ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Treatment of tuberculosis infection (latent tuberculosis) in nonpregnant adults with HIV infection

Treatment of tuberculosis infection (latent tuberculosis) in nonpregnant adults with HIV infection
Literature review current through: Jan 2024.
This topic last updated: Jun 02, 2023.

INTRODUCTION — Treatment of individuals with active tuberculosis (TB) is the first priority for TB control; an important second priority is identification and treatment of individuals with latent TB infection (LTBI) [1-6]. Issues related to treatment of LTBI in nonpregnant patients with human immunodeficiency virus (HIV) infection will be reviewed in more detail here.

The diagnosis of LTBI is discussed separately. (See "Tuberculosis infection (latent tuberculosis) in adults: Approach to diagnosis (screening)" and "Use of the tuberculin skin test for diagnosis of tuberculosis infection (tuberculosis screening) in adults" and "Use of interferon-gamma release assays for diagnosis of tuberculosis infection (tuberculosis screening) in adults".)

Issues related to treatment of LTBI in individuals without HIV are discussed separately. (See "Treatment of tuberculosis infection (latent tuberculosis) in nonpregnant adults without HIV infection" and "Tuberculosis disease (active tuberculosis) in pregnancy" and "Tuberculosis infection (latent tuberculosis) in children".)

TERMINOLOGY — TB terminology is inconsistent in the literature [7]. Relevant terms are defined in the table (table 1).

RATIONALE FOR TBI TREATMENT — Tuberculosis infection (TBI) is caused by inhalation of viable tuberculous bacilli. These organisms usually persist in an inactive state, known as latent TBI; in some cases, there is rapid progression to active TB disease.

Individuals with TBI are asymptomatic and not infectious. Latent TB bacilli remain viable and may reactivate years later, causing active symptomatic and transmissible TB disease [8]. The natural history of TB is discussed further separately. (See "Tuberculosis: Natural history, microbiology, and pathogenesis".)

Following primary TB infection, patients with HIV infection are at greater risk of progressing to active TB disease than individuals without HIV [8,9]; the interval between exposure and disease can be less than two months [9,10]. In addition, patients with untreated HIV and TBI are 30 to 100 times more likely to reactivate with TB disease than individuals without HIV [11-14]. Antiretroviral therapy reduces risk of developing active TB disease among individuals with HIV and TBI [15-18]. (See 'Initiation of antiretroviral therapy' below.)

Treatment of TBI in patients with HIV is associated with two important benefits:

Treatment of TBI reduces the risk of progression to active TB disease [4,19]. Among patients in Africa with HIV and TBI, the risk of developing active TB disease is 5 to 10 percent per year [20,21]. Overall, treatment of TBI in individuals with HIV reduces active TB disease by 62 percent and mortality by 26 percent; most studies have demonstrated the benefit is greatest among those with a positive test for TBI (via tuberculin skin test or interferon-gamma release assay) [20].

Treatment of TBI reduces TB transmission [15,20,22]. Mathematical models have predicted that provision of TBI therapy to at least half of individuals with HIV and TBI would be cost effective and would result in a substantial reduction in TB disease [22,23].

For these reasons, treatment of TBI in people with HIV has been favored by expert panels for many years [24]. However, despite consistent and clear recommendations from authoritative agencies (including the World Health Organization) [5], it is estimated that in 2018 and 2019 less than 50 percent of patients with HIV newly accessing HIV care received TBI treatment worldwide [25,26].

WHOM TO TREAT — TBI treatment is warranted for individuals with HIV infection in the following circumstances (in some cases, it may not be possible to definitively establish the presence of TBI) [4]:

Individuals with recent contact with a person with active TB disease

Individuals with clinical suspicion for prior TB (eg, fibrotic disease on chest radiograph consistent with healed TB) and no documented history of adequate TB treatment

Individuals with positive tuberculin skin test (TST) or interferon-gamma release assay (IGRA) in the absence of active TB

If TBI testing is not available − All individuals with HIV infection (particularly if the CD4 cell count is <200 cells/microL, which confers greater risk for development of TB disease), once active TB has been excluded

Testing for TBI via TST or IGRA is warranted prior to initiation of therapy; multiple studies suggest that the benefits of treatment are greatest among patients with HIV infection and positive TST or IGRA [4,15,17,20,27,28]. In one systematic review of longitudinal studies, the incidence of active TB was 11 times higher among individuals with HIV infection and a positive IGRA or TST than those with a negative test [29]. Similarly, in two systematic reviews of 12 controlled trials evaluating treatment regimens administered for 2 to 12 months [20], and three trials of extended duration isoniazid (INH) [30], TBI treatment was associated with reduction in TB incidence [20,30] and death [30] only among patients with HIV who had positive TST at enrollment.

For patients with HIV infection in whom there is strong likelihood of TB exposure, but TBI testing is not available (despite all efforts of the provider), TBI treatment is warranted. This approach is supported by a randomized trial in South Africa including 1329 individuals with HIV treated with INH or placebo; the incidence of TB over a three-year period was lower among those who received treatment than those who did not (2.3 versus 3.6 per 100 person-years; hazard ratio 0.63, 95% CI 0.41-0.94) [31].

Issues related to diagnosis of TBI are discussed further separately. (See "Tuberculosis infection (latent tuberculosis) in adults: Approach to diagnosis (screening)".)

PRIOR TO TREATMENT

Exclude active TB disease — Prior to initiation of treatment for TBI, all patients must be evaluated for active TB disease, to avoid inadvertent undertreatment of active disease leading to development of drug resistance [32]. The evaluation includes clinical history, physical examination, and chest radiograph; it also may require microbiologic testing of respiratory samples. (See "Tuberculosis infection (latent tuberculosis) in adults: Approach to diagnosis (screening)", section on 'Excluding TB disease'.)

The increased risk of active TB begins as soon as HIV infection is acquired and is especially high among patients with a CD4 cell count <200 cells/mm3 [33]. Patients with advanced immunosuppression may present with atypical clinical manifestations including extrapulmonary TB disease and/or mediastinal lymphadenopathy or with subclinical infection and a normal chest radiograph.

Excluding active TB in individuals with HIV may be more difficult than in individuals without HIV. Among individuals with HIV, symptoms may be minimal or nonspecific, and the yield of diagnostic procedures may be limited because of many factors; these include presence of extrapulmonary disease (with absence of lung involvement in some cases) and normal chest radiography (pulmonary disease present but relative lack of inflammation related to immune suppression).

The World Health Organization (WHO) suggests symptoms screening is adequate to exclude active TB [34]; however, in a systematic review of 21 studies including more than 15,000 people with HIV, the sensitivity of symptoms screen alone was only 51 percent in those on antiretroviral therapy (ART). Based on data from two studies, the addition of chest radiography improved the sensitivity of active TB detection to 89 percent [35]. Hence, we strongly advise chest radiography to exclude active TB before initiation of TBI treatment.

The investigation for active TB should include multiple respiratory samples for acid-fast bacilli (AFB) smear and TB culture; sputum induction may be warranted. If sputum is nondiagnostic, bronchoscopy may be useful to obtain additional respiratory samples for diagnosis of TB as well as to facilitate evaluation for other causes of pulmonary infiltrates and respiratory symptoms.

For individuals with HIV in whom it is difficult to exclude active TB (for example, individuals with relevant clinical manifestations [cough >2 weeks' duration, fevers, night sweats, and/or weight loss] but negative sputum AFB smear), initiation of treatment for active disease (pending microbiology data) is reasonable, particularly in patients with advanced immunosuppression or in resource-limited settings. If there is no improvement in symptoms or radiographic findings with empiric treatment for active TB and microbiologic studies are negative, additional evaluation for an alternative cause should be pursued, and the patient may be switched to a regimen for treatment of TBI. In such cases, if the patient has taken two months of standard therapy for active TB (ie, isoniazid [INH], rifampin, and pyrazinamide), the patient may be considered to have completed adequate treatment for TBI [1].

The diagnosis of active TB disease is discussed in detail separately. (See "Diagnosis of pulmonary tuberculosis in adults".)

Assess comorbidities — Comorbidities that should be considered in planning treatment of TBI include underlying liver disease, neuropathy, and concomitant medications.

Regimens for treatment of TBI are associated with hepatotoxicity; the risk of hepatotoxicity is greatest with INH and less so for the rifamycin-based regimens. Baseline liver enzyme testing is appropriate for the following individuals [4,24,36]:

Patients receiving ART. In earlier guidelines, HIV infection was considered a risk factor for drug-induced liver injury [24]. Subsequently, an extensive review concluded that the likelihood of developing INH-related hepatotoxicity among individuals with HIV is comparable with that of individuals without HIV [37].

Patients with underlying liver disease (eg, hepatitis B or C, alcoholic hepatitis, or cirrhosis) or risk of underlying liver disease

Pregnant and postpartum women within three months of delivery

Patients who consume alcohol regularly

Patients on other medication(s) with potential hepatotoxicity

Baseline liver enzyme testing may also be considered on an individual basis for other patients but need not be performed routinely. Active hepatitis and end-stage liver disease are relative contraindications to the use of hepatotoxic agents for TBI treatment. For patients with baseline serum transaminase levels greater than three times the upper limit of normal, TBI treatment decisions should be deferred pending evaluation of the underlying cause.

The risks and benefits of initiating treatment of TBI in patients with abnormal baseline liver function (alanine aminotransferase [ALT] >3 times the upper limit of normal) should be considered carefully. Clinical considerations include baseline ALT level, alcohol use, age, presence of viral hepatitis, and the risk of TB progression [37]. If therapy is required, serum transaminases, bilirubin, and international normalized ratio should be monitored every two to four weeks [37].

The potential for drug-drug interactions with rifamycins is high for individuals taking the following drugs: warfarin, oral or other hormonal contraceptives, corticosteroids, some antihypertensives, some antiarrhythmics, some antidepressants, some anticonvulsants, methadone, and some antiretroviral drugs (including tenofovir alafenamide, some non-nucleoside reverse transcriptase inhibitors, all protease inhibitors, and all integrase inhibitors). Details about specific interactions may be obtained by using the drug interactions program included within UpToDate. (See "Rifamycins (rifampin, rifabutin, rifapentine)".)

SELECTING A REGIMEN — The approach to treatment of TBI in individuals with HIV depends on a number of factors, including possible drug interactions in individuals taking antiretroviral therapy (ART), known or suspected susceptibility of the presumed infecting organism, likelihood of patient adherence with longer regimens, and local TB transmission rates.

Settings with TB transmission <500 per 100,000

General approach — For treatment of TBI in patients with HIV infection on ART, we favor either 3HP (isoniazid [INH] and rifapentine [RPT] administered weekly for three months via directly observed therapy) or 4R (rifampin [RIF] self-administered daily for four months) over INH monotherapy (table 2). We favor this approach in view of the similar efficacy, improved treatment completion rates, and significantly lower rates of hepatotoxicity of these two rifamycin-based regimens compared with INH, in HIV uninfected [38,39] and HIV infected [40,41] persons.

This approach differs from that of the World Health Organization (WHO), which favors daily administration of INH for six months [5]. It also differs somewhat from the United States Department of Health and Human Services (DHHS) Opportunistic Infection Guidelines in which the preferred therapies are 3HP and 3HR [42].

Use of a rifamycin-based regimen requires careful attention to drug interactions [1]. A discussion of the available data on the efficacy for these regimens is presented below. (See 'Regimens: efficacy and toxicity' below.)

Drug interactions related to rifamycins are a very important consideration for management of patients with HIV infection on ART. Treatment decisions must be individualized, given that (1) integrase strand transfer inhibitor (INSTIs)-based regimens are preferred for treatment of HIV infection, (2) rifamycin-based regimens (3HP and 4R) are preferred for treatment of TBI, and (3) coadministration of INSTIs and rifamycins is associated with significant risk of adverse drug effects.

Treatment decisions should be guided by the patient's current ART regimen; in some circumstances, we consider switching to an alternative ART regimen with minimal rifamycin-based drug interactions for the duration of TBI treatment.

Use of 3HP – The regimen 3HP (INH and RPT administered weekly for three months via directly observed therapy [DOT]) is preferred for treatment of TBI in patients with HIV infection, given greatest efficacy and safety data (see 'Isoniazid and rifapentine (3HP)' below). This regimen may be used in the following circumstances:

Patients on a dolutegravir-based regimen (administered once daily) who are virologically suppressed

Patients taking (or willing to switch to) a raltegravir- or efavirenz- (EFV) based regimen

Patients on tenofovir alafenamide (TAF) would need to switch to tenofovir disoproxil fumarate (TDF) for the duration of TBI treatment.

Use of 4R – The regimen 4R (RIF self-administered daily for four months) may be used if DOT (for administration of 3HP) is not feasible. This regimen may be used for patients taking (or willing to change to) an EFV-based regimen; patients on a dolutegravir-based or raltegravir-based regimen require a doubling of the dose. This regimen may not be used for patients on bictegravir, TAF, and a number of other antiretroviral agents.

4R regimen is safe and efficacious in patients without HIV, but data among patients with HIV are limited (see 'Rifampin (4R)' below).

Use of INH – For patients who are unable or unwilling to switch to a rifamycin-compatible regimen, we use INH monotherapy (daily for nine months) for treatment of TBI. The WHO favors daily INH for six months because of better completion rates, but this carries the risk of lower efficacy [5]. The United States DHHS Opportunistic Infection Guidelines also list INH as the preferred therapy [4]. (See 'Isoniazid monotherapy' below.)

Regimens not recommended

3HR 3HR regimen has been studied in three randomized trials among adults with HIV infection [21,43,44]. In these studies, completion rates with 3HR were not significantly better than with 6H [43], and adverse events with 3HR were comparable to 6H [21,43] although less than with 12H [45]. The efficacy of 3HR and isoniazid monotherapy regimens was similar in the three studies [21,43,45].

Hence, 3HR confers no benefit over INH monotherapy in terms of completion, efficacy, or toxicity, but it does confer risk of rifampin-induced drug interactions. Our approach is similar to Canadian recommendations [46], but differs from DHHS recommendations which prefer 3HP or 3HR [42]. The Centers for Disease Control and Prevention (CDC) have conditionally recommended 3HR [1], while WHO recommends use of 3HR in settings where rifamycins are only available as fixed dose combinations of INH and RIF [5].

While EFV may be used with 3HR, dolutegravir requires doubling the dose and other antiretroviral agents (raltegravir, bictegravir, TAF, and others) may not be used.

RIF plus pyrazinamide – RIF plus pyrazinamide should not be used for treatment of TBI due to hepatotoxicity [47].

Nitrosamine impurities – Given detection of nitrosamine impurities in samples of RIF and RPT announced by the US Food and Drug Administration (FDA) in August 2020 [48], INH monotherapy may be considered as an alternative regimen for adults with newly diagnosed TBI. However, for adults at highest risk of new TB infection or TB reactivation (such as close contacts, recent converters, immunocompromised individuals, and those who will be receiving tumor necrosis factor-alpha inhibitors), and particularly for those at increased risk of liver toxicity (age >50 years, hepatitis C coinfection, or other liver disease), use of rifamycin-containing regimens may be considered after individual patient discussion of potential risks and benefits. To date, there is no information on the clinical significance of these nitrosamine impurities, nor is there information on possible impurities in rifabutin.

Additional considerations − Important considerations with respect to regimen selection and administration include:

RIF and RPT have significant interactions with many medications including the following: warfarin, oral and other hormonal contraceptives, corticosteroids, some antihypertensives, some antiarrhythmics, some antidepressants, some anticonvulsants, methadone, buprenorphine, and some antiretroviral drugs (including TAF, all non-nucleoside reverse transcriptase inhibitors [NNRTIs], all protease inhibitors, and all INSTIs). Specific interactions can be determined by use of the drug interactions program included within UpToDate. (See 'Drug interactions: ART and rifamycins' below and "Rifamycins (rifampin, rifabutin, rifapentine)".)

Regimens for treatment of TBI are associated with hepatotoxicity. The risk of hepatotoxicity is greatest with INH and significantly less with 4R or 3HP. The risk appears to be somewhat less with 3HR than with 6 to 12 months of INH [21,43,45], although in these studies overall rates of drug discontinuation due to adverse events was similar with 3HR and the longer isoniazid monotherapy regimens. Additional side effects associated with rifamycins include rash and thrombocytopenia. (See 'Assess comorbidities' above and 'Monitoring and adherence' below.)

Peripheral neuropathy occurs in up to 2 percent of patients taking INH due to interference with metabolism of pyridoxine and can be prevented with pyridoxine supplementation [49]. For all patients with HIV infection on an TBI regimen containing INH, we coadminister pyridoxine (25 to 50 mg daily). We also administer pyridoxine to infants of breastfeeding mothers receiving INH.

Important side effects of 3HP include hypersensitivity or flu-like symptoms (eg, lightheadedness, dizziness, headache, nausea or vomiting, syncope, rash, or angioedema); these occurred in 3.8 percent of patients treated with 3HP in one trial [38]. For this reason, 3HP usually is administered via DOT, to facilitate side effect review and treatment withholding if needed. However, self-administration may be acceptable for patients who can reliably take their medications on schedule and inform their providers promptly of side effects (while withholding the next dose pending provider review) [38,50,51]. (See "Treatment of tuberculosis infection (latent tuberculosis) in nonpregnant adults without HIV infection", section on 'Isoniazid and rifapentine (3HP)'.)

Self-administered INH is frequently associated with poor adherence; for patients with poor adherence and contraindications to rifamycins, administration of INH via DOT may be the optimal approach.

Regimens: efficacy and toxicity

Rifamycin-based regimens — Use of a rifamycin-based regimen over INH monotherapy for treatment of TBI in patients with HIV infection on antiretroviral therapy is supported by a network meta-analysis including 20 studies (including 16 randomized trials) with median sample size 616; administration of INH monotherapy (6 to 12 months) was no more efficacious for TB prevention than rifamycin-containing regimens (incidence rate ratio [IRR] 1.0, 95% CI 0.8-1.4); however, INH monotherapy was associated with higher all-cause mortality (IRR 1.6, 95% CI 1.2-2.0) and higher risk of severe hepatotoxicity (risk difference 8.9 per 100 persons treated, 95% CI 2.8-14.9) [52].

The FDA announced detection of nitrosamine impurities in samples of RIF and RPT in August 2020. This is discussed further above. (See 'General approach' above.)

If RIF or RPT is used to treat TBI, clinicians should assess the potential for drug-drug interactions among the rifamycins and different ART regimens. (See 'Drug interactions: ART and rifamycins' below.)

Isoniazid and rifapentine (3HP) — Thus far, the greatest cumulative clinical and trial experience with INH and RPT, although limited, has been a weekly regimen for three months (3HP) in which doses are directly observed. A regimen of INH and RPT administered daily for one month (1HP) has been studied in patients with HIV, as described below. However, given limited data for this approach, combination therapy with INH and RPT should be administered for three months as 3HP pending further study (table 2). (See "Treatment of tuberculosis infection (latent tuberculosis) in nonpregnant adults without HIV infection", section on 'Isoniazid and rifapentine (3HP)'.)

Use of ART with 3HP − ART regimens that may be used for patients treated with 3HP are discussed above. (See 'General approach' above.)

Dosing − Weight-based RPT dosing may result in underdosing among the lower weight categories; in addition, RPT levels are approximately 30 percent lower in people with HIV [53]. Pending further data and consideration by authoritative agencies, we continue to dose RPT as summarized in the table (table 2).

Side effects − Important side effects of 3HP include hypersensitivity or flu-like symptoms (eg, lightheadedness, dizziness, headache, nausea or vomiting, syncope, rash, or angioedema); these occurred in 3.8 percent of patients treated with 3HP in one trial [38]. For this reason, 3HP usually is administered via DOT, to facilitate side effect review and treatment withholding if needed. In one trial including more than 1000 patients with TBI comparing self-administration of 3HP with DOT, the rate of treatment completion was lower in the self-administered group [50]. However, self-administration may be acceptable for patients who can reliably take their medications on schedule and inform their providers promptly of side effects (while withholding the next dose pending provider review) [38,50,51].

Efficacy data for 3HP – Use of 3HP in patients for treatment of TBI in patients with HIV infection is supported by the following trial data:

In a trial including more than 4000 patients with HIV infection on ART (largely efavirenz-based regimens; median CD4 count 440 cells/mcL) in South Africa, Ethiopia, and Mozambique, patients were randomly assigned to treatment with 3HP given once, 3HP given annually for two years, or INH daily for six months (6H) [54]. Approximately 60 percent of participants had negative QuantiFERON TB Gold Plus test results. The incidence of TB among the two groups treated with 3HP was similar (1.21 versus 1.26 per 100 person-years; hazard ratio 0.96, 95% CI 0.61-1.50), suggesting no benefit of repeat 3HP treatment. The incidence of TB was comparable among those treated with 3HP or 6H (hazard ratio 1.60, 95% CI 0.58-4.42), despite more favorable treatment completion rates with 3HP (90.4 versus 50.5 percent; risk ratio 1.78, 95% CI 1.61-1.95).

In a trial including nearly 400 patients with HIV infection (median CD4 count approximately 500 cells/mcL) in the United States, Brazil, Spain, Peru, Canada, and Hong Kong, patients were randomly assigned to treatment with 3HP or INH daily for 9 months (9H) [40]. Enrolled participants were not taking ART or planning to initiate ART in the next 90 days. The TB case rate was lower among those treated with 3HP (0.39 versus 1.25 per person-year). The treatment completion rate was higher with 3HP than 9H (89 versus 64 percent); the rate of adverse drug reactions was comparable between the groups.

Efficacy data for 1HP − In a randomized trial including nearly 3000 patients with HIV infection (of whom 43 percent were receiving EFV-based ART, 7 percent were receiving nevirapine-based ART, and 50 percent were not on ART), a regimen of one month of daily self-administered INH and RPT (<35 kg: 300 mg, 35 to 45 kg: 450 mg, >45 kg: 600 mg) was compared with nine months of daily INH [55]. Among the study participants, 23 percent had a positive test for TBI; 60 percent had a negative test; the remainder were not tested but resided in a region with TB prevalence 60 to 100 cases per 100,000 population. The one-month regimen was noninferior to the nine-month regimen for preventing TB; during the follow-up period (median 3.3 years), the incidence rates for TB disease or death were 0.65 and 0.67 per 100 person-years, respectively. The rate of serious adverse events over the full period of follow-up was comparable (6 versus 7 percent). The treatment completion rate was very high in both arms, although higher among those who received the one-month regimen (97 versus 90 percent).

The high completion rates emphasize the high quality of trial execution, but it may be difficult to extrapolate results to routine clinical settings. Limitations of the trial include broad eligibility criteria and relatively low proportion of patients with CD4 <250 cells/microL (13 percent); despite this, most patients were tuberculin skin test (TST) negative. In addition, given that toxicity is a major limitation of the nine-month regimen, safety of the one-month regimen requires further evaluation in other settings and populations.

Rifampin (4R) — Thus far, safety data available from randomized trials for use of 4R in patients with HIV are limited.

In a secondary analysis of two randomized trials of the 270 patients with HIV infection treated with 4R or 9 months of INH, drug-related adverse events were less common among patients treated with 4R than those treated with 9H (risk difference -2.1 percent, 95% CI -5.9 to 1.6) [41]. Rates of active TB were also nonsignificantly lower with 4R than 9H (rate difference per 100 person-years -0.4: 95% CI -2.2 to 1.3).

ART regimens that may be used for patients treated with 4R are discussed above. (See 'General approach' above.)

Among patients without HIV, of the available TBI regimens, 4R is associated with the lowest rate of hepatotoxicity [1,56,57]. (See "Treatment of tuberculosis infection (latent tuberculosis) in nonpregnant adults without HIV infection".)

There are no data to support use of rifabutin monotherapy for treatment of TBI in patients with HIV on ART.

Isoniazid and rifampin (3HR) — We do not favor use of 3HR for treatment of TBI in patients with HIV infection; this regimen confers no benefit over INH monotherapy in terms of efficacy or toxicity, but it does confer risk of drug interactions associated with rifampin. (See 'General approach' above.)

The efficacy and hepatotoxicity of 3HR is similar to that of INH administered for six months. In a randomized trial among more than 2000 individuals with HIV evaluating 3HR and INH for six months, the relative risk of TB with INH alone (compared with placebo) was 0.33 (95% CI 0.14-0.77) and with INH and RIF was 0.40 (95% CI 0.18-0.86) [21].

Some trials supporting recommendations for use of 3HR actually used four months of HR [58]. In a meta-analysis of five trials among patients with and without HIV infection, the toxicity of 3HR or 4HR was the same as that of INH administered for 6 to 12 months [58].

Isoniazid monotherapy — Of the available TBI regimens, INH is associated with the highest rate of hepatotoxicity in adults [56,57,59,60]. This can result in hospitalization [59,61] and fatal hepatoxicity [59,60]. In addition, the risk of mortality due to INH hepatotoxicity in adults with HIV may be under-recognized in high TB incidence settings; in one cohort including more than 4900 patients with HIV in Eritrea treated with 6H, life-threatening liver toxicity was observed in 15 cases and 4 died [60].

Prior to the availability of clinical trial data supporting use of rifamycin-based regimens, INH monotherapy was favored for treatment of TBI in patients with HIV, given its efficacy and compatibility with ART [17,18,20]. This approach is based on inference from clinical trials in patients without HIV and subgroup analyses of data from trials using treatment interventions of different durations [4,24,62]. INH monotherapy remains an acceptable alternative agent for patients with contraindications to use of rifamycins (such as unmanageable drug-drug interactions or hypersensitivity). (See "Treatment of tuberculosis infection (latent tuberculosis) in nonpregnant adults without HIV infection", section on 'Isoniazid monotherapy'.)

We favor daily INH dosing (300 mg) for nine months; the DHHS recommends six to nine months [42], and the WHO favors daily dosing for six months [5]. Based on limited data in patients without HIV infection, the efficacy of six months is lower than the efficacy of nine months (risk reduction 69 versus 93 percent) [63], but completion rates are better [24]. In the setting of difficulty with adherence, providers may prefer to concentrate efforts in ensuring six months of therapy.

Any ART regimen may be used for patients on INH monotherapy [3]. For patients who cannot tolerate INH and who are on an ART regimen containing drugs that interact with rifamycins and cannot be managed with dose adjustments and/or substitutions, the approach requires careful individualized review of medications and potential drug interactions. In such cases, consultation with an expert is warranted prior to selecting a regimen for treatment of TBI.

Drug interactions: ART and rifamycins — The potential for drug-drug interactions with rifamycins in individuals taking ART is high. These interactions often are bi-directional and may be complex. Antiretroviral drugs should be reviewed carefully for compatibility with rifamycins [3,64].

Rifamycins accelerate metabolism of TAF, all NNRTIs, all protease inhibitors, all integrase strand transfer inhibitors (INSTIs; bictegravir cabotegravir, dolutegravir, elvitegravir, and raltegravir), and the CCR5 antagonist maraviroc. RPT appears to cause less severe drug interactions than RIF [2]. (See "Rifamycins (rifampin, rifabutin, rifapentine)" and "Treatment of drug-susceptible pulmonary tuberculosis in nonpregnant adults with HIV infection: Initiation of therapy", section on 'Drug interactions' and "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach".)

TAF is a potent nucleotide reverse transcription inhibitor that is used commonly in HIV treatment regimens. Strong P-glycoprotein inducers like RIF, rifabutin, and RPT likely inhibit the absorption of TAF, and the United States FDA Prescribing Information recommends against administration of these agents with TAF because of risk of treatment failure [65]. However, while administration of TAF with RIF to healthy volunteers reduced both plasma and intracellular TAF exposure by 36 percent relative to administration of TAF alone, intracellular concentrations of the active drug metabolite were more than four times higher than levels following administration of tenofovir disoproxil fumarate (TDF) alone; these findings suggest that coadministration of TAF and rifampin may be acceptable [66]. More data are needed to understand these interactions, before rifamycins can be used safely with TAF.

RIF only leads to modest reduction in EFV concentrations, and standard doses of EFV can still achieve good clinical outcomes [3]. Use of nevirapine with RIF is not recommended due to low levels with RIF [67]. RIF significantly decreases concentrations of the other NNRTIs (doravirine, etravirine, nevirapine, and rilpivirine) and they should not be used together [68-71]. RIF cannot be used with any HIV protease inhibitor, due to significant decreases in protease inhibitor concentrations [3,72].

RIF causes decreases in concentrations of INSTIs; this can be managed by doubling the dose of dolutegravir or raltegravir; however, other INSTIs (bictegravir, cabotegravir, elvitegravir-cobicistat) may not be coadministered with RIF [73,74].

RPT induces cytochrome P450 isoenzymes and can cause significant interactions [51]. 3HP may be used for patients on dolutegravir-, EFV-, or raltegravir-based regimens (in combination with either abacavir-lamivudine or TDF fumarate-emtricitabine) [4,75,76]. RPT significantly reduces other integrase inhibitor concentrations (bictegravir, cabotegravir, elvitegravir-cobicistat) [4], and coadministration is contraindicated.

Guidelines issued in 2020 to 2021 indicate that 3HP be administered to patients with HIV infection who are on dolutegravir [4,77], based on a phase 1/2 study among 61 South African patients with HIV infection who received dolutegravir and 3HP; grade 3 adverse events were observed in only 5 percent of cases [76]. However, we favor close observation in such cases, given one pharmacokinetic study including four volunteers without HIV who took 3HP with dolutegravir, in which two individuals developed a severe flu-like syndrome with elevated transaminases and endogenous cytokine release. However, thus far, this adverse effect has not been observed among patients with HIV infection [78].

Settings with high TB transmission rates (≥500 per 100,000) — The WHO recommends that adults and adolescents with HIV and a positive or unknown TBI test in settings with "high" TB transmission be treated with INH for at least 36 months [5]; however, the regional TB transmission level is not always known. In general, we support the use of extended INH (≥36 months) for settings with TB incidence >500 per 100,000 population (table 3); this is the incidence rate in the settings where randomized trials demonstrating benefit of extended INH were conducted (eg, Botswana and South Africa) [17,18]. Since the trial, Botswana's TB incidence has fallen from 900 to <500 per 100,000 [17].

Given the patient burden, cost, and potential toxicity of an extended of duration of INH, the decision to administer extended INH should be guided by careful assessment of the local epidemiology and the likelihood of ongoing or recurrent exposure, as well as the potential for adverse events and capacity for monitoring [18,79].

The above approach is supported by a systematic review including three randomized trials evaluating the efficacy of extended INH in Botswana, South Africa, and India [30]; the risk of active TB was lower among patients treated with extended-duration INH than among patients receiving a 6-month duration of INH (relative risk [RR] 0.62, 95% CI 0.42-0.89) and lower among those with a positive TST at enrollment (RR 0.51, 95% CI 0.30-0.86) [30]. Two studies found no increase in adverse events among those treated with extended INH, whereas a third study (using different definitions) found strong evidence of increase. There was no evidence of increased drug resistance among those treated with INH. In contrast, in a randomized trial in India (TB incidence 209 per 100,000 population) including 712 patients with HIV infection treated with extended INH or a 6-month course of INH and ethambutol, no significant benefit of extended INH was observed [79]. In a network meta-analysis of 16 trials [52], the incidence of active TB was significantly lower with 24 to 72 months of INH monotherapy than with 6 to 12 months INH, or shorter rifamycin-based regimens, but grade 3-4 hepatotoxicity was significantly worse, particularly relative to the rifamycin regimens (risk difference per 100 persons 20.7, 95% CI 12.5-28.9).

Settings with extremely high TB transmission rates — In a few settings, such as the South African gold mines, where close quarters, poor ventilation, and frequent HIV coinfection produced some of the highest TB rates in the world, the optimal approach to TBI treatment is uncertain. In one cluster-randomized study in this setting, mass screening and treatment for TBI had no significant effect on TB control, despite successful use of INH in preventing TB during TBI treatment [80]. In such settings, there are no data on the impact of any short rifamycin-containing regimen; further study is needed.

Empiric initiation of treatment for active TB disease (four-drug therapy) does not appear superior to preventive therapy for reduction of mortality. (See "Treatment of drug-susceptible pulmonary tuberculosis in nonpregnant adults with HIV infection: Initiation of therapy", section on 'Role of empiric TB therapy'.)

MONITORING AND ADHERENCE — The optimal approach to liver enzyme testing for patients with HIV receiving treatment for TBI (in the absence of other risk factors) is uncertain. The risk of drug-induced hepatotoxicity associated with isoniazid (INH) increases with age [81] and alcohol use [81]. There is no consistent evidence of increased toxicity of TBI therapy in individuals with HIV [82,83]. (See "Treatment of pulmonary tuberculosis in adults with HIV infection: Follow-up after initiation of therapy", section on 'Drug-related side effects'.)

Issues related to baseline laboratory testing are discussed above. (See 'Assess comorbidities' above.)

For patients with abnormal baseline aminotransferases and for patients with additional risk factors for hepatotoxicity (eg, pre-existing liver disease), we favor subsequent laboratory monitoring of serum transaminases, bilirubin, and international normalized ratio every two to four weeks [4,24,37].

For patients with normal baseline aminotransferases who are initiating treatment for TBI, we favor a single aminotransferase assay after approximately one month of therapy. Otherwise, laboratory monitoring is warranted only in the setting of clinical symptoms or other concern for hepatotoxicity.

We are in agreement with some experts who favor withholding TBI treatment in patients with symptomatic hepatitis and an alanine aminotransferase level (ALT) greater than three times the upper limit of normal, as well as among those with an ALT greater than five times the upper limit of normal (whether symptomatic hepatitis is present or not) [24,37].

Patients should be counseled on the symptoms and signs of drug-induced hepatitis (eg, right upper quadrant pain, jaundice, nausea, vomiting, loss of appetite, dark urine) and be instructed to stop therapy and report these symptoms promptly should they be experienced. Patients should be monitored monthly for signs and symptoms of adverse drug events and development of active TB [4]; regular patient-doctor interactions also provide an opportunity to reinforce the importance of drug adherence.

Issues related to clinical monitoring for patients on INH are discussed further separately. (See "Isoniazid: An overview".)

INITIATION OF ANTIRETROVIRAL THERAPY — Treatment for TBI may be started at any time, regardless of antiretroviral therapy (ART) status. We prefer to stagger initiation of ART and TBI treatment, to allow for assessment of side effects should any occur. For patients with a new diagnosis of HIV and TBI, we begin ART promptly and initiate TBI treatment a month later. (See 'Selecting a regimen' above.)

Early initiation of ART has been associated with lower incidence of active TB among patients with HIV in TB-endemic areas [15,17,27,84-86]:

In a randomized trial including more than 2000 patients with HIV in the Ivory Coast with CD4 <800 cells/mm3, both ART and six months of isoniazid preventive therapy were independently associated with lower rates of active TB [27]. There was a significant benefit of early ART even among participants with CD4 of 500 to 800 cells/mm3 at enrollment. (See "Use and impact of antiretroviral therapy for HIV infection in resource-limited settings", section on 'Tuberculosis'.)

In a randomized trial including more than 4600 patients with HIV around the world with CD4 counts >500 cells/mm3, active TB developed less frequently among those who received immediate ART than among those who started ART only when their CD4 count dropped to 350 cells/mm3 or when they developed a clinical condition that required ART (14 versus 20 percent) [87].

SPECIAL CIRCUMSTANCES

Drug-resistant TBI — Issues related to TBI among patients with HIV and possible drug-resistant TB are discussed separately. (See "Treatment of drug-resistant pulmonary tuberculosis in adults".)

Pregnancy — Issues related to TBI during pregnancy among patients with HIV are discussed separately. (See "Tuberculosis disease (active tuberculosis) in pregnancy".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Diagnosis and treatment of tuberculosis".)

SUMMARY AND RECOMMENDATIONS

Rationale – Treatment of tuberculosis infection (TBI) in patients with HIV is associated with two important benefits: reducing the risk for progression to active tuberculosis (TB) disease (which is greater among individuals with HIV those without HIV) and reducing TB transmission. (See 'Rationale for TBI treatment' above.)

Whom to treat – We recommend TBI treatment for nonpregnant adults with HIV in the following circumstances (see 'Whom to treat' above):

Individuals with recent contact with a person with active TB disease (Grade 1A)

Individuals with no signs or symptoms of active TB and clinical suspicion for prior TB (eg, fibrotic disease on chest radiograph consistent with healed TB) with no documented history of adequate TB treatment (Grade 1A)

Individuals with positive tuberculin skin test (TST) or interferon-gamma release assay (IGRA) in the absence of active TB (Grade 1A)

If TBI testing is not available − All individuals with HIV infection (particularly if CD4 cell count <200 cells/microL), once active TB has been excluded (Grade 1B)

Exclude active TB – Patients must be evaluated for active TB disease prior to initiation of TBI treatment, to avoid monotherapy and risk of TB drug resistance. We strongly advise chest radiography; respiratory samples for acid-fast bacilli smear and TB culture should also be obtained. (See 'Exclude active TB disease' above.)

Settings with TB transmission <500 per 100,000 – For treatment of TBI in patients with HIV infection on antiretroviral therapy (ART), we suggest a rifamycin-based regimen over isoniazid (INH) monotherapy (table 2) (Grade 2B), given the efficacy, favorable completion rates, and relatively low hepatotoxicity rates.

However, use of such regimens requires careful attention to drug interactions. Treatment decisions should be guided by the patient's current ART regimen; in some circumstances, we consider switching to an alternative regimen with minimal rifamycin-based drug interactions for the duration of TBI treatment. (See 'General approach' above.)

Use of 3HP – The regimen 3HP (INH and rifapentine [RPT] administered weekly for 3 months via directly observed therapy [DOT]) is preferred for treatment of TBI in patients with HIV infection. 3HP may be given to patients on a raltegravir- or efavirenz- (EFV) based regimen; it may also be given to patients on a dolutegravir-based regimen (administered once daily), if virologically suppressed. Patients on tenofovir alafenamide would need to switch for tenofovir disoproxil fumarate for the duration of TBI treatment.

Use of 4R – The regimen 4R (rifampin [RIF] self-administered daily for four months) may be used if administration of 3HP via DOT is not feasible. 4R may be given to patients on an EFV-based regimen; patients on a dolutegravir or raltegravir-based regimen require a doubling of the dose. 4R may not be used for patients on bictegravir, TAF, and a number of other antiretroviral agents.

Use of INH – For patients who are unable or unwilling to switch to a rifamycin-compatible regimen, we suggest INH monotherapy (daily for nine months) for treatment of TBI (Grade 2B). The World Health Organization (WHO) favors daily INH for six months because of better completion rates, but this carries risk of lower efficacy. The United States Department of Health and Human Services Opportunistic Infection Guidelines also list INH as the preferred therapy.

No role for 3HR − We do not favor use of 3HR for treatment of TBI in patients with HIV infection; this regimen confers no benefit over INH monotherapy in terms of efficacy or toxicity, but it does confer risk of rifampin-induced drug interactions. This approach differs from the WHO, which suggests use of 3HR in settings where rifamycins are only available as fixed dose combinations of INH and RIF.

Settings with TB transmission ≥500 per 100,000 – For patients in high transmission settings (table 3), we suggest treatment with INH for a minimum 36-month duration (Grade 2B) with particular attention to toxicity monitoring, given evidence of greater efficacy than shorter durations for reducing the risk of active TB in trials conducted in such settings. (See 'Settings with high TB transmission rates (≥500 per 100,000)' above.)

Monitoring and adherence (See 'Monitoring and adherence' above.)

Symptoms − Patients receiving TBI treatment should have monthly monitoring for symptoms of hepatitis. In addition, patients should be educated to stop their medication and seek prompt evaluation if they develop such symptoms.

Laboratory testing − We perform baseline liver enzyme testing for patients on ART. For patients with abnormal baseline liver enzymes and for patients with risk for hepatotoxicity, we perform subsequent laboratory monitoring every two to four weeks.

Treatment discontinuation − TBI treatment should be discontinued for symptomatic patients with serum transaminase levels greater than three times the upper limit of normal and for asymptomatic patients with serum transaminase levels greater than five times the upper limit of normal. TBI treatment should not be resumed even after liver function tests have improved.

Initiating ART – Treatment for TBI may be started at any time, regardless of ART status. For patients with a new diagnosis of HIV and TBI, we begin ART promptly and initiate TBI treatment a month later. (See 'Initiation of antiretroviral therapy' above.)

  1. Sterling TR, Njie G, Zenner D, et al. Guidelines for the Treatment of Latent Tuberculosis Infection: Recommendations from the National Tuberculosis Controllers Association and CDC, 2020. MMWR Recomm Rep 2020; 69:1.
  2. González Fernández L, Casas EC, Singh S, et al. New opportunities in tuberculosis prevention: implications for people living with HIV. J Int AIDS Soc 2020; 23:e25438.
  3. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV. Department of Health and Human Services. https://clinicalinfo.hiv.gov/en/guidelines/adult-and-adolescent-arv/whats-new-guidelines (Accessed on June 08, 2021).
  4. Panel on Opportunistic Infections in Adults and Adolescents with HIV. Guidelines for the prevention and treatment of opportunistic infections in adults and adolescents with HIV: recommendations from the Centers for Disease Control and Prevention, the National Institutes of Health, and the HIV Medicine Association of the Infectious Diseases Society of America. Available at https://clinicalinfo.hiv.gov/en/guidelines/adult-and-adolescent-opportunistic-infection/whats-new-guidelines (Accessed on June 08, 2021).
  5. World Health Organization. Latent TB Infection: Updated and consolidated guidelines for programmatic management, 2018. https://apps.who.int/iris/handle/10665/260233 (Accessed on March 06, 2018).
  6. Mehtani NJ, Puryear S, Pham P, et al. Infectious Diseases Learning Unit: Understanding Advances in the Treatment of Latent Tuberculosis Infection Among People With Human Immunodeficiency Virus. Open Forum Infect Dis 2021; 8:ofab319.
  7. Behr MA, Kaufmann E, Duffin J, et al. Latent Tuberculosis: Two Centuries of Confusion. Am J Respir Crit Care Med 2021; 204:142.
  8. Shwander SK, Ellner J. The human host: Immunology and susceptibility. In: Tuberculosis, 20th ed, Raviglione MC (Ed), Informa Healthcare, New York 2006. p.117.
  9. Menzies D, Fanning A, Yuan L, Fitzgerald M. Tuberculosis among health care workers. N Engl J Med 1995; 332:92.
  10. Daley CL, Small PM, Schecter GF, et al. An outbreak of tuberculosis with accelerated progression among persons infected with the human immunodeficiency virus. An analysis using restriction-fragment-length polymorphisms. N Engl J Med 1992; 326:231.
  11. Page KR, Godfrey-Fausset P, Chaisson RE. Tuberculosis-HIV coinfection: epidemiology, clinical aspects, and interventions. In: Reichman and Hershfield's Tuberculosis: A Comprehensive International Approach, Raviglione MC (Ed), Informa Healthcare, New York 2006. p.371.
  12. Selwyn PA, Hartel D, Lewis VA, et al. A prospective study of the risk of tuberculosis among intravenous drug users with human immunodeficiency virus infection. N Engl J Med 1989; 320:545.
  13. Wood R, Maartens G, Lombard CJ. Risk factors for developing tuberculosis in HIV-1-infected adults from communities with a low or very high incidence of tuberculosis. J Acquir Immune Defic Syndr 2000; 23:75.
  14. Comstock GW, Edwards LB, Livesay VT. Tuberculosis morbidity in the U.S. Navy: its distribution and decline. Am Rev Respir Dis 1974; 110:572.
  15. Elzi L, Schlegel M, Weber R, et al. Reducing tuberculosis incidence by tuberculin skin testing, preventive treatment, and antiretroviral therapy in an area of low tuberculosis transmission. Clin Infect Dis 2007; 44:94.
  16. Golub JE, Pronyk P, Mohapi L, et al. Isoniazid preventive therapy, HAART and tuberculosis risk in HIV-infected adults in South Africa: a prospective cohort. AIDS 2009; 23:631.
  17. Samandari T, Agizew TB, Nyirenda S, et al. 6-month versus 36-month isoniazid preventive treatment for tuberculosis in adults with HIV infection in Botswana: a randomised, double-blind, placebo-controlled trial. Lancet 2011; 377:1588.
  18. Martinson NA, Barnes GL, Moulton LH, et al. New regimens to prevent tuberculosis in adults with HIV infection. N Engl J Med 2011; 365:11.
  19. Sterling TR, Bethel J, Goldberg S, et al. The scope and impact of treatment of latent tuberculosis infection in the United States and Canada. Am J Respir Crit Care Med 2006; 173:927.
  20. Akolo C, Adetifa I, Shepperd S, Volmink J. Treatment of latent tuberculosis infection in HIV infected persons. Cochrane Database Syst Rev 2010; :CD000171.
  21. Whalen CC, Johnson JL, Okwera A, et al. A trial of three regimens to prevent tuberculosis in Ugandan adults infected with the human immunodeficiency virus. Uganda-Case Western Reserve University Research Collaboration. N Engl J Med 1997; 337:801.
  22. Cohen T, Lipsitch M, Walensky RP, Murray M. Beneficial and perverse effects of isoniazid preventive therapy for latent tuberculosis infection in HIV-tuberculosis coinfected populations. Proc Natl Acad Sci U S A 2006; 103:7042.
  23. Foster S, Godfrey-Faussett P, Porter J. Modelling the economic benefits of tuberculosis preventive therapy for people with HIV: the example of Zambia. AIDS 1997; 11:919.
  24. Targeted tuberculin testing and treatment of latent tuberculosis infection. This official statement of the American Thoracic Society was adopted by the ATS Board of Directors, July 1999. This is a Joint Statement 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 of America. (IDSA), September 1999, and the sections of this statement. Am J Respir Crit Care Med 2000; 161:S221.
  25. Harries AD, Zachariah R, Corbett EL, et al. The HIV-associated tuberculosis epidemic--when will we act? Lancet 2010; 375:1906.
  26. World Health Organization. Global tuberculosis report 2020. https://apps.who.int/iris/bitstream/handle/10665/336069/9789240013131-eng.pdf (Accessed on January 04, 2021).
  27. TEMPRANO ANRS 12136 Study Group, Danel C, Moh R, et al. A Trial of Early Antiretrovirals and Isoniazid Preventive Therapy in Africa. N Engl J Med 2015; 373:808.
  28. Golub JE, Cohn S, Saraceni V, et al. Long-term protection from isoniazid preventive therapy for tuberculosis in HIV-infected patients in a medium-burden tuberculosis setting: the TB/HIV in Rio (THRio) study. Clin Infect Dis 2015; 60:639.
  29. Campbell JR, Winters N, Menzies D. Absolute risk of tuberculosis among untreated populations with a positive tuberculin skin test or interferon-gamma release assay result: systematic review and meta-analysis. BMJ 2020; 368:m549.
  30. Den Boon S, Matteelli A, Ford N, Getahun H. Continuous isoniazid for the treatment of latent tuberculosis infection in people living with HIV. AIDS 2016; 30:797.
  31. Rangaka MX, Wilkinson RJ, Boulle A, et al. Isoniazid plus antiretroviral therapy to prevent tuberculosis: a randomised double-blind, placebo-controlled trial. Lancet 2014; 384:682.
  32. Mtei L, Matee M, Herfort O, et al. High rates of clinical and subclinical tuberculosis among HIV-infected ambulatory subjects in Tanzania. Clin Infect Dis 2005; 40:1500.
  33. Day JH, Charalambous S, Fielding KL, et al. Screening for tuberculosis prior to isoniazid preventive therapy among HIV-infected gold miners in South Africa. Int J Tuberc Lung Dis 2006; 10:523.
  34. World Health Organization. WHO operational handbook on tuberculosis Module 1: Prevention - Tuberculosis preventive treatment https://apps.who.int/iris/bitstream/handle/10665/331525/9789240002906-eng.pdf (Accessed on November 10, 2020).
  35. Hamada Y, Lujan J, Schenkel K, et al. Sensitivity and specificity of WHO's recommended four-symptom screening rule for tuberculosis in people living with HIV: a systematic review and meta-analysis. Lancet HIV 2018; 5:e515.
  36. Ngongondo M, Miyahara S, Hughes MD, et al. Hepatotoxicity During Isoniazid Preventive Therapy and Antiretroviral Therapy in People Living With HIV With Severe Immunosuppression: A Secondary Analysis of a Multi-Country Open-Label Randomized Controlled Clinical Trial. J Acquir Immune Defic Syndr 2018; 78:54.
  37. Saukkonen JJ, Cohn DL, Jasmer RM, et al. An official ATS statement: hepatotoxicity of antituberculosis therapy. Am J Respir Crit Care Med 2006; 174:935.
  38. Sterling TR, Villarino ME, Borisov AS, et al. Three months of rifapentine and isoniazid for latent tuberculosis infection. N Engl J Med 2011; 365:2155.
  39. Menzies D, Adjobimey M, Ruslami R, et al. Four Months of Rifampin or Nine Months of Isoniazid for Latent Tuberculosis in Adults. N Engl J Med 2018; 379:440.
  40. Sterling TR, Scott NA, Miro JM, et al. Three months of weekly rifapentine and isoniazid for treatment of Mycobacterium tuberculosis infection in HIV-coinfected persons. AIDS 2016; 30:1607.
  41. Campbell JR, Al-Jahdali H, Bah B, et al. Safety and Efficacy of Rifampin or Isoniazid Among People With Mycobacterium tuberculosis Infection and Living With Human Immunodeficiency Virus or Other Health Conditions: Post Hoc Analysis of 2 Randomized Trials. Clin Infect Dis 2021; 73:e3545.
  42. Guidelines for the Prevention and Treatment of Opportunistic Infections in Adults and Adolescents with HIV. Mycobacterium tuberculosis Infection and Disease. Available at: https://clinicalinfo.hiv.gov/en/guidelines/hiv-clinical-guidelines-adult-and-adolescent-opportunistic-infections/mycobacterium-0?view=full (Accessed on April 20, 2023).
  43. Rivero A, López-Cortés L, Castillo R, et al. [Randomized clinical trial investigating three chemoprophylaxis regimens for latent tuberculosis infection in HIV-infected patients]. Enferm Infecc Microbiol Clin 2007; 25:305.
  44. Martínez Alfaro E, Solera J, Serna E, et al. [Compliance, tolerance and effectiveness of a short chemoprophylaxis regimen for the treatment of tuberculosis]. Med Clin (Barc) 1998; 111:401.
  45. Martínez Alfaro EM, Cuadra F, Solera J, et al. [Evaluation of 2 tuberculosis chemoprophylaxis regimens in patients infected with human immunodeficiency virus. The GECMEI Group]. Med Clin (Barc) 2000; 115:161.
  46. Alvarez GG, Pease C, Menzies D. Chapter 6: Tuberculosis preventive treatment in adults. Canadian Tuberculosis Standards, 8th Edition 2022; 6:77.
  47. Centers for Disease Control and Prevention (CDC), American Thoracic Society. Update: adverse event data and revised American Thoracic Society/CDC recommendations against the use of rifampin and pyrazinamide for treatment of latent tuberculosis infection--United States, 2003. MMWR Morb Mortal Wkly Rep 2003; 52:735.
  48. US Food and Drug Administration. FDA Updates and Press Announcements on Nitrosamines in Rifampin and Rifapentine. https://www.fda.gov/drugs/drug-safety-and-availability/fda-works-mitigate-shortages-rifampin-and-rifapentine-after-manufacturers-find-nitrosamine (Accessed on October 06, 2020).
  49. OESTREICHER R, DRESSLER SH, MIDDLEBROOK G. Peripheral neuritis in tuberculous patients treated with isoniazid. Am Rev Tuberc 1954; 70:504.
  50. Belknap R, Holland D, Feng PJ, et al. Self-administered Versus Directly Observed Once-Weekly Isoniazid and Rifapentine Treatment of Latent Tuberculosis Infection: A Randomized Trial. Ann Intern Med 2017; 167:689.
  51. Borisov AS, Bamrah Morris S, Njie GJ, et al. Update of Recommendations for Use of Once-Weekly Isoniazid-Rifapentine Regimen to Treat Latent Mycobacterium tuberculosis Infection. MMWR Morb Mortal Wkly Rep 2018; 67:723.
  52. Yanes-Lane M, Ortiz-Brizuela E, Campbell JR, et al. Tuberculosis preventive therapy for people living with HIV: A systematic review and network meta-analysis. PLoS Med 2021; 18:e1003738.
  53. Hibma JE, Radtke KK, Dorman SE, et al. Rifapentine Population Pharmacokinetics and Dosing Recommendations for Latent Tuberculosis Infection. Am J Respir Crit Care Med 2020; 202:866.
  54. Churchyard G, Cárdenas V, Chihota V, et al. Annual Tuberculosis Preventive Therapy for Persons With HIV Infection : A Randomized Trial. Ann Intern Med 2021; 174:1367.
  55. Swindells S, Ramchandani R, Gupta A, et al. One Month of Rifapentine plus Isoniazid to Prevent HIV-Related Tuberculosis. N Engl J Med 2019; 380:1001.
  56. Campbell JR, Trajman A, Cook VJ, et al. Adverse events in adults with latent tuberculosis infection receiving daily rifampicin or isoniazid: post-hoc safety analysis of two randomised controlled trials. Lancet Infect Dis 2020; 20:318.
  57. Zenner D, Beer N, Harris RJ, et al. Treatment of Latent Tuberculosis Infection: An Updated Network Meta-analysis. Ann Intern Med 2017; 167:248.
  58. Ena J, Valls V. Short-course therapy with rifampin plus isoniazid, compared with standard therapy with isoniazid, for latent tuberculosis infection: a meta-analysis. Clin Infect Dis 2005; 40:670.
  59. Ronald LA, FitzGerald JM, Bartlett-Esquilant G, et al. Treatment with isoniazid or rifampin for latent tuberculosis infection: population-based study of hepatotoxicity, completion and costs. Eur Respir J 2020; 55.
  60. Russom M, Debesai M, Zeregabr M, et al. Serious hepatotoxicity following use of isoniazid preventive therapy in HIV patients in Eritrea. Pharmacol Res Perspect 2018; 6:e00423.
  61. Smith BM, Schwartzman K, Bartlett G, Menzies D. Adverse events associated with treatment of latent tuberculosis in the general population. CMAJ 2011; 183:E173.
  62. Comstock GW. How much isoniazid is needed for prevention of tuberculosis among immunocompetent adults? Int J Tuberc Lung Dis 1999; 3:847.
  63. Efficacy of various durations of isoniazid preventive therapy for tuberculosis: five years of follow-up in the IUAT trial. International Union Against Tuberculosis Committee on Prophylaxis. Bull World Health Organ 1982; 60:555.
  64. University of Liverpool. HIV Drug Interactions. Interaction Checker. https://www.hiv-druginteractions.org/ (Accessed on April 25, 2022).
  65. Wassner C, Bradley N, Lee Y. A Review and Clinical Understanding of Tenofovir: Tenofovir Disoproxil Fumarate versus Tenofovir Alafenamide. J Int Assoc Provid AIDS Care 2020; 19:2325958220919231.
  66. Cerrone M, Alfarisi O, Neary M, et al. Rifampicin effect on intracellular and plasma pharmacokinetics of tenofovir alafenamide. J Antimicrob Chemother 2019; 74:1670.
  67. Bhatt NB, Barau C, Amin A, et al. Pharmacokinetics of rifampin and isoniazid in tuberculosis-HIV-coinfected patients receiving nevirapine- or efavirenz-based antiretroviral treatment. Antimicrob Agents Chemother 2014; 58:3182.
  68. Crauwels H, van Heeswijk RP, Stevens M, et al. Clinical perspective on drug-drug interactions with the non-nucleoside reverse transcriptase inhibitor rilpivirine. AIDS Rev 2013; 15:87.
  69. Rajoli RKR, Curley P, Chiong J, et al. Predicting Drug-Drug Interactions Between Rifampicin and Long-Acting Cabotegravir and Rilpivirine Using Physiologically Based Pharmacokinetic Modeling. J Infect Dis 2019; 219:1735.
  70. Gagliardini R, Fabbiani M, Fortuna S, et al. Pharmacokinetics of etravirine in HIV-infected patients concomitantly treated with rifampin for tuberculosis. Infection 2014; 42:775.
  71. Yee KL, Khalilieh SG, Sanchez RI, et al. The Effect of Single and Multiple Doses of Rifampin on the Pharmacokinetics of Doravirine in Healthy Subjects. Clin Drug Investig 2017; 37:659.
  72. Maartens G, Decloedt E, Cohen K. Effectiveness and safety of antiretrovirals with rifampicin: crucial issues for high-burden countries. Antivir Ther 2009; 14:1039.
  73. 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 Syndr 2013; 62:21.
  74. 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.
  75. Weiner M, Egelund EF, Engle M, et al. Pharmacokinetic interaction of rifapentine and raltegravir in healthy volunteers. J Antimicrob Chemother 2014; 69:1079.
  76. Dooley KE, Savic R, Gupte A, et al. Once-weekly rifapentine and isoniazid for tuberculosis prevention in patients with HIV taking dolutegravir-based antiretroviral therapy: a phase 1/2 trial. Lancet HIV 2020; 7:e401.
  77. Saag MS, Gandhi RT, Hoy JF, et al. Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2020 Recommendations of the International Antiviral Society-USA Panel. JAMA 2020; 324:1651.
  78. Brooks KM, George JM, Pau AK, et al. Cytokine-Mediated Systemic Adverse Drug Reactions in a Drug-Drug Interaction Study of Dolutegravir With Once-Weekly Isoniazid and Rifapentine. Clin Infect Dis 2018; 67:193.
  79. 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:e47400.
  80. Churchyard GJ, Fielding KL, Lewis JJ, et al. A trial of mass isoniazid preventive therapy for tuberculosis control. N Engl J Med 2014; 370:301.
  81. Kopanoff DE, Snider DE Jr, Caras GJ. Isoniazid-related hepatitis: a U.S. Public Health Service cooperative surveillance study. Am Rev Respir Dis 1978; 117:991.
  82. Gordin F, Chaisson RE, Matts JP, et al. Rifampin and pyrazinamide vs isoniazid for prevention of tuberculosis in HIV-infected persons: an international randomized trial. Terry Beirn Community Programs for Clinical Research on AIDS, the Adult AIDS Clinical Trials Group, the Pan American Health Organization, and the Centers for Disease Control and Prevention Study Group. JAMA 2000; 283:1445.
  83. Gao XF, Wang L, Liu GJ, et al. Rifampicin plus pyrazinamide versus isoniazid for treating latent tuberculosis infection: a meta-analysis. Int J Tuberc Lung Dis 2006; 10:1080.
  84. Lawn SD, Myer L, Edwards D, et al. Short-term and long-term risk of tuberculosis associated with CD4 cell recovery during antiretroviral therapy in South Africa. AIDS 2009; 23:1717.
  85. Cohen MS, Chen YQ, McCauley M, et al. Prevention of HIV-1 infection with early antiretroviral therapy. N Engl J Med 2011; 365:493.
  86. Severe P, Juste MA, Ambroise A, et al. Early versus standard antiretroviral therapy for HIV-infected adults in Haiti. N Engl J Med 2010; 363:257.
  87. INSIGHT START Study Group, Lundgren JD, Babiker AG, et al. Initiation of Antiretroviral Therapy in Early Asymptomatic HIV Infection. N Engl J Med 2015; 373:795.
Topic 8011 Version 65.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟