: from pathogenesis to prediction. Expert Rev Clin Immunol. 2014;10(5):631–45.

    Article  CAS  PubMed  Google Scholar 

  151. Lawn SD, Meintjes G. Pathogenesis and prevention of immune reconstitution disease during antiretroviral therapy. Expert Rev Anti Infect Ther. 2011;9(4):415–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Narendran G, Andrade BB, Porter BO, et al. Paradoxical tuberculosis immune reconstitution inflammatory syndrome (TB-IRIS) in HIV patients with culture confirmed pulmonary tuberculosis in India and the potential role of IL-6 in prediction. PLoS ONE. 2013;8(5):63541.

    Article  CAS  Google Scholar 

  153. Musselwhite LW, Andrade BB, Ellenberg SS, et al. Vitamin D, d-dimer, interferon gamma, and sCD14 levels are independently associated with immune reconstitution inflammatory syndrome: a prospective, international Study. EBioMedicine. 2016;4:115–23.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Price P, Morahan G, Huang D, et al. Polymorphisms in cytokine genes define subpopulations of HIV-1 patients who experienced immune restoration diseases. AIDS. 2002;16(15):2043–7.

    Article  CAS  PubMed  Google Scholar 

  155. Bourgarit A, Carcelain G, Martinez V, et al. ExPLOS ion of tuberculin-specific Th1-responses induces immune restoration syndrome in tuberculosis and HIV co-infected patients. AIDS. 2006;20(2):F1–7.

    Article  CAS  PubMed  Google Scholar 

  156. Manabe YC, Campbell JD, Sydnor E, et al. Immune reconstitution inflammatory syndrome: risk factors and treatment implications. J Acquir Immune Defic Syndr. 2007;46(4):456–62.

    Article  CAS  PubMed  Google Scholar 

  157. Seddiki N, Sasson SC, Santner-Nanan B, et al. Proliferation of weakly suppressive regulatory CD4+ T cells is associated with over-active CD4+ T-cell responses in HIV-positive patients with mycobacterial immune restoration disease. Eur J Immunol. 2009;39(2):391–403.

    Article  CAS  PubMed  Google Scholar 

  158. Tadokera R, Meintjes GA, Wilkinson KA, et al. Matrix metalloproteinases and tissue damage in HIV-tuberculosis immune reconstitution inflammatory syndrome. Eur J Immunol. 2014;44(1):127–36.

    Article  CAS  PubMed  Google Scholar 

  159. Lawn SD, Bekker LG, Miller RF. Immune reconstitution disease associated with mycobacterial infections in HIV-infected individuals receiving antiretrovirals. Lancet Infect Dis. 2005;5(6):361–73.

    Article  PubMed  Google Scholar 

  160. Buckingham SJ, Haddow LJ, Shaw PJ, et al. Immune reconstitution inflammatory syndrome in HIV-infected patients with mycobacterial infections starting highly active anti-retroviral therapy. Clin Radiol. 2004;59(6):505–13.

    Article  CAS  PubMed  Google Scholar 

  161. Swaminathan S, Narendran G. HIV and tuberculosis in India. J Biosci. 2008;33(4):527–37.

    Article  PubMed  Google Scholar 

  162. Shelburne SA, Montes M, Hamill RJ. Immune reconstitution inflammatory syndrome: more answers, more questions. J Antimicrob Chemother. 2006;57(2):167–70.

    Article  CAS  PubMed  Google Scholar 

  163. French MA, Price P, Stone SF, et al. Immune restoration disease after antiretroviral therapy. AIDS. 2004;18:1615–27.

    Article  CAS  PubMed  Google Scholar 

  164. Lawn SD. Point-of-care detection of lipoarabinomannan (LAM) in urine for diagnosis of HIV-associated tuberculosis: a state of the art review. BMC Infect Dis. 2012;12:103.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Lawn SD, Macallan DC. Hypercalcemia: a manifestation of immune reconstitution complicating tuberculosis in an HIV-infected person. Clin Infect Dis. 2004;38(1):154–5.

    Article  PubMed  Google Scholar 

  166. Meintjes G, Rangaka MX, Maartens G, et al. Novel relationship between tuberculosis immune reconstitution inflammatory syndrome and antitubercular drug resistance. Clin Infect Dis. 2009;48(5):667–76.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Colebunders R, John L, Huyst V, et al. Tuberculosis immune reconstitution inflammatory syndrome in countries with limited resources. Int J Tuberc Lung Dis. 2006;10(9):946–53.

    CAS  PubMed  Google Scholar 

  168. Knysz B, Kuliszkiewicz-Janus M, Jelen M, et al. Non-Hodgkin’s lymphoma as a rare manifestation of immune reconstitution disease in HIV-1 positive patients. Postepy Hig Med Dosw. 2006;60:547–51.

    Google Scholar 

  169. Narendran G, Swaminathan S. TB-HIV co-infection: a catastrophic comradeship. Oral Dis. 2016;22(Suppl 1):46–52.

    Article  PubMed  Google Scholar 

  170. Meintjes G, Wilkinson RJ, Morroni C, et al. Randomized placebo-controlled trial of prednisone for paradoxical tuberculosis-associated immune reconstitution inflammatory syndrome. AIDS. 2010;24(15):2381–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Brunel AS, Reynes J, Tuaillon E, et al. Thalidomide for steroid-dependent immune reconstitution inflammatory syndromes during AIDS. AIDS. 2012;26(16):2110–2.

    Article  PubMed  Google Scholar 

  172. Sierra-Madero JG, Ellenberg S, Rassool MS, et al. A randomized, double-blind, placebo-controlled clinical trial of a chemokine receptor 5 (CCR5) antagonist to decrease the occurrence of immune reconstitution inflammatory syndrome in HIV-infection: The CADIRIS Study. Lancet HIV. 2014;1(2):e60–7.

    Article  PubMed Central  Google Scholar 

  173. Knight RL, Allen DR, Birch HL, et al. Development of CXCR3 antagonists. Part 4: discovery of 2-amino-(4-tropinyl)quinolines. Bioorg Med Chem Lett. 2008;18(2):629–33.

    Article  CAS  PubMed  Google Scholar 

  174. Kanai T, Kamada N, Hisamatsu T. Clinical strategies for the blockade of IL-18 in inflammatory bowel diseases. Curr Drug Targets. 2013;14(12):1392–9.

    Article  CAS  PubMed  Google Scholar 

  175. Hennigan S, Kavanaugh A. Interleukin-6 inhibitors in the treatment of rheumatoid arthritis. Ther Clin Risk Manag. 2008;4(4):767–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Suntharalingam G, Perry MR, Ward S, et al. Cytokine storm in phase I trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med. 2006;355(10):1018–28.

    Article  CAS  PubMed  Google Scholar 

  177. de Jong BC, Israelski DM, Corbett EL, Small PM. Clinical management of tuberculosis in the context of HIV infection. Annu Rev Med. 2004;55:283–301.

    Article  CAS  PubMed  Google Scholar 

  178. DART Virology Group and Trial Team. Virological response to a triple nucleoside/nucleotide analogue regimen over 48 weeks in HIV-1-infected adults in Africa. AIDS. 2006;20:1391–9.

    Article  CAS  Google Scholar 

  179. Ribera E, Pou L, Lopez RM, et al. Pharmacokinetic interaction between nevirapine and rifampicin in HIV-infected patients with tuberculosis. J Acquir Immune Defic Syndr. 2001;28:450–3.

    Article  CAS  PubMed  Google Scholar 

  180. Ramachandran G, Hemanthkumar AK, Rajasekaran S, et al. Increasing nevirapine dose can overcome reduced bioavailability due to rifampicin co-administration. J Acquir Immune Defic Syndr. 2006;42:36–41.

    CAS  PubMed  Google Scholar 

  181. Manosuthi W, Ruxrungtham K, Likanonsakul S, et al. Nevirapine levels after discontinuation of rifampicin therapy and 60-week efficacy of nevirapine-based antiretroviral therapy in HIV-infected patients with tuberculosis. Clin Infect Dis. 2007;44:141–4.

    Article  CAS  PubMed  Google Scholar 

  182. Autar RS, Wit FW, Sankote J, et al. Nevirapine plasma concentrations and concomitant use of rifampin in patients co-infected with HIV-1 and tuberculosis. Antivir Ther. 2005;10:937–43.

    CAS  PubMed  Google Scholar 

  183. Ngaimisi E, Mugusi S, Minzi O, et al. Effect of rifampicin and CYP2B6 genotype on long-term efavirenz auto induction and plasma exposure in HIV patients with or without tuberculosis. Clin Pharmacol Ther. 2011;90:406–13.

    Article  CAS  PubMed  Google Scholar 

  184. Cohen K, Grant A, Dandara C, et al. Effect of rifampicin-based antitubercular therapy and the cytochrome P450 2B6 516G> T polymorphism on efavirenz concentrations in adults in South Africa. Antivir Ther. 2009;14:687–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Kakuda TN, Scholler-Gyure M, Hoetelmans RM. Pharmacokinetic interactions between etravirine and non-antiretroviral drugs. Clin Pharmacokinet. 2011;50:25–39.

    Article  CAS  PubMed  Google Scholar 

  186. [Anonymous]. Edurant package insert. © Janssen Products, LP 2011. http://www.edurant.com/sites/default/files/EDURANT-PI.pdf. Accessed Jun 2013.

  187. Decloedt EH, McIlleron H, Smith P, et al. Pharmacokinetics of lopinavir in HIV-infected adults receiving rifampin with adjusted doses of lopinavir-ritonavir tablets. Antimicrob Agents Chemother. 2011;55:3195–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Schmitt C, Riek M, Winters K, et al. Unexpected hepatotoxicity of rifampin and saquinavir/ritonavir in healthy male volunteers. Arch Drug Inf. 2009;2:8–16.

    Article  CAS  PubMed  Google Scholar 

  189. Haas DW, Koletar SL, Laughlin L, et al. Hepatotoxicity and gastrointestinal intolerance when healthy volunteers taking rifampin add twice-daily atazanavir and ritonavir. J Acquir Immune Defic Syndr. 2009;50:290–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Nijland HM, L’homme RF, Rongen GA, et al. High incidence of adverse events in healthy volunteers receiving rifampicin and adjusted doses of lopinavir/ritonavir tablets. AIDS. 2008;22:931–5.

    Article  CAS  PubMed  Google Scholar 

  191. Wenning LA, Hanley WD, Brainard DM, et al. Effect of rifampin, a potent inducer of drug-metabolizing enzymes, on the pharmacokinetics of raltegravir. Antimicrob Agents Chemother. 2009;53:2852–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Mena A, Vazquez P, Castro A, et al. Clinical experience of raltegravir-containing regimens in HIV-infected patients during rifampicin-containing treatment of tuberculosis. J Antimicrob Chemother. 2011;66:951–2.

    Article  CAS  PubMed  Google Scholar 

  193. Burger DM, Magis-Escurra C, van den Berk GE, et al. Pharmacokinetics of double-dose raltegravir in two patients with HIV infection and tuberculosis. AIDS. 2010;24:328–30.

    Article  PubMed  Google Scholar 

  194. Ramanathan S, Wang H, Stondell T, et al. Pharmacokinetics and drug interaction profile of cobicistat boosted-elvitegravir with atazanavir, rosuvastatin or rifabutin. Abstract O-03. 13th International Workshop on Clinical Pharmacology of HIV Therapy. Barcelona. 16–18 Apr 2012.

Download references

Authors’ contributions

GN, CP and ST prepared various sections of the manuscript. GN and ST compiled the final manuscript with valuable inputs from SS. All authors read and approved the final manuscript.

Acknowledgements

The authors wish to thank to Dr. Sanjay Mehendale MD, MPH, Director, NIRT and Dr. Mohan Natrajan HOD, Dept. of Clinical Research, National Institute for Research in Tuberculosis Chennai, the medical officers of TB sanatorium Tambaram, Chest physicians and ART medical officers of Rajiv Gandhi Government General hospital, Chennai for their valuable inputs, the staff of HIV and clinic, NIRT for their support and Mr. N. Santhanakumar for his excellent secretarial assistance.

Competing interests

The authors declare that they have no competing interests.

Consent for publication

Consent has been obtained from the patients whose images have been displayed.

Ethics approval and consent to participate

Not applicable as it is review of published literature and does not deal with individual patient or data.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Srikanth Tripathy.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gopalan, N., Chandrasekaran, P., Swaminathan, S. et al. Current trends and intricacies in the management of HIV-associated pulmonary tuberculosis. AIDS Res Ther 13, 34 (2016). https://doi.org/10.1186/s12981-016-0118-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12981-016-0118-7

Keywords