You are on page 1of 13

CHEST Special Features

GLOBAL MEDICINE

Relevance of Latent TB Infection in


Areas of High TB Prevalence
Surendra K. Sharma, MD, PhD; Sandeep Mohanan, MD; and Abhishek Sharma, MBBS

About one-third of the world population has latent TB infection (LTBI), the majority of which is
distributed in 22 high-burden countries. Early diagnosis and treatment of active TB remains the
top priority in resource-poor countries with high TB prevalence. Notwithstanding, because LTBI
contributes significantly to the pool of active TB cases later on, its diagnosis and treatment is essen-
tial, especially in high-risk groups. The lack of a gold standard and several limitations of currently
available tools, namely the tuberculin skin test and interferon-g release assays, are major con-
straints for LTBI diagnosis. In areas with high TB prevalence, interferon-g release assays have
not shown superiority over the conventional tuberculin skin test and are yet to be systematically
studied. Decisions regarding LTBI treatment with isoniazid preventive therapy should be made,
keeping in mind the high prevalence of isoniazid resistance in these settings. Although efforts
to shorten the LTBI treatment duration are encouraging, most trials have focused on adher-
ence and toxicity. Future trials on short-duration regimens in high-burden settings should address
drug efficacy issues as well. LTBI management, therefore, should comprise a targeted screen-
ing approach and individualization of LTBI treatment protocols. In addition, efforts should
focus on airborne infection control measures in high-burden countries. A high prevalence of
drug-resistant TB, the HIV epidemic, and delays in the diagnosis of active TB cases are other
major concerns in areas of high TB prevalence. There is ample space for further research in these
countries, whose outcomes may strengthen future national guidelines.
CHEST 2012; 142(3):761–773

Abbreviations: ART 5 antiretroviral therapy; BCG 5 Bacillus Calmette-Guérin; CDC 5 Centers for Disease Control
and Prevention; HBC 5 high-burden country; HCW 5 health-care worker; IGRA 5 interferon-g release assay; INH 5 iso-
niazid; IPT 5 isoniazid preventive therapy; LTBI 5 latent TB infection; MDR 5 multidrug resistant; Mtb 5 Mycobacterium
tuberculosis; NTM 5 nontubercular mycobacteria; TST 5 tuberculin skin test; WHO 5 World Health Organization

Latent TB infection (LTBI) is the presence of


Mycobacterium tuberculosis (Mtb) in an individ-
believed to have LTBI.1 This burden is distributed
largely among resource-poor countries. LTBI preva-
ual without clinical, imaging, or microbiologic evi- lence rates of up to 79% have been noted, with an
dence of active disease. Based on the tuberculin skin annual risk of TB infection ranging from 0.5% to
test (TST), which is also known as the Mantoux test, 14.3% among health-care workers (HCWs) in resource-
approximately one-third of the world population is limited countries.2 Poverty, malnutrition, tropical
infections, addictions, overcrowding, illiteracy, poorly
Manuscript received January 17, 2012; revision accepted May 11, functioning health-care systems, and political imbal-
2012. ance are important contributors to this situation.
Affiliations: From the Department of Internal Medicine
(Drs S. K. Sharma, Mohanan, and A. Sharma), All India Institute Worldwide, TB prevalence seems to be decreasing;
of Medical Sciences, New Delhi, India; and Medical University- about 12 million TB cases were estimated in 2010
Pleven (Dr A. Sharma), Pleven, Bulgaria. (178 cases per 100,000 population).1 According to the
Correspondence to: Surendra K. Sharma, MD, PhD, Depart-
ment of Medicine, All India Institute of Medical Sciences, New World Health Organization (WHO), 82% of this global
Delhi 110 029, India; e-mail: sksharma.aiims@gmail.com burden is distributed among 22 high-burden countries
© 2012 American College of Chest Physicians. Reproduction (HBCs).1 The highest prevalence was estimated in
of this article is prohibited without written permission from the
American College of Chest Physicians. See online for more details. African countries, such as South Africa and Congo
DOI: 10.1378/chest.12-0142 Republic, and southeast Asian countries, like Cambodia

journal.publications.chestnet.org CHEST / 142 / 3 / SEPTEMBER 2012 761

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


and Myanmar. India holds the global rank-one position associated with the susceptibility of developing TB
in the total estimated cases of TB (. 3.4 million), fol- have also been identified and include the natural
lowed by China, South Africa, Nigeria, and Indonesia. resistance-associated macrophage protein (NRAMP),
Although treatment of active TB is the overriding vitamin-D receptor (VDR), and nitric oxide synthase
priority in TB control, the diagnosis and treatment (NOS2A). Their polymorphisms may play a signifi-
of LTBI is equally important because it significantly cant role in populous HBCs.11
contributes to active cases later on. Several guide- Growing urbanization, explosive economic growth,
lines are available for LTBI management, but from and aggressive medical treatment in developing coun-
the priority point of view, these are applicable only tries have led to a surge in altered immunologic states
to countries with low TB prevalence. In areas with and diseases that multiply TB reactivation risk (Table 1).
high TB prevalence, the enormous number of sub- Added to the background environmental Mtb persis-
jects with LTBI who would have to be screened and tence, these tend to push the present goals of TB
treated would have to be balanced against the lim- control farther away. This review deals with the pre-
ited resources available for treating active TB cases. sent principles of LTBI diagnosis and treatment and
Repeated exogenous infections due to persistent their role in controlling the TB epidemic in HBCs.
mycobacterial sources in the environment along with
poor airborne infection control policies further com- Diagnosis of LTBI
plicate this situation. The administration of Bacillus
Calmette-Guérin (BCG) vaccination early in life, which Even as TB continues to intimidate the human race
so far has been the prime strategy, has done little to since time immemorial, the lack of a gold standard
solve this predicament. Thus, decisions regarding the test for LTBI diagnosis remains a major impediment
diagnosis and treatment of LTBI should address the in recognizing the population at risk. Contact investi-
various high-risk groups within the population. This gation promptly identifies new sources of environ-
is known as targeted screening, and it holds the key mental transmission and is the best method to isolate
in implementing focused therapy. In addition to the person at risk. This should ideally be incorporated
socioeconomic concerns, the presence of high-level into national programs, but inadequate finances and
background isoniazid (INH) resistance and several lack of trained human resources limit such reforms
uncertainties regarding diagnosis and treatment have in several countries. The TST, . 100 years old, is still
hampered formal synthesis of guidelines for LTBI the most widely used tool. Even though the advent of
management in resource-limited countries. interferon-g release assays (IGRAs) has shown promise
in overcoming several drawbacks of the TST,13 as its
utility unfolds, the presumed superiority of IGRAs seem
Immunopathogenesis and Risk Factors to diminish, particularly for high-TB-burden settings.
Various outcomes following exposure to Mtb are
Tuberculin Skin Test
depicted in Figure 1. In the general population, the
lifetime risk of progression from LTBI to active dis- TST quantifies the cell-mediated immune response
ease is about 5% to 10%; with HIV coinfection, this using a protein derivative of the bacilli to elicit delayed-
rises to 30% (annual risk, 10%).5 The pathogenic state type hypersensitivity. Table 2 shows criteria for inter-
of bacterial infection and probability of reactivation pretation of TST in various clinical settings. When
depend on the balance between host immunity and a smaller reaction is used as a cutoff for TST posi-
the influence of exogenous factors (Fig 2). It was tivity, sensitivity increases for those who are at high-
previously suggested that Mtb exists in a dormant est risk for reactivation TB. However, TST specificity
state during LTBI, which may later be resuscitated decreases with increased BCG vaccination coverage
to actively growing bacilli by certain resuscitation- and presence of environmental nontubercular myco-
promoting factors.7 However, perspectives propose a bacteria (NTM).
dynamic relationship, between a quiescent and an Currently, one-time BCG vaccination at birth is a
active state having bidirectional shifts, depending on routine practice of national programs in developing
the mycobacterial load and virulence, and the host’s countries. Although it is not possible to distinguish
immune mileu.8 This concept suggests that a bulk of between a tuberculin reaction that is caused by true
the so-called latent infection may actually be asymp- infection vs that caused by BCG, only 8% of subjects
tomatic active infection. Future research may help to who receive the BCG vaccine at birth have a positive
develop novel vaccines against LTBI persistence and TST result 15 years later.15 Accordingly, the history
reactivation.9 of BCG vaccination may be ignored when TST is
In addition to immune mechanisms, several latency- interpreted in adults.16 Similarly, the older the child,
related genes, such as a crystalline (acr) and s factor the lesser the probability for the reaction to be con-
(sigF), have been identified.10 Genetic determinants founded by prior BCG administration.

762 Special Features

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


Figure 1. Outcomes following exposure to Mycobacterium tuberculosis in an individual: scope for
interventions in TB disease control. Following environmental exposure with M tuberculosis, about 30% of
individuals become infected. Out of these, 5% to 10% progress to primary pulmonary TB, whereas the
rest (90%-95%) undergo containment but persist to LTBI because a balance strikes between protective
host immunity and bacterial virulence. The lifetime reactivation risk is about 10%, 20%,3,4 30%,5 and
40%3,4 for healthy adults, children aged , 5 years, people with HIV, and children aged , 2 years, respec-
tively. Various other high-risk clinical settings, such as administration of biologic agents or steroids,
chronic renal failure, diabetes mellitus, and so forth (see Table 1) tilt this balance in favor of increased
bacterial multiplication. The reactivation risk is greatest after the first 2 years of exposure. TB reactiva-
tion may express in the form of pulmonary, extrapulmonary, or miliary TB. Pulmonary TB cases again
perpetuate the cycle of environmental transmission. Thus, interventions to decrease global TB burden
can be considered at the following three levels: (1) airborne infection control, (2) intensive case finding
and treatment, and (3) diagnosis and treatment of latent infection. LTBI 5 latent TB infection.

Various studies conducted in India show NTM of the practice of boiling milk, although this may not
prevalence rates varying from 1% to 28%, with be the case in many African nations.
Mycobacterium fortuitum and Mycobacterium chelonae
being the most common isolates.17 Most individuals
Interferon-g Release Assays
harboring LTBI (due to Mtb) elicit a larger reaction
compared with that with NTM antigens, especially The IGRAs are a new class of in vitro assays that
when there is a history of contact. Sometimes, it may measure interferon-g released by effector T cells fol-
be difficult to distinguish the reactions induced by lowing stimulation with Mtb antigens (namely ESAT-6,
Mycobacterium bovis from Mtb. However, M bovis CFP-10, and TB7.7). The QuantiFERON-TB Gold
infections have become relatively rare in India because In-Tube (Cellestis) and the T-SPOT.TB (Oxford

journal.publications.chestnet.org CHEST / 142 / 3 / SEPTEMBER 2012 763

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


Figure 2. Immune mechanisms of containment and persistence of M tuberculosis. Following infection, dendritic cells prime T lympho-
cytes, which recruit and activate macrophages; this contains the initial phase of bacterial multiplication. Failure of this containment process
results in primary TB. Viable bacteria may persist either within focal lesions or outside a focal lesion. Within focal lesions, they may either
exist as sequestered foci or may be walled off by fibrosis and calcification. When outside focal lesions they are held in check by host immu-
nity. DC-SIGN 5 dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin; IFN-g 5 interferon-g; MHC 5 major
histocompatibility complex; TCR 5 T-cell antigen receptor; TLR-2 5 toll-like receptor 2; TNF-a 5 tumor necrosis factor-a. See Figure 1
legend for expansion of other abbreviation. (Reprinted with permission from Stewart et al.6)

Immunotec) tests are now widely used in the devel- detect recent as well as remote T-cell responses.
oped world. The growing economies of developing Dheda et al4 reported a pooled sensitivity of
nations have made them a lucrative market for com- about 77% for the QuantiFERON-Gold test to detect
mercial IGRAs. This has led to the indiscriminate use culture-confirmed TB, but when analyzed separately
of IGRAs, even for diagnosis of active TB, where they for high- and low-burden areas, it was 69% and 86%,
play no role.18,19 This should be highly discouraged respectively. In individuals with immunodeficiency,
because several controversies still exist regarding their the sensitivity of IGRAs decreases with higher degrees
operational value, such as their discordance with the of immunosuppression.25 A meta-analysis on the value
TST,20 role in immunocompromised subgroups,21 role of IGRAs in active pulmonary TB found pooled sen-
in HCWs,22 role of serial testing,23 and ability to iden- sitivities in the range of 84% to 88% for both the
tify people who are likely to progress to active TB.24 QuantiFERON-Gold and T-SPOT.TB tests in individ-
In high-burden settings, IGRAs tend to have uals without HIV, but the same decreased to 65% to
decreased sensitivity because of the confounding 68% in individuals with HIV infection18 (Figs 3, 4).
effects of malnutrition, NTM exposure (especially As noted earlier, Mtb persistence and reinfection
Mycobacterium kansasii and Mycobacterium marinum), is a major concern in IGRA interpretation. Likewise,
leprosy, and parasitic and other tropical infections strong IFN-g responses persisted even after 6 months
that may alter the host’s T helper 1/T helper 2 cell bal- of INH therapy in Indian HCWs,26 implying contin-
ance. Additionally, in such settings, IGRAs seem to uous nosocomial exposure, although high levels of
INH resistance may also contribute. This raises doubts
Table 1—Relative Risk of Reactivation TB in Various about the practical value of the currently recom-
Clinical Settings mended LTBI treatment regimen in high-burden
settings. Further, cost of IGRAs is another factor that
Clinical Setting Relative Risk
makes it less preferable to TST. Future studies should
AIDS 110-170 focus on improved versions of these tests that are
HIV infection 50-110 more user friendly and cost-effective. Table 3 com-
Solid organ transplantation (renal) 20-74
Silicosis 30
pares the utility of IGRAs and TST.
Jejunoileal bypass 27-63
Chronic renal failure on dialysis 10-25 TST, IGRA, or Both?
Carcinoma of head and neck 16
Abnormal chest radiograph with upper-lobe 6-19 Accumulated evidence at present suggests that in
fibronodular disease typical of healed TB infection HBCs, neither IGRAs nor TST accurately identify
TNF-a inhibitor therapy 1.7-9 people who will benefit from treatment. A systematic
Glucocorticoid therapy 4.9
review on their predictive value by Rangaka et al24
Children aged , 4 y 2.2-5
Diabetes mellitus 2-3.6 reported false-positive rates . 50% for both. Despite
Gastrectomy 2-5 its several limitations, the TST still remains a simple
Smoker (1 pack/d) 2-3 and cost-effective tool for both epidemiologic research
TNF-a 5 tumor necrosis factor-a. (Adapted with permission from and TB control. Accordingly, the latest European
Lobue and Menzies.12) Centre for Disease Prevention and Control guidelines32

764 Special Features

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


Table 2—Criteria for TST Positivity in Various TB prevalence, further evidence is required before
Clinical Settings this can be routinely recommended.
Width of Induration Positive Reaction

ⱖ 5 mm People with HIV infection Treatment of LTBI


Close contacts of patients with active TB
Patients with iatrogenic immunosuppressiona Before considering treatment of LTBI, a meticu-
Patients having fibrotic changes in chest lous assessment to rule out active TB is mandatory;
radiograph suggestive of old TB otherwise the patient may develop complications in
Chronic medical disease increasing the form of emergence of drug-resistant strains, dis-
susceptibility to infectionb
ease progression, and mortality. Many popular LTBI
ⱖ 10 mm Residents and staff of long-term-care
facilities regimens have been described in the past 2 decades,
Homeless people broadly, isoniazid preventive therapy (IPT) and short-
IV drug users course rifamycin-containing regimens (Table 4 ).
Health-care workers However, the overall efficacy of various regimens is
Recent TST conversion (increase by 10 mm thwarted by issues concerning adherence, duration
over past 2 y)
Children aged , 4 y
of protection, and drug toxicity.
ⱖ 15 mm People with no particular risk factors A Cochrane meta-analysis that examined the per-
Data from Reference 14. TST 5 tuberculin skin test.
formance of 6- and 12-month INH therapy (300 mg
aOrgan transplant recipients, people taking biologic agents or steroids daily) in people without HIV reported 60% effi-
(equivalent of 15 mg/d prednisolone for . 1 mo). cacy, which was only slightly higher for the 12-month
bPatients with silicosis, diabetes mellitus, chronic renal failure, lym-
regimen.39 One person was saved from developing
phoreticular/solid malignancies, or gastrectomy/ileojejunal bypass or active TB when 35 people were treated with INH for
with a weight loss of ⱖ 10% of ideal body weight.
6 months.39 However, a major practical difficulty for
IPT, especially among people with low socioeconomic
and a policy statement issued by the WHO19 dis- status, is poor adherence. It is important to under-
courage the use of IGRAs over TST, especially in stand that if proper adherence is ensured, 12-month
high-prevalence settings. A two-step strategy using a INH therapy is definitely more effective than 6-month
combination of TST and IGRA in sequence has been INH therapy.37 The WHO currently recommends
suggested by the National Institute for Health and IPT for LTBI treatment at a daily dose of 5 mg/kg
Clinical Excellence33 and European Centre for Disease (max 300 mg/kg) for at least 6 months and ideally
Prevention and Control32 guidelines, especially for for 9 months.40 Table 4 summarizes and compares
very-high-risk groups, such as those with HIV infec- short-course LTBI treatment regimens with IPT.
tion with low CD4 cell counts (, 200/mm), in low- The reported Three Months of Weekly Rifapentine
prevalence settings. However, for countries with high and Isoniazid for M Tuberculosis Infection study
(PREVENT TB) showed that a directly observed
weekly regimen of rifapentine (900 mg) and INH
(maximum dose 900 mg) for 12 weeks was noninfe-
rior to 9 months of IPT and had better adherence
rates than the latter.36 Accordingly, the Centers for
Disease Control and Prevention (CDC) revised its
treatment guidelines for people aged . 12 years in
countries with low and medium TB prevalence. Fur-
ther clinical trials are required to demonstrate the
efficacy and cost-effectiveness of this regimen for
high-prevalence settings.
It has long been argued that in developing coun-
tries, available resources should be conserved for
managing active TB. In populous nations like India
or China, the prospects of treating the majority of
the population over 6 or 9 months seem unfeasible
and nonproductive. In addition, high-prevalence rates
of INH resistance in these countries, ranging from
Figure 3. Pooled sensitivity of QFT-GIT and TSPOT among peo- 10% to 39%,41,42 preclude widespread use of INH
ple with HIV infection with confirmed active pulmonary TB from monotherapy for LTBI. Failure of IPT because of
various studies conducted in low- and middle-income countries.
QFT-GIT 5 QuantiFERON-TB Gold In-Tube; TSPOT 5 T-SPOT. the emergence of drug-resistant strains has been
TB. (Reprinted with permission from Metcalfe et al.18) reported.43 However, the fear of emergence of drug

journal.publications.chestnet.org CHEST / 142 / 3 / SEPTEMBER 2012 765

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


In addition, 6-month IPT had only a limited role
compared with extended therapy for 36 months in
the high-TB-burden setting of Botswana among indi-
viduals with HIV.47 This again emphasizes the priority
of individual TB case management and environmen-
tal infection control above LTBI treatment in resource-
limited countries. Once the decision to treat LTBI
has been made, the choice of regimen has to be indi-
vidualized based on toxicity risk, pill burden, and
clinical setting. Effective information, education, and
counseling activities along with directly observed pre-
ventive therapy48 may help to achieve higher comple-
tion rates.

Pertinent Issues in Endemic Countries


LTBI and the HIV Epidemic
Figure 4. Pooled sensitivity of QFT-GIT and TSPOT among Globally, about 13% of TB cases occur among
people without HIV infection with confirmed active pulmonary
TB from various studies conducted in low- and middle-income people with HIV (. 50% in some African countries).1
countries. See Figure 3 legend for expansion of abbreviations. Figure 5 illustrates global TB-HIV coinfection rates.
(Reprinted with permission from Metcalfe et al.18) People with HIV infection and Mtb have a roughly
30% lifetime reactivation risk, and this increases with
resistance should not hamper the administration of disease progression.5 This is further complicated by
IPT to high-risk groups provided that the importance late presentation and significant delay in accessing
of ruling out active TB and continued drug resis- antiretroviral therapy (ART) in resource-limited coun-
tance surveillance is emphasized.44,45 Further, sys- tries. Therefore, in the HBCs, prevention of TB is
tematic background INH resistance mapping should sine qua non to reduce the morbidity and mortality
be carried out in these countries. attributed to HIV infection. Exogenous infection
Although definite data on the duration of effective- plays a significant role in HIV-associated TB in set-
ness of LTBI treatment are lacking, risk of reexposure tings of high TB prevalence.49 Continuous environ-
and degree of immunosuppression are major deter- mental exposure to Mtb also increases the chance of
minants of the waning protection with time. A study TB reactivation by increasing the total host mycobac-
in the setting of high TB-HIV prevalence in Zambia terial load.50 Unfortunately, only DNA fingerprinting
revealed durability of protection for at least 2.5 years.46 technology can distinguish reinfection vs reactivation

Table 3—Comparison of the Utility of TST and IGRAs in Areas of High TB Endemicity

Characteristics TST IGRA


Sensitivity4,24,27,28, a 72%-100% QFT-GIT test, 69%
T-SPOT.TB test, 72%-90%
Specificity24,27, a,b 41%-97% 50%-99%
Quantity of evidence Abundant Fewer
Material costs Inexpensive Costly
Reporting Patient needs to return after 2 d Single visit
Interpretation Subjective to technique and observer bias Yes/no answer (may also be indeterminate)
Predictive value for reactivation TB Poor Poor
Resource input Minimum laboratory equipment, but manpower Requires laboratory expertise, expensive
and cold storage required equipment, and phlebotomy facilities
HIV population29 Overall sensitivity less than that for Sensitivity wanes with the progression of
immunocompetent population disease; overall performance is similar to TST
Pediatric population30,31 More sensitive, preferred Less-sensitive and more-indeterminate results
IGRA 5 interferon-g release assay; LTBI 5 latent TB infection; QFT-GIT 5 QuantiFERON-TB Gold In-Tube. See Table 2 legend for expansion of
other abbreviation.
aData are based on comparison studies among active TB cases. Absolute sensitivity and specificity for LTBI cannot be obtained in the absence

of a gold standard. TST sensitivity is higher for a lower cutoff for positivity.
bSpecificity tends to be relatively lower for TST in the Bacillus Calmette-Guérin-vaccinated cohort in low-prevalence countries. In adults from

high-burden countries, the specificity of TST tends to be higher (due to waning Bacillus Calmette-Guérin effect), and that of IGRAs tends to be
lower because of repeated infections.

766 Special Features

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


Table 4—Summary of Various Popular Regimens That Have Been Used for LTBI Treatment

Regimen Efficacy Adherence Incidence of Drug-Induced Hepatitis


Daily H: 6 mo 69% 50% 1%-5%
Daily H: 9-12 mo 90%-93% , 50% 1%-5%
Daily RH: 3-4 mo Equal to 6-mo H Slightly better (by 6%) than 9- to 12-mo H 1%-5%
Daily RZa: 2 mo Equal to 9- to 12-mo H Slightly better (by 6%) than 9- to 12-mo H 3%-5%
Daily R: 3-4 mo34 65% Much better (by 22%) than 9- to 12-mo H , 1%
Twice-weekly RH: 3 mo35, b Equal to 6-mo H 95% 2.4%
Weekly RH: 3 mo35,36, b Equal to 9-mo H 82%-96% 0.4%-1.5%
H 5 isoniazid 5 mg/kg; R 5 rifampicin 10 mg/kg; Z 5 pyrazinamide. See Table 3 legend for expansion of other abbreviation. (Adapted with permis-
sion from Menzies et al.37)
aRZ regimen is no longer recommended because of high incidence of drug toxicity.38

bIn the weekly and twice-weekly regimens, H and R were used at doses of 900 mg each.35

TB and is not easily available in resource-limited set- in the risk of developing TB with any LTBI treatment
tings.51 Several issues complicate TB diagnosis in the regimen, which increased to 62% when the TST result
setting of HIV coinfection, which can also create con- was positive.60 However, IPT was found to be rela-
fusion in differentiating latent from active disease tively safer than the combination regimens. Significant
(Table 5). pharmacokinetic drug interactions and additive toxic-
The influence of HIV disease stage on the efficacy ities with ART preclude the use of rifamycin-containing
of LTBI treatment is still not known and warrants combination regimens as the first choice for LTBI
further study. The recommendation of WHO to ini- treatment. Accordingly, current WHO guidelines
tiate ART at a CD4 cell count of 350/mm3 may signif- for resource-limited countries strongly recommend
icantly bring down TB reactivation rates by restoring at least 6-month INH therapy for both adults and
immunity at an earlier stage of the disease. Resource- children, regardless of TST, after excluding active TB.45
limited countries need to undertake IPT feasibility It also conditionally recommends a 36-month IPT
studies to evaluate the impact of this recommendation. regimen in high-prevalence settings, although adher-
Nevertheless, a study conducted in Brazil reported ence issues may interfere with treatment completion.
efficacy of IPT, regardless of the CD4 cell count, in Among people with HIV, IPT was found to reduce
addition to the protective effect offered by ART.56 the risk of reactivation by 82% when given immedi-
Unlike the situation in low-prevalence countries, ately after cure of a first TB episode61 and by 55%
studies among individuals with HIV in high-burden when given for a history of cured TB.62 Accordingly,
settings have shown poor concordance between the WHO guidelines advocate secondary prevention with
TST and IGRAs, with only modest predictive values INH for patients with HIV who have successfully
for either.29,57 Increased number of indeterminate completed their TB treatment.45
test results and reduced sensitivity at lower CD4 cell The fear of developing drug-resistant TB is one of
counts were reported with IGRAs.25 Among the two the arguments cited for not offering INH for latent
commercially available kits, a higher sensitivity was TB-HIV coinfection in endemic settings. However,
reported with T-SPOT.TB (about 72%) than with the this was not noted in a large prospective study from
QuantiFERON-TB Gold In-Tube (about 61%).29
Table 6 describes the major lacunae identified in the
interpretation of IGRAs in the population with HIV
infection. Based on available evidence, IGRAs cur-
rently cannot be recommended over TST to screen
people living with HIV for eligibility to receive IPT
in high-TB-prevalence areas.19
For intensified case finding, WHO recognizes that
the absence of four symptoms, namely fever, current
cough, night sweats, and weight loss, has a nega-
tive predictive value of 97.7% for active TB in high
TB-HIV coinfection areas and that such patients can
be safely treated for LTBI.45,59 This holds true irre-
spective of the degree of immunosuppression, people
receiving ART, people with a history of TB, and preg-
nant women.59 A Cochrane meta-analysis among Figure 5. Prevalence of HIV in new TB cases reported in 2010.
patients with HIV infection revealed a 32% reduction (Reprinted with permission from the World Health Organization.1)

journal.publications.chestnet.org CHEST / 142 / 3 / SEPTEMBER 2012 767

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


Table 5—Difficulties in the Diagnosis of TB in People of active TB in household contacts of patients with
With TB-HIV Coinfection in Endemic Countries MDR-TB range from 1.9% in adults to up to 11.2%
Characteristic Reference in children.67,69,70 The Revised National TB Control
Program of India advocates early diagnosis and treat-
Increased prevalence of asymptomatic active TB that 52
may preclude symptom-based diagnosis
ment in symptomatic contacts of patients with MDR-TB
Increased rates of occult disseminated TB that may 53 as an important element of TB prevention. However,
cause gross underestimation of the true prevalence the insurgence of extensively drug-resistant strains
Higher incidence of extrapulmonary TB compared 54,55 and reports on the appearance of totally drug-resistant
with the non-HIV population strains are further blows to TB-control measures.
More frequent sputum smear-negative active TB cases 55
Higher proportion of TST-negative TB infection 54
A paucity of evidence-based guidelines for the
treatment of LTBI in MDR-TB contacts is very much
See Table 2 legend for expansion of abbreviation.
acknowledged. A careful evaluation to assess the
reactivation risk should be made, and a combination
South Africa63 and, therefore, should not be a hin- regimen should be started, considering the resistance
drance to the use of IPT in developing nations.45 In profile of the suspected Mtb isolates in question, for
countries with high prevalence of INH resistance, 6 to 12 months. LTBI-proven contacts of INH mono-
IPT provision may have to be based on regional resis- resistant TB have been successfully treated with the
tance mapping, as mentioned previously. In response 4-month rifampicin regimen.16,34 Among the fluoro-
to the dual epidemic, the WHO highlights interven- quinolones, moxifloxacin is a promising drug for the
tions like ART provision and the three Is for HIV/TB, treatment of both active TB infection and LTBI.71
namely intensified TB case finding, infection control, Unfortunately, their unrestricted use for community-
and IPT, as part of prevention, care, and treatment acquired pneumonia has facilitated the entry of drug-
services.64 With the current scale-up of ART services, resistant strains by inadvertent monotherapy for
future trials on LTBI treatment should assess the unsuspected TB.72 In high-prevalence countries, flu-
duration of protection, cost-effectiveness, and value oroquinolone use should be safeguarded through
of combination with ART. strict regulatory policies to prevent weakening of
the drug arsenal. Newer promising molecules, such as
LTBI and Drug Resistance TMC-207, SQ109, PA-824, and FAS20013, have potent
Globally, about 3.6% of incident TB cases are mul- in vitro bactericidal action against MDR-TB and latent
tidrug resistant (MDR) (ie, resistant to INH and TB isolates.38,51 These molecules are being evaluated
rifampicin), one-half of which are estimated to occur in phase 1 and 2 clinical trials but have a long way to
in China and India.65 Estimates in India reveal that go before they see daylight in developing nations.
up to 20% of patients with TB who are retreated, have
relapsed, or have previously defaulted are infected LTBI in Children
with MDR isolates.66 Household contacts of patients
with MDR TB are at high risk of infection, particularly Childhood TB has been neglected in national TB
in the first 2 years.67 A prospective study from Peru control programs for a long time. In high-burden set-
observed 4,500 household contacts of patients with tings, reactivation rates of up to 40% are observed
MDR-TB and revealed the high probability of devel- in children aged , 2 years, with most occurring in
oping MDR-TB over a period of 4 years.68 This exposes the first year following infection.4 The importance of
limitations of current guidelines on the duration of LTBI diagnosis and treatment in this cohort, there-
contact monitoring in this scenario. The prevalence fore, should be acknowledged in view of children’s
developing immune systems, especially for those
aged ⱕ 5 years.33 In children, IGRAs perform poorly
Table 6—Knowledge Gaps Restricting the Use of IGRAs
in the Diagnosis of TB-HIV Coinfection in terms of sensitivity30 and provide more indeter-
minate results,31 and the specificity is not better for a
Knowledge Gap higher cutoff for a positive TST result.30 In those who
Accuracy and reliability for the diagnosis of LTBI received BCG at birth, . 50% will have a negative
Value in predicting who to treat TST result at age 9 to 12 months and almost all will
Identification of cutoff points for various ranges of CD4 cell counts be negative by 5 years of age.73 Currently, TST is
Prognostic ability to predict progression from latent to active disease
the more-valuable and cost-effective tool for LTBI
Role in monitoring response to treatment in latent and active disease
Feasibility, acceptability, and cost-effectiveness diagnosis in children living in high-prevalence coun-
Role in point-of-care setting compared with TST for diagnosing tries, regardless of BCG status,74,75 and should not be
latent disease replaced by IGRAs.19
Data from Pai et al.58 See Table 2-4 legends for expansion of The CDC,76 WHO,44 and National Institute for
abbreviations. Health and Clinical Excellence30 guidelines recommend

768 Special Features

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


at least 6 months of IPT (10 mg/kg daily) for children three-level hierarchy: (1) administrative control,
with LTBI. However, a trial failed to reveal signifi- (2) environmental control, and (3) respiratory protec-
cant survival benefits in children with HIV coinfec- tion control.76 Administrative control is the prompt
tion.77 The Revised National TB Control Program identification, segregation, triaging, and case manage-
guidelines in India recommend treatment of all close ment at the facility level, a prerequisite for further
household contacts aged , 6 years with 6 months of control measures. Environmental (airborne infection)
IPT (5 mg/kg daily), regardless of BCG vaccination control works on the principle of dilution of infectious
status.78 However, operational analyses in develop- particles through natural or mechanical methods. Sug-
ing countries have revealed that less than one-fourth gested basic recommendations are summarized in
of the deserving pediatric population receives screen- Table 8. Additionally, future design of health-care
ing and IPT services.79,80 This must be dealt with strict facilities should consider patient flow patterns and
surveillance protocols and education of field-level isolation strategies to minimize the spread of airborne
staff and program managers in national TB-control nosocomial infections. Particular attention should be
programs. given to facilities that provide integrated TB-HIV
management services and to those that deal with
Relevance of Airborne Infection Control drug-resistant TB.90 Respiratory protection devices,
Measures to LTBI Management pertaining mainly to people who are at highest risk
(laboratory staff and various categories of health-care
Rapid expansion of medical therapeutics in devel- providers [eg, ICU staff, bronchoscope operators,
oping countries has greatly amplified the susceptible thoracic surgeons]), include N95 and FFP2 masks.
population for reactivation TB. HCWs from endemic Individual guidelines on airborne infection control
countries were found to have an annual risk of TB have now come up in several developing nations.90
infection of 8.4% (range, 2.7%-14%).81 In addition, Political commitment is essential to plan, control, and
the prevalence of diseases impairing host immunity, supervise these measures at the community level for
such as diabetes mellitus and chronic renal disease, a good TB control program.
are ever increasing. However, routine screening and
treatment of LTBI is not currently advisable in HBCs,
especially because of the large population they repre- Conclusion
sent and the increased risks for drug toxicity. Hence,
decisions regarding LTBI treatment should be indi- In developing countries, the problem of high TB
vidualized until large-scale studies have conclusively burden sometimes eclipses the real issues of poor
shown favorable results in specific risk groups. A reliability of diagnostic tools and treatment-related
summary of evidence for the diagnosis and treatment concerns. Delayed diagnosis of active TB cases and
of LTBI in various iatrogenic risk groups appears in the emergence of MDR-TB strains are additional
Table 7. issues. The moral argument that every infected person
The overwhelming danger of reinfection and mul- deserves treatment, just as in other chronic diseases
tiplying risk groups underscore the need for com- like HIV and hypertension, may be overshadowed by
mensurate airborne infection control measures. To the real problem of scarce resources, but this matter of
tackle this, the CDC has proposed the following contention seems largely one of political commitment

Table 7—Evidence-Based Updates on the Diagnosis and Management of LTBI in Selected High-Risk Groups

Risk Factor Diagnostic Protocola Treatment Regimen


Organ transplantation b82,83 TST and IGRAs perform similarly 9- to 12-mo H
Hemodialysis16,84,85 IGRAs preferred to TST (two -step testing may be advisable) 9-mo H
Biologic therapyc86-88 IGRAs preferred to TST (TST if effect of BCG is minor) Preferred, 9- to 12-mo H; alternative, 3-mo HR
Health-care workersd23,77 No evidence for superiority of either TST or IGRA, but Preferred, 9-mo H; alternatives, 6-mo H or 4-mo R
one-time IGRA may prevent excess false positives and
thus overtreatment in low burden settings
BCG 5 Bacillus Calmette-Guérin. See Table 1-4 legends for expansion of other abbreviations.
aA person is considered to be infected if he or she has a history of TB or if the chest radiograph shows suggestive infiltrates despite negative
immunologic test results. Recommendations supporting IGRAs are based primarily on studies conducted in low-burden countries and cannot be
extrapolated to high-burden settings where IGRAs, in general, have not been shown to be superior to TST. Large-scale studies that are conducted
exclusively in these selected risk groups may reveal their exact role in endemic settings.
bIncreased toxicity has been reported with the H regimen following liver transplantation.83,84

cBiologic agents include anti-TNF-a therapy, especially infliximab, and monoclonal antibodies against various immune mediators, which are used

against inflammatory diseases or malignancies.


dSerial testing for health-care workers is not advised in high-prevalence settings.23

journal.publications.chestnet.org CHEST / 142 / 3 / SEPTEMBER 2012 769

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


Table 8—Basic Recommendations on Environmental human immunodeficiency virus infection. N Engl J Med.
Control of Airborne Infection 1989;320(9):545-550.
6. Stewart GR, Robertson BD, Young DB. Tuberculosis: a
Recommendation problem with persistence. Nat Rev Microbiol. 2003;1(2):
97-105.
Adequate natural ventilation in all patient-care zones of health-care
7. Biketov S, Potapov V, Ganina E, Downing K, Kana BD,
facilities should exist.
Kaprelyants A. The role of resuscitation promoting factors in
Requirements for natural ventilation are an hourly average
pathogenesis and reactivation of Mycobacterium tuberculosis
ventilation rate of the following:
during intra-peritoneal infection in mice. BMC Infect Dis.
160 L/s per patient for isolation rooms (minimum of 80 L/s
2007;7:146.
per patient)
8. Barry CE III, Boshoff HI, Dartois V, et al. The spectrum of
60 L/s per patient for general wards and outpatient departments
latent tuberculosis: rethinking the biology and intervention
2.5 L/s/m3 for corridors and spaces not used for fixed patient care
strategies. Nat Rev Microbiol. 2009;7(12):845-855.
Alternative modes such as hybrid (mixed-mode) ventilation,
9. Commandeur S, van Meijgaarden KE, Lin MY, et al. Iden-
mechanical ventilation, ultraviolet germicidal irradiation
tification of human T-cell responses to Mycobacterium tuber-
devices, and use of HEPA filters should be considered
culosis resuscitation-promoting factors in long-term latently
when natural ventilation is inadequate.
infected individuals. Clin Vaccine Immunol . 2011;18(4):
The airflow should start from the source and be directed outdoors.
676-683.
Data from Atkinson et al.89 HEPA 5 high-efficiency particulate air. 10. Parrish NM, Dick JD, Bishai WR. Mechanisms of latency
in Mycobacterium tuberculosis. Trends Microbiol. 1998;6(3):
107-112.
rather than one purely of finances. Nevertheless, eco- 11. Dye C, Williams BG. The population dynamics and control
nomic evaluations are necessary to justify the diver- of tuberculosis. Science. 2010;328(5980):856-861.
sion of money and trained human resources. Despite 12. Lobue P, Menzies D. Treatment of latent tuberculosis infec-
these obstacles, the success seen among individuals tion: An update. Respirology. 2010;15(4):603-622.
13. Menzies D, Pai M, Comstock G. Meta-analysis: new tests for
with HIV infection suggest that targeted screening the diagnosis of latent tuberculosis infection: areas of uncer-
and therapy, especially for young children and other tainty and recommendations for research. Ann Intern Med.
high-risk groups, which have so far been ignored, 2007;146(5):340-354.
should be pursued. Rather than comprehensive cov- 14. Screening for tuberculosis and tuberculosis infection in high-
erage, individualization of treatment protocols would risk populations. Recommendations of the Advisory Council
for the Elimination of Tuberculosis. MMWR Recomm Rep.
be a step in the right direction. However, prudence 1995;44(RR-11):19-34.
should be maintained to ensure that LTBI treatment 15. Menzies R, Vissandjee B. Effect of Bacille Calmette-Guérin
does not divert resources from treatment of active TB vaccination on tuberculin reactivity. Am Rev Respir Dis.
or from offering ART services to needy patients. The 1992;145(3):621-625.
need for systematic trials focusing on high-risk groups 16. American Thoracic Society. Targeted tuberculin testing and
treatment of latent tuberculosis infection. This official state-
in resource-limited settings is very much felt, and this ment of the American Thoracic Society was adopted by the
may help to formulate clear-cut individual national ATS Board of Directors, July 1999. This is a Joint Statement
guidelines on LTBI management in the future. of the American Thoracic Society (ATS) and the Centers for
Disease Control and Prevention (CDC). This statement was
endorsed by the Council of the Infectious Diseases Society
Acknowledgments of America. (IDSA), September 1999, and the sections of
this statement. Am J Respir Crit Care Med. 2000;161(4 pt 2):
Financial/nonfinancial disclosures: The authors have reported S221-S247.
to CHEST that no potential conflicts of interest exist with any
companies/organizations whose products or services may be dis- 17. Chakrabarti A, Sharma M, Dubey ML. Isolation rates of dif-
cussed in this article. ferent mycobacterial species from Chandigarh (north India).
Indian J Med Res. 1990;91:111-114.
18. Metcalfe JZ, Everett CK, Steingart KR, et al. Interferon-g
release assays for active pulmonary tuberculosis diagnosis
References in adults in low- and middle-income countries: systematic
1. World Health Organization. WHO Report 2011: Global review and meta-analysis. J Infect Dis. 2011;204(suppl 4):
Tuberculosis Control. World Health Organization website. S1120-S1129.
http://www.who.int/tb/publications/global_report/2011/gtbr11_ 19. World Health Organization. Use of Tuberculosis Interferon-
main.pdf. Accessed May 27, 2012. Gamma Release Assays (IGRAs) in Low- and Middle-Income
2. Joshi R, Reingold AL, Menzies D, Pai M. Tuberculosis among Countries: Policy Statement. Publication WHO/HTM/TB/2011.
health-care workers in low- and middle-income countries: 18. Geneva, Switzerland: World Health Organization; 2011.
a systematic review. PLoS Med. 2006;3(12):e494. http://whqlibdoc.who.int/publications/2011/9789241502672_
3. Brailey M. Mortality in the children of tuberculous house- eng.pdf. Accessed May 27, 2012.
holds. Am J Public Health Nations Health. 1940;30(7):816-823. 20. Hill PC, Brookes RH, Fox A, et al. Large-scale evaluation of
4. Dheda K, van Zyl Smit R, Badri M, Pai M. T-cell interferon- enzyme-linked immunospot assay and skin test for diagnosis
gamma release assays for the rapid immunodiagnosis of of Mycobacterium tuberculosis infection against a gradient of
tuberculosis: clinical utility in high-burden vs. low-burden exposure in The Gambia. Clin Infect Dis. 2004;38(7):966-973.
settings. Curr Opin Pulm Med. 2009;15(3):188-200. 21. Pai M, Lewinsohn DM. Interferon-gamma assays for tuber-
5. Selwyn PA, Hartel D, Lewis VA, et al. A prospective study of culosis: is anergy the Achilles’ heel? Am J Respir Crit Care
the risk of tuberculosis among intravenous drug users with Med. 2005;172(5):519-521.

770 Special Features

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


22. Harada N, Nakajima Y, Higuchi K, Sekiya Y, Rothel J, 37. Menzies D, Al Jahdali H, Al Otaibi B. Recent developments
Mori T. Screening for tuberculosis infection using whole- in treatment of latent tuberculosis infection. Indian J Med
blood interferon-gamma and Mantoux testing among Japanese Res. 2011;133:257-266.
healthcare workers. Infect Control Hosp Epidemiol. 2006; 38. Zhang T, Li SY, Williams KN, Andries K, Nuermberger EL.
27(5):442-448. Short-course chemotherapy with TMC207 and rifapentine
23. Zwerling A, van den Hof S, Scholten J, Cobelens F, Menzies D, in a murine model of latent tuberculosis infection. Am J
Pai M. Interferon-gamma release assays for tuberculosis Respir Crit Care Med. 2011;184(6):732-737.
screening of healthcare workers: a systematic review. Thorax. 39. Smieja MJ, Marchetti CA, Cook DJ, Smaill FM. Isoniazid
2012;67(1):62-70. for preventing tuberculosis in non-HIV infected persons.
24. Rangaka MX, Wilkinson KA, Glynn JR, et al. Predictive value Cochrane Database Syst Rev. 2000; (2):CD001363.
of interferon-g release assays for incident active tuberculo- 40. World Health Organization. Implementing the WHO Stop
sis: a systematic review and meta-analysis. Lancet Infect Dis. TB Strategy: A Handbook for National Tuberculosis Control
2012;12(1):45-55. Programmes. Publication WHO/HTM/TB/2008.401. Geneva,
25. Aabye MG, Ravn P, PrayGod G, et al. The impact of HIV Switzerland: World Health Organization; 2008. http://
infection and CD4 cell count on the performance of an inter- whqlibdoc.who.int/publications/2008/9789241546676_eng.
feron gamma release assay in patients with pulmonary tuber- pdf. Accessed May 28, 2012.
culosis. PLoS ONE. 2009;4(1):e4220. 41. Singh P, Katoch VM. Multi-drug resistant tuberculosis: cur-
26. Pai M, Joshi R, Dogra S, et al. Persistently elevated T cell rent status and emerging tools for its management in India.
interferon-gamma responses after treatment for latent tuber- J Commun Dis. 2006;38(3):216-229.
culosis infection among health care workers in India: a pre- 42. He GX, Zhao YL, Jiang GL, et al. Prevalence of tuberculosis
liminary report. J Occup Med Toxicol. 2006;1:7. drug resistance in 10 provinces of China. BMC Infect Dis.
27. Pai M, Zwerling A, Menzies D. Systematic review: T-cell- 2008;8:166.
based assays for the diagnosis of latent tuberculosis infection: 43. Fernández-Guerrero ML, Esteban J, Acebes C, Górgolas M.
an update. Ann Intern Med. 2008;149(3):177-184. Failure of isoniazid chemoprophylaxis during infliximab
28. Jackson-Sillah D, Hill PC, Fox A, et al. Screening for tuber- therapy. Emerg Infect Dis. 2007;13(9):1428-1429.
culosis among 2381 household contacts of sputum-smear- 44. Balcells ME, Thomas SL, Godfrey-Faussett P, Grant AD.
positive cases in The Gambia. Trans R Soc Trop Med Hyg. Isoniazid preventive therapy and risk for resistant tuberculo-
2007;101(6):594-601. sis. Emerg Infect Dis. 2006;12(5):744-751.
29. Cattamanchi A, Smith R, Steingart KR, et al. Interferon- 45. World Health Organization, Department of HIV/AIDS, Stop
gamma release assays for the diagnosis of latent tuberculo- TB Department. Guidelines for Intensified Tuberculosis
sis infection in HIV-infected individuals: a systematic review Case-Finding and Isoniazid Preventive Therapy for People
and meta-analysis. J Acquir Immune Defic Syndr. 2011;56(3): Living With HIV in Resource-Constrained Settings. Geneva,
230-238. Switzerland: World Health Organization; 2011. http://whqlibdoc.
30. Machingaidze S, Wiysonge CS, Gonzalez-Angulo Y, et al. who.int/publications/2011/9789241500708_eng.pdf. Accessed
The utility of an interferon gamma release assay for diagno- May 28, 2012.
sis of latent tuberculosis infection and disease in children: 46. Quigley MA, Mwinga A, Hosp M, et al. Long-term effect
a systematic review and meta-analysis. Pediatr Infect Dis J. of preventive therapy for tuberculosis in a cohort of HIV-
2011;30(8):694-700. infected Zambian adults. AIDS. 2001;15(2):215-222.
31. Connell TG, Curtis N, Ranganathan SC, Buttery JP. Perfor- 47. Samandari T, Agizew TB, Nyirenda S, et al. 6-month versus
mance of a whole blood interferon gamma assay for detecting 36-month isoniazid preventive treatment for tuberculosis
latent infection with Mycobacterium tuberculosis in children. in adults with HIV infection in Botswana: a randomised,
Thorax. 2006;61(7):616-620. double-blind, placebo-controlled trial. Lancet. 2011;377(9777):
32. European Centre for Disease Prevention and Control. 1588-1598.
Use of Interferon-Gamma Release Assays in Support of 48. Hirsch-Moverman Y, Daftary A, Franks J, Colson PW.
TB Diagnosis. Stockholm, Sweden: European Centre for Adherence to treatment for latent tuberculosis infection:
Disease Prevention and Control; 2011. http://ecdc.europa.eu/ systematic review of studies in the US and Canada. Int J
en/publications/Publications/1103_GUI_IGRA.pdf. Accessed Tuberc Lung Dis. 2008;12(11):1235-1254.
May 28, 2012. 49. Houben RM, Crampin AC, Ndhlovu R, et al. Human immu-
33. National Collaborating Centre for Chronic Conditions and nodeficiency virus associated tuberculosis more often due to
the Centre for Clinical Practice at NICE. Tuberculosis: recent infection than reactivation of latent infection. Int J
Clinical Diagnosis and Management of Tuberculosis, and Tuberc Lung Dis. 2011;15(1):24-31.
Measures for Its Prevention and Control. NICE clinical 50. Wood R, Liang H, Wu H, et al. Changing prevalence of
guideline 117. London, England: Royal College of Physicians tuberculosis infection with increasing age in high-burden
of London; 2011. http://www.nice.org.uk/nicemedia/live/ townships in South Africa. Int J Tuberc Lung Dis. 2010;14(4):
13422/53638/53638.pdf. Accessed May 28, 2012. 406-412.
34. Menzies D, Long R, Trajman A, et al. Adverse events with 51. Lienhardt C, Vernon A, Raviglione MC. New drugs and new
4 months of rifampin therapy or 9 months of isoniazid therapy regimens for the treatment of tuberculosis: review of the
for latent tuberculosis infection: a randomized trial. Ann drug development pipeline and implications for national
Intern Med. 2008;149(10):689-697. programmes. Curr Opin Pulm Med. 2010;16(3):186-193.
35. Martinson NA, Barnes GL, Moulton LH, et al. New regimens 52. Kranzer K, Houben RM, Glynn JR, Bekker LG, Wood R,
to prevent tuberculosis in adults with HIV infection. N Engl Lawn SD. Yield of HIV-associated tuberculosis during
J Med. 2011;365(1):11-20. intensified case finding in resource-limited settings: a sys-
36. Sterling TR, Villarino ME, Borisov AS, et al; TB Trials tematic review and meta-analysis. Lancet Infect Dis. 2010;
Consortium PREVENT TB Study Team. Three months of 10(2):93-102.
rifapentine and isoniazid for latent tuberculosis infection. 53. Ansari NA, Kombe AH, Kenyon TA, et al. Pathology and
N Engl J Med. 2011;365(23):2155-2166. causes of death in a group of 128 predominantly HIV-positive

journal.publications.chestnet.org CHEST / 142 / 3 / SEPTEMBER 2012 771

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


patients in Botswana, 1997-1998. Int J Tuberc Lung Dis. adult multidrug-resistant pulmonary tuberculosis: a 30-month
2002;6(1):55-63. follow-up. Pediatrics. 2002;109(5):765-771.
54. Elliott AM, Halwiindi B, Hayes RJ, et al. The impact of 70. Attamna A, Chemtob D, Attamna S, et al. Risk of tuberculo-
human immunodeficiency virus on presentation and diagno- sis in close contacts of patients with multidrug resistant tuber-
sis of tuberculosis in a cohort study in Zambia. J Trop Med culosis: a nationwide cohort. Thorax. 2009;64(3):271.
Hyg. 1993;96(1):1-11. 71. Nuermberger E, Tyagi S, Williams KN, Rosenthal I, Bishai WR,
55. Getahun H, Harrington M, O’Brien R, Nunn P. Diagnosis Grosset JH. Rifapentine, moxifloxacin, or DNA vaccine
of smear-negative pulmonary tuberculosis in people with HIV improves treatment of latent tuberculosis in a mouse model.
infection or AIDS in resource-constrained settings: informing Am J Respir Crit Care Med. 2005;172(11):1452-1456.
urgent policy changes. Lancet. 2007;369(9578):2042-2049. 72. Drlica K, Zhao X, Kreiswirth B. Minimising moxifloxacin
56. Golub JE, Saraceni V, Cavalcante SC, et al. The impact of resistance with tuberculosis. Lancet Infect Dis. 2008;8(5):
antiretroviral therapy and isoniazid preventive therapy on 273-275.
tuberculosis incidence in HIV-infected patients in Rio de 73. Starke JR, Smith M. Tuberculosis. Textbook of Pediatric
Janeiro, Brazil. AIDS. 2007;21(11):1441-1448. Infectious Diseases. 5th ed. Philadelphia, PA: WB Saunders;
57. Mandalakas AM, Hesseling AC, Chegou NN, et al. High level 2004:1337-1379.
of discordant IGRA results in HIV-infected adults and chil- 74. Dogra S, Narang P, Mendiratta DK, et al. Comparison
dren. Int J Tuberc Lung Dis. 2008;12(4):417-423. of a whole blood interferon-gamma assay with tuberculin
58. Pai M, Dheda K, Cunningham J, Scano F, O’Brien R. T-cell skin testing for the detection of tuberculosis infection in
assays for the diagnosis of latent tuberculosis infection: moving hospitalized children in rural India. J Infect. 2007;54(3):
the research agenda forward. Lancet Infect Dis. 2007;7(6): 267-276.
428-438. 75. Mandalakas AM, Detjen AK, Hesseling AC, Benedetti A,
59. Getahun H, Kittikraisak W, Heilig CM, et al. Development Menzies D. Interferon-gamma release assays and child-
of a standardized screening rule for tuberculosis in people hood tuberculosis: systematic review and meta-analysis. Int J
living with HIV in resource-constrained settings: individ- Tuberc Lung Dis. 2011;15(8):1018-1032.
ual participant data meta-analysis of observational studies. 76. Jensen PA, Lambert LA, Iademarco MF, Ridzon R; Centers
PLoS Med. 2011;8(1):e1000391. for Disease Control and Prevention. Guidelines for prevent-
60. Akolo C, Adetifa I, Shepperd S, Volmink J. Treatment ing the transmission of Mycobacterium tuberculosis in health-
of latent tuberculosis infection in HIV infected persons. care settings, 2005. MMWR Recomm Rep. 2005;54(RR-17:
Cochrane Database Syst Rev. 2010;(1):CD000171. 1-141.
61. Fitzgerald DW, Desvarieux M, Severe P, Joseph P, 77. Madhi SA, Nachman S, Violari A, et al; P1041 Study Team.
Johnson WD Jr, Pape JW. Effect of post-treatment isoniazid Primary isoniazid prophylaxis against tuberculosis in HIV-
on prevention of recurrent tuberculosis in HIV-1-infected indi- exposed children. N Engl J Med. 2011;365(1):21-31.
viduals: a randomised trial. Lancet. 2000;356(9240):1470-1474. 78. Revised National Tuberculosis Control Program. Technical
62. Churchyard GJ, Fielding K, Charalambous S, et al. Effi- and Operational Guidelines for Tuberculosis Control. New
cacy of secondary isoniazid preventive therapy among HIV- Delhi, India: Central Tuberculosis Division; Directorate
infected Southern Africans: time to change policy? AIDS. General of Health Services, Ministry of Health and Family
2003;17(14):2063-2070. Welfare, Government of India; 2005. http://health.bih.nic.in/
63. van Halsema CL, Fielding KL, Chihota VN, et al. Tubercu- Docs/Guidelines-TB-Control.pdf. Accessed May 26, 2012.
losis outcomes and drug susceptibility in individuals exposed 79. Banu Rekha VV, Jagarajamma K, Wares F, Chandrasekaran V,
to isoniazid preventive therapy in a high HIV prevalence set- Swaminathan S. Contact screening and chemoprophylaxis
ting. AIDS. 2010;24(7):1051-1055. in India’s Revised Tuberculosis Control Programme: a situa-
64. World Health Organization, HIV/AIDS Department. WHO tional analysis. Int J Tuberc Lung Dis. 2009;13(12):1507-1512.
Three I’s Meeting: Intensified Case Finding (ICF), Isoniazid 80. Van Wyk SS, Hamade H, Hesseling AC, Beyers N, Enarson DA,
Preventive Therapy (IPT) and TB Infection Control (IC) Mandalakas AM. Recording isoniazid preventive therapy
for People Living with HIV. Geneva, Switzerland: World delivery to children: operational challenges. Int J Tuberc Lung
Health Organization; 2008. http://www.who.int/hiv/pub/meeting Dis. 2010;14(5):650-653.
reports/WHO_3Is_meeting_report.pdf. Accessed May 26, 81. Baussano I, Nunn P, Williams B, Pivetta E, Bugiani M,
2012. Scano F. Tuberculosis among health care workers. Emerg
65. World Health Organziation. Multidrug and Extensively Infect Dis. 2011;17(3):488-494.
Drug Resistant TB (M/SDR-TB): 2010 Global Report on 82. Jafri SM, Singal AG, Kaul D, Fontana RJ. Detection and
Surveillance and Response. Publication WHO/HTM/TB/ management of latent tuberculosis in liver transplant patients.
2010.3. Geneva, Switzerland: World Health Organization; 2010. Liver Transpl. 2011;17(3):306-314.
http://whqlibdoc.who.int/publications/2010/9789241599191_ 83. Naqvi R, Naqvi A, Akhtar S, et al. Use of isoniazid chemo-
eng.pdf. Accessed May 26, 2012. prophylaxis in renal transplant recipients. Nephrol Dial
66. Sharma SK, Kumar S, Saha PK, et al. Prevalence of multi- Transplant. 2010;25(2):634-637.
drug-resistant tuberculosis among category II pulmonary 84. Sester M, Sester U, Clauer P, et al. Tuberculin skin testing
tuberculosis patients. Indian J Med Res. 2011;133:312-315. underestimates a high prevalence of latent tuberculosis
67. Teixeira L, Perkins MD, Johnson JL, et al. Infection and infection in hemodialysis patients. Kidney Int. 2004;65(5):
disease among household contacts of patients with multidrug- 1826-1834.
resistant tuberculosis. Int J Tuberc Lung Dis. 2001;5(4): 85. Soysal A, Toprak D, Koc M, Arikan H, Akoglu E, Bakir M.
321-328. Diagnosing latent tuberculosis infection in haemodialysis
68. Becerra MC, Appleton SC, Franke MF, et al. Tuberculosis patients: T-cell based assay (T-SPOT.TB) or tuberculin skin
burden in households of patients with multidrug-resistant test? Nephrol Dial Transplant. 2011;27(4):1645-1650.
and extensively drug-resistant tuberculosis: a retrospective 86. Kowada A. Cost effectiveness of interferon-gamma release
cohort study. Lancet. 2011;377(9760):147-152. assay for tuberculosis screening of rheumatoid arthritis
69. Schaaf HS, Gie RP, Kennedy M, Beyers N, Hesseling PB, patients prior to initiation of tumor necrosis factor-a antago-
Donald PR. Evaluation of young children in contact with nist therapy. Mol Diagn Ther. 2010;14(6):367-373.

772 Special Features

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016


87. Solovic I, Sester M, Gomez-Reino JJ, et al. The risk of tuber- Health-Care Settings. Geneva, Switzerland: World Health
culosis related to tumour necrosis factor antagonist therapies: Organization; 2009. http://whqlibdoc.who.int/publications/
a TBNET consensus statement. Eur Respir J. 2010;36(5): 2009/9789241547857_eng.pdf. Accessed May 28, 2012.
1185-1206. 90. Agarwal A, Anand S, Chadha S, et al National Airborne Infec-
88. Mariette X, Salmon D. French guidelines for diagnosis and tion Control Committee. Guidelines on Airborne Infection
treating latent and active tuberculosis in patients with RA Control in Healthcare and Other Settings. New Delhi, India:
treated with TNF blockers. Ann Rheum Dis. 2003;62(8):791. TBC India; 2010. http://tbcindia.nic.in/pdfs/Guidelines_on_
89. Atkinson J, Chartier Y, Pessoa-Silva CL, Jensen P, Li Y, Airborne_Infection_Control_April2010Provisional.pdf. Accessed
Seto W-H. Natural Ventilation for Infection Control in May 28, 2012.

journal.publications.chestnet.org CHEST / 142 / 3 / SEPTEMBER 2012 773

Downloaded From: http://journal.publications.chestnet.org/ on 09/07/2016

You might also like