Suggestions
Guide for authors
Searcher
Journal Information
Visits
189
SEPAR/ALAT Voice
Full text access
Available online 16 January 2026

Recommendations on the Diagnosis and Treatment of Tuberculosis Infection: SEPAR/SEIMC/Spanish Ministry of Health Consensus Statement

Visits
189
José Antonio Camineroa,b,1, Luís Anibarroc,d,1,
Corresponding author
, Eva Taberneroe,f, Fernando Alcaideg,h, Guillermo Pérez-Mendozai,j, Joan-Pau Milletk,l,m, Remedio Gunan,o, Maria-Luiza de Souza-Galvãop, Paula López-Roaq, Adrián Sánchez-Montalvár,s,t, José A. Gullón-Blancou,2, Verónica González-Galánv,x,2
a Prof. Head of the Pulmonology Service at the Dr. Negrín General University Hospital, Las Palmas de GC, Spain
b Director of Scientific Activities ALOSA TB ACADEMY, Spain
c Tuberculosis Unit, Infectious Diseases, Internal Medicine Department, University Hospital Complex, Pontevedra, Spain
d Immunology Research Group, Galicia Sur Health Research Institute, Spain (ISS-Galicia Sur) Uvigo, Vigo (Pontevedra), Spain
e Pulmonology Service, Cruces University Hospital (OSI EEC), Barakaldo, Spain
f BioBizkaia Health Research Institute, Spain
g Microbiology Department, Hospital Universitari de Bellvitge-IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
h Department of Pathology and Experimental Therapy, University of Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain
i Pulmonology Department, Dr. Negrin University Hospital of Gran Canaria, Las Palmas de GC, Spain
j PII-TB&MNT & SEPAR Study Group, Spain
k Epidemiology Service, Barcelona Public Health Agency, Barcelona, Spain
l Serveis Clínics Social and Healthcare Centre, Barcelona, Spain
m CIBER of Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
n Microbiology Service, General University Hospital Consortium of Valencia, Valencia, Spain
o Departament of Microbiology, Faculty of Medicine, University of Valencia, Valencia, Spain
p Tuberculosis Unit, Vall d’Hebron-Drassanes, Vall d’Hebron University Hospital, Barcelona, Spain
q Microbiology Service, 12 de Octubre University Hospital, Madrid, Spain
r International Health Unit Vall d’Hebron-Drassanes Centre, Infectious Diseases Department, Vall d’Hebron University Hospital, PROSICS Barcelona, Autonomous University of Barcelona, Spain
s Mycobacterial Infections Study Group (GEIM) of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), Madrid, Spain
t Biomedical Research Centre in Infectious Diseases Network (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
u Pulmonology Clinical Management Unit, San Agustín University Hospital, Avilés, Spain
v Micobacteria Laboratory, Microbiology Section, UCEIM, Virgen de Valme University Hospital, South Seville Healthcare Management Area, Seville, Spain
x Clinical and Molecular Microbiology Group, Biomedicine Institute of Seville, Spain
Ver más
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (3)
fig0005
fig0010
fig0015
Show moreShow less
Tables (4)
Table 1. IGRA techniques used for the diagnosis of tuberculosis infection.
Tables
Table 2. Risk of developing tuberculosis among individuals infected with M. tuberculosis.
Tables
Table 3. TPT regimens shown to be effective. Doses and observations.
Tables
Table 4. Adverse effects of drugs used in tuberculosis preventive treatment.
Tables
Show moreShow less
Abstract

All strategies on the pathway to tuberculosis (TB) elimination prioritise, among other measures, addressing tuberculosis infection (TBI), particularly the identification of individuals and population groups who should be candidates for tuberculosis preventive treatment (TPT).

In Spain, a TBI test is required before recommending TPT. An interferon-gamma release assay (IGRA) is preferred over the tuberculin skin test (TST), although the latter may be used in settings where IGRAs are not available or when deemed necessary to increase diagnostic sensitivity. On the other hand, new skin tests employing specific antigens (TBST) may play a key role.

As a general principle, screening for TBI should include all individuals at high risk for progressing from TBI to TB, as described in this guideline, prioritising pulmonary TB contacts, people living with HIV, immunocompromised individuals and those in other situations associated with an elevated risk of developing TB.

Once TBI has been diagnosed in these high-risk groups, the algorithm set out in this guideline should be applied to rule out TB disease. Once TB has been excluded, TPT should be recommended. The preferred regimen is daily isoniazid (H) plus rifampicin (R) for 3 months (3HR). However, once rifapentine becomes available in Spain, both the 1-month daily (1HP) and the 3-month weekly (3HP) regimens combining H and rifapentine (P) may also be used. Finally, measures must be taken to ensure adherence to TPT and to monitor and manage potential drug-related adverse effects.

Keywords:
Tuberculosis
Tuberculosis infection (TBI)
Latent tuberculosis infection
Tuberculosis preventive treatment (TPT)
Interferon-gamma release assay (IGRA)
Tuberculin skin test (TST)
Consensus
Abbreviations:
BCG
CFP-10
ESAT-6
H
HIV
IFN-γ
IGRA
MDR-TB
P
PCR
R
SEIMC
SEPAR
TB
TBST
TBI
TNF
TPT
TST
WHO
Graphical abstract
Full Text
Considerations concerning terminology

In recent years, great advances have been made in understanding the pathogenesis of TB and the complex interaction between this pathogen and the immune response. Infection by Mycobacterium tuberculosis complex is now recognised to be a continuous spectrum from exposure to the mycobacterium up to overt disease. Hence, numerous authors consider the traditional dichotomous terminology of latent infection versus active tuberculosis to be insufficient or inadequate [1,2].

Nonetheless, this classification has been used effectively for decades, because it is simple, practical, and easy to understand for patients and healthcare workers: an individual with a latent infection could be treated with just one drug and would not spread the disease to others, while an individual with a diagnosis of TB disease would require several drugs and could transmit the disease, even in the absence of signs and symptoms in the case of respiratory TB.

Currently, there is no consensus among researchers regarding the use of this terminology. Terms such as TBI, latent TB infection, infection by M. tuberculosis complex, incipient TB, subclinical TB, and asymptomatic TB overlap and may cause confusion among patients, clinicians, and even TB experts themselves [3–6]. Further, given the indirect diagnostic techniques currently used, these terms cannot distinguish between the persistence of the mycobacterium in a quiescent or low-activity state and the presence of an adaptive immunological response after exposure to M. tuberculosis complex antigens.

Acknowledging these limitations, in this document, we have adopted the terminology recommended by the World Health Organization (WHO) and other expert groups, which suggest using the term “TB infection (TBI)” rather than latent TB. TBI is defined as the presence of an acquired immune response detectable by currently available diagnostic techniques against M. tuberculosis antigens (although not exclusive to this species) with no signs or symptoms of the disease. Consequently, we use the term “tuberculosis preventive treatment (TPT)” instead of “treatment of latent TB” [7]. Accordingly, we have opted to use the terms TB disease or simply “TB”, instead of “active TB”.

The importance of addressing tuberculosis infection

Tuberculosis remains the leading infectious cause of death worldwide. According to the WHO, out of the 10.8 million new cases estimated in 2023, 1.25 million died [8]. Notably, TBI affects a quarter of the world's population [9]. Recognising and diagnosing TBI, and providing treatment at this stage, is key to preventing progression to TB disease and contributing to disease control worldwide [10,11].

In 2014, the WHO launched the END TB strategy with the ambitious goals of reducing TB incidence by 90% and TB-related deaths by 95% by 2035, compared to 2015. In this strategy, tackling TBI is a key measure to achieve these goals. In Spain, notified TB cases have progressively decreased over recent decades to 10 per 100,000 [12], allowing the country to be considered low-incidence. Despite this, the burden of TBI remains substantial and therefore must be addressed. TPT in populations at risk of developing TB has proven to be both effective and cost-effective in reducing TB incidence [13,14].

However, TPT is not without adverse effects and may entail significant costs. It is therefore essential to carefully identify the population at the greatest risk of progression to TB, ensuring the benefits outweigh the potential adverse effects. Factors such as demographic change, increasing use of immunosuppressive therapies and social vulnerability have resulted in a rise in the number of individuals with TBI who develop TB disease. It should be emphasised that TPT is crucial not only from a medical and public health perspective, but also from one of social justice. For the first time, the 2024 WHO report included data on the economic impact of TB on households, indicating that half of those in which a member has TB face catastrophic costs. In this context, preventing progression from TBI to the disease is also a measure that may mitigate social and healthcare inequalities [8].

The introduction of diagnostic techniques such as IGRAs and new skin tests based on specific antigens, along with shorter and more effective TPT regimens, has led the Spanish Society of Pulmonology and Thoracic Surgery (SEPAR) and the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), within the framework of the agreement signed with the Spanish Ministry of Health's Division for HIV, Sexually Transmitted Infections, Viral Hepatitis, and Tuberculosis Control, to consider it necessary to develop a joint statement updating the national consensus on the diagnosis and treatment of TBI from 2010 [15] and 2016 [16]. This document is intended for healthcare professionals caring for populations at risk of having TBI and at risk of progression to TB disease.

In this consensus statement, a multidisciplinary group of experts addresses the integrated and sustainable management of TBI in Spain, with the objective of adapting the guidelines of the END TB strategy, the commitments made at the 2023 United Nations High-Level Meeting on the Fight Against Tuberculosis and WHO recommendations to recent technological advances and national requirements.

Pathogenesis of tuberculosis. The continuum from exposure to infection and disease

The pathogenesis of TB is a complex process involving a dynamic interaction between the invading microorganism and the host immune system. This interaction determines the course of the infection, from an initial innate immune response to the containment or clearance of the infection by the adaptive immune response in later stages, or the progression of the infection to disease. Understanding these processes is essential for the development of strategies for the diagnosis, monitoring and treatment of M. tuberculosis.

Following exposure to M. tuberculosis, the bacilli are inhaled into the respiratory tract and cross the mucosal barriers. The presence of the microorganism in lung tissue triggers an innate immune response, mainly involving resident alveolar macrophages, dendritic cells and neutrophils [17,18]. This effector immune response may clear the bacilli, eliminating the invader without leaving an imprint on immunological memory, and therefore, with current diagnostic tools, such exposed individuals are indistinguishable from those who have not been exposed to M. tuberculosis.

When clearance does not occur, the mycobacteria migrate to the lymph nodes via immune system cells and replicate in macrophages. In the lymph nodes, antigen-presenting cells interact with naïve T cells, stimulating their differentiation into active phenotypes that migrate to the lung and initiate an adaptive immune response, mainly mediated by interferon gamma (IFN-γ). This leads to macrophage activation, the production of stimulating cytokines and the release of microbicidal factors that contribute to bacterial control.

Replication of M. tuberculosis may be inhibited by a balanced immune response that restricts bacterial replication in macrophages and traps extracellular bacteria in containment structures called granulomas. In humans, granulomas consist of a core region composed of infected macrophages, which have differentiated into multinucleated giant cells, epithelioid cells, foamy macrophages and neutrophils, surrounded by lymphocytes (CD4 and CD8 T cells and B cells) and fibroblasts that form a peripheral fibrotic capsule [19]. In TBI, bacilli persist in the central region in a state of low metabolic activity, whereas in TB disease, they replicate within the granuloma.

Various survival and immune evasion mechanisms have been described, such as the creation of a microenvironment that hinders immune cell activity and inhibits the fusion of the phagolysosome through, among other mechanisms, the release of proteins such as 6kDa early secreted antigenic target (ESAT-6) and 10kDa culture filtrate protein (CFP-10) [20,21].

Granulomas recruit macrophages that become infected and may subsequently migrate to other sites, potentially contributing to disease dissemination [22,23]. Failure to contain M. tuberculosis within the granuloma may allow the bacilli to escape, spread through the circulatory system and cause pulmonary or extrapulmonary TB.

Immune responses to TBI or TB vary depending on age, sex and immune status [24]. It should be emphasised that the immune response in TBI is not static. Certain factors may induce subclinical reactivation, in which bacilli remain in a state of low replication but are still capable of being activated in situations that compromise the immune system. These episodes of partial reactivation do not always result in disease with clinical manifestations, but could increase the bacterial burden and risk of transmission [25].

Thus, the simplified concept of individuals being classified as non-infected, with TBI or with TB disease has been replaced by a broader continuum of possibilities, where interactions between the environment, the host and the pathogen determine an individual's status at any given time. This paradigm shift, however, does not invalidate the diagnostic and treatment strategies for TBI in individuals at high risk of reactivation, given that they have shown to be effective in the control of TB in the community [17].

Current picture of tuberculosis infection and disease worldwide and in Spain

Worldwide, around 10.8 million people develop TB every year. In 2023, 1.25 million people died from TB, including 161,000 living with HIV [8]. In Spain, 4207 cases were reported in 2023, with an incidence 8.2 cases per 100,000 among autochthonous individuals (n=3944), representing a 7% increase compared to 2022, but a 22.5% decline relative to 2015, when 4913 cases were reported (10.6/105). In 2023, 241 deaths were attributed to TB, a mortality of 0.5 per 100,000 (0.8 in men and 0.23 in women). Based on these data, Spain is considered a low-incidence country [12,26]. Nevertheless, further efforts are needed to develop a national plan for TB prevention and control [27].

Globally, the prevalence of TBI lies between 21.2% and 24.8%, corresponding to some 2 thousand million people infected [9,28]. There is a downward trend over the years, with rates decreasing from a WHO estimate in 1999 that a third of the world's population was infected to a mean of 23.7% reported in 2019 [29,30]. TBI prevalence is related to transmission rates and duration of the infection [31,32], and varies across continents, regions, age groups and other risk factors. By WHO region, the highest prevalence of TBI is found in Southeast Asia (30.8%), followed by the Western Pacific (27.7%), Africa (22.4%), the Eastern Mediterranean (16.3%), Europe (13.7%), and the Americas (11%) [9].

In Spain, the prevalence of TBI varies by regions. Among TB contact cases in Madrid, Galicia and Barcelona, the prevalence rates in 2022–23 were 20.8%, 27.2% and 23.4%, respectively [33–35]. In migrants from high-prevalence countries and in vulnerable situations, TBI rates vary depending on their origin and context, from 17.9% to 40% [36–38], similar to rates in their regions of origin. Notably, the highest rates of TBI are found among people deprived of liberty—about 50–54%—though prevalence varies by age, region, and length of incarceration. When IGRAs are used for diagnosis, lower rates (19–30.1%) are reported [39–41].

Healthcare workers are at increased risk of exposure, and the prevalence observed in this group ranges from 11.1% to 15.5%, depending on their origin and professional profile [42,43].

Among vulnerable populations at a high risk of progression to TB, the prevalence of TBI varies from 27.2% in nursing homes [44], to 2.6% in minors who visit their parents’ high-TB-incidence country of origin [45]. In patients who are candidates for biological therapies, a prevalence of TBI of around 4.5% has been observed [46–48].

Diagnosis of tuberculosis infection

The diagnosis of TBI is indirect and based on the detection of a host immune response to M. tuberculosis complex antigens, without assessing the viability of the microorganism. The currently available diagnostic tests are described below, highlighting their strengths and limitations.

Interferon gamma release assays

These are in vitro blood tests that measure either the amount of IFN-γ released or the number of T cells releasing IFN-γ in response to specific antigens of M. tuberculosis complex (ESAT-6 and CFP-10). These antigens are absent from the Mycobacterium bovis BCG vaccine and most non-tuberculous mycobacteria, except for Mycobacteriummarinum, Mycobacterium kansasii, Mycobacterium szulgai and Mycobacterium flavescens. Several commercial kits are available [49] (Table 1), the most widely studied being:

  • a)

    QuantiFERON-TB Gold Plus (QFT-Plus) [50–52]. This test is based on four tubes, two of which contain specific polypeptide antigens (CFP-10 and ESAT-6) that stimulate CD4+ and CD8+ T cells, potentially relevant for identifying recent infections or active infectious states. The response is measured using ELISA or chemiluminescence immunoassays [53].

  • b)

    T-SPOT.TB [54]. This test quantifies IFN-γ-producing T cells, and readings are based on ELISPOT assays.

Table 1.

IGRA techniques used for the diagnosis of tuberculosis infection.

Technique  T-SPOT.TB  QFT-plus  LIAISON QFTplus  AdvanSure™TB-IGRA  WantaiTB-IGRA  Standard ETB-feron  QIAreach QFT  ichroma™IGRA-TB  VIDAS™TB-IGRA  T-Track® TB 
Manufacturer  Oxford, Immunotec, UK  Hilden, Germany  DiaSorin S.P.A., Italy  LG Life sciences, South Korea  Wantai, China  SD Biosensor, South Korea  Hilden, Germany  Boditech Med Inc., South Korea  bioMerieux SA, Marcy-l’Etoile, France  Mikrogen GmbH, Neuried, Germany 
Sample  PBMC  Whole blood  Whole blood  Whole blood  Whole blood  Whole blood  Whole blood  Whole blood  Whole blood  Whole blood 
Type of antigen  Panel A: ESAT-6Panel B: CFP-10  ESAT-6, CFP-10  ESAT-6, CFP-10  ESAT-6, CFP-10  ESAT-6, CFP-10  ESAT-6, CFP-10, and TB7.7 recombinant proteins  Peptide cocktail simulating ESAT-6 and CFP-10  ESAT-6, CFP-10  ESAT-6, CFP-10  ESAT-6, CFP-10 
Tubes  1 tube  Nil, TB1, TB2, and mitogen tubes  PC, TB-A, TB-B, and negative control  Nil, TB1, TB2, and mitogen tubes  Nil, TB1, TB2, and mitogen tubes  Nil, TB1, TB2, and mitogen tubes  Blood collection tube and processing tube  Nil, TB1, TB2, and mitogen tubes  Nil, TB1, TB2, and mitogen tubes  Nil, TB1, B2, and mitogen tubes 
Technology platform  ELISPOT  ELISA  CLIA  CLIA  ELISA  ELISA  LFIA  FIA  ELISA  RT-qPCR 
Results  Number of cells that release IFN-γ  Amount of IFN-γ released by CD4/CD8 T cells  Amount of IFN-γ released by CD4/CD8 T cells  Amount of IFN-γ released by CD4/CD8 T cells  Amount of IFN-γ released by CD4/CD8 T cells  Amount of IFN-γ released by CD4/CD8 T cells  Amount of IFN-γ released by CD4/CD8 T cells  Amount of IFN-γ released by CD4/CD8 T cells  Amount of IFN-γ released by CD4/CD8 T cells  Changes in IFNG and CXCL10 mRNA levels 
Sensitivity  83%  91.4%  78.6%  98.48%  86.4%  88%  96.5%  95.76%  97.5%  94.9% 
Specificity  83%  97.8%  94.7%  97.95%  85.9%  95%  94.2%  88%  97.6%  93.8% 

PBMC: peripheral blood mononuclear cells; ELISPOT: enzyme-linked immunosorbent spot; ELISA: enzyme-linked immunosorbent assay; CLIA: chemiluminescent immunoassay; IFN-γ: interferon gamma; LFIA: lateral flow immunoassay; FIA: fluorescence immunoassay; T.SPOT: T cell spot test; QFT-Plus: QuantiFERON-TB Gold-In-Tube; IGRA: Interferon gamma release assay, QIAreach: Qiagen kit for detecting M. tuberculosis; VIDAS: registered trademark of bioMérieux; ESAT-6; 6kDa early secreted antigenic target; CFP-10: 10kDa culture filtrate antigen, coded by the esxB gene and secreted to the medium; Nil: Null or negative control; PC: positive control; TB-A: tuberculosis A; TB-B tuberculosis-B; NC: negative control; RT-qPCR, reverse transcription quantitative polymerase chain reaction; IFN-G: interferon gamma gene; CXCL: chemokine (C-X-C motif) ligand.

Adapted from Li LS, Yang L, Zhuang L, Ye ZY, Zhao WG, Gong WP. From immunology to artificial intelligence: revolutionizing latent tuberculosis infection diagnosis with machine learning. Mil Med Res. 2023 Nov 28;10 [1]:58. doi:10.1186/s40779-023-00490-8.

Results are interpreted as positive, negative or indeterminate, based on the response to stimulation with specific antigens, taking into account the internal positive and negative controls.

Indeterminate results are uncommon and can be due to: (1) pre-analytical or analytical errors, or failure of the positive and/or negative controls, and (2) patient-related factors: viral infections that impair cellular immunity (e.g. HIV with low CD4 T cell counts), immunosuppressive medications (corticosteroids, chemotherapy, biologics), autoimmune and/or haematological malignant disease, chronic conditions (diabetes, chronic kidney failure, advanced liver disease) or extreme age.

The main strength of IGRAs is their high specificity, as results are not affected by prior BCG vaccination, and only a few non-tuberculous mycobacterial species interfere with the results. Moreover, IGRAs have been shown to be more sensitive than TSTs in immunosuppression, slightly more so with the ELISPOT assay [55], they require only a single patient visit, and provide objective results.

Their main limitations relate to their higher cost, the need for blood sampling and the requirements for laboratory facilities and specific technical training. In young children, their use is limited due to the weaker supporting evidence and higher rates of indeterminate results. As with TSTs, the sensitivity may be lower in immunosuppressed individuals, and the results cannot distinguish between TBI and TB [56–58].

Tuberculin skin test

The TST is the oldest diagnostic test currently recommended. It measures the delayed-type hypersensitivity response mediated by T cells, through an intradermal injection of a purified protein derivative (PPD) of M. tuberculosis into the upper third of the flexor surface of the forearm (Mantoux method) [59].

For interpretation, the diameter of the induration (skin thickening) should be measured rather than the erythema. Although different cut-off points have been accepted depending on individual and epidemiological risk factors for developing the disease [60], the test should be performed in people for whom a positive result would prompt preventive treatment. In this context, an induration of ≥5mm would be the main outcome used as a cut-off for recommending TPT. Higher cut-offs (≥10 or ≥15mm) may be appropriate in epidemiological studies or in individual cases where the efficacy of TPT has not been sufficiently demonstrated.

The main strengths of the TST are its low cost, wide availability, and performance at the point of care, without the need for laboratory facilities. In addition, the test is supported by almost a century of evidence of its utility, particularly in children.

Its limitations are related to the lack of specificity, with positive reactions occurring due to prior BCG vaccination or exposure to non-tuberculous mycobacteria. Further, it has variable sensitivity, which is influenced by immunosuppression. The test has no internal positive and/or negative controls. Operationally, it requires two patient visits (one for the injection, and another 48–72h later, for reading the response), and the results are more subjective and less reproducible than those of IGRAs.

Skin tests with specific antigens of M. tuberculosis complex (TBST)

In recent years, skin tests have been developed and marketed that, unlike the TST, only contain highly specific antigens of the M. tuberculosis complex. Consequently, their specificity is higher than that of the TST [61–64]. They are also performed using the Mantoux method, with the transverse diameter of the induration measured after 48–72h. The results are interpreted according to the same criteria as the TST [65].

Various commercial tests are available including:

  • a)

    Cy-Tb (SIILTIBCY®) formerly known as the C-Tb test. It contains two recombinant proteins, ESAT-6 and CFP-10. It is the only test currently approved and marketed in Spain.

  • b)

    Diaskintest®. It is based on a recombinant protein produced using a genetically modified culture of Escherichia coli BL21 (DE3)/pCFP-ESAT.

  • c)

    C-TST. The active ingredient is a recombinant fusion protein ESAT-6–CFP-10, expressed in genetically modified E. coli.

Recent innovations in the diagnosis of tuberculous infection

Alongside numerous improvements in established testing kits, rapid or ultra-rapid IGRAs are being developed, using miniaturised microfluidic systems and flow cytometry-based platforms to achieve same-day results (within 24h). In addition, researchers are exploring immunological biomarkers and molecular and transcriptomic tests, with higher positive predictive values for disease progression, but these are not yet available for clinical use [49,55,66–68]. The ultimate goal is to develop a test that not only diagnoses TBI but also identifies individuals at highest risk of progression to TB disease, thereby enabling more targeted and efficient TPT.

Current diagnostic algorithms

The diagnosis of TBI requires integrating microbiological, clinical and epidemiological factors. In high-prevalence regions, which are often those with fewer economic resources, WHO recommends prioritising TSTs as the initial screening tool, with subsequent confirmation by IGRA (for example, in individuals previously vaccinated with BCG) [65,69,70].

In low-prevalence regions, which typically have greater economic and social resources – such as Spain, the recommendation is to perform an IGRA, particularly in people who have received the BCG vaccine. In certain circumstances, a combination of TST and IGRA may be considered, or even the simultaneous use of two different IGRA techniques, to enhance diagnostic sensitivity [65,71–73].

Recent evidence indicates that a negative IGRA or TST result has an almost 100% negative predictive value for the development of disease. This requires that testing is technically well performed, before the initiation of any immunosuppressive or immunomodulatory treatment, and in the case of contacts, at least 8–12 weeks after exposure (window period) [56,70,74–78].

Several studies have also demonstrated the superiority of IGRAs over TSTs in predicting the development of the disease [69,74,79–81].

In children <5 years of age (and especially in those <2 years of age), TST (or TBST) plus IGRA is recommended to achieve maximum diagnostic sensitivity. In immunocompetent individuals>5 years of age, either TST (or TBST) or IGRA may be used. IGRAs or TBSTs, are particularly recommended for children who have received BCG vaccination [82].

Pending further data on the role of new antigen-specific skin tests, this guideline supports the use of IGRAs as the initial diagnostic test, reserving TSTs for settings with limited access to IGRAs or operational constraints, such as large-scale community contact tracing studies (e.g. schools, universities, or workplaces). In situations where higher diagnostic sensitivity is needed, both tests may be performed.

In any case, it should be noted that no TBI diagnostic technique, whether alone or in combination, can guarantee with absolute certainty the prevention of tuberculosis disease [83]

Tuberculosis preventive treatmentIdentification of populations requiring preventive treatment

It is estimated that 5–15% of individuals infected with M. tuberculosis complex will develop TB disease during their lifetime. Therefore, when selecting populations to receive TPT, emphasis should be placed on those groups at the highest risk of progression to disease. This requires consideration of the context in which testing is performed and the strategy applied, whether through contact tracing or population screening.

In the context of a TB control programme, it is essential to consider the number of infected individuals who must be treated to prevent one case of disease, together with the efficacy of, and adherence to, preventive treatment. In short, the aim is to maximise the efficiency of this intervention. Accordingly, the highest-priority group for evaluation is contacts of patients with pulmonary TB, in whom the risk of progression to disease is markedly increased, particularly among household and close contacts. This risk varies as a function of age, being highest in children under 5 years, although in older individuals it may reach up to 28% [84–86]. Therefore, all contacts of a pulmonary TB case should be prioritised for TPT, irrespective of age, especially if they have shared living space for more than 3–6h a day.

In contacts at high risk of rapid progression to TB disease (children under 5 years or individuals with severe immunosuppression), initiation of TPT should be considered even in the presence of a negative IGRA or TST result. In individuals exposed to patients with pulmonary TB who initially test negative by TST or IGRA, testing should be repeated 8–12 weeks after the latest exposure; TPT is indicated if the result subsequently becomes positive [77,87,88]. Reverse contact tracing (to identify the source of infection) should be performed in cases of childhood TB, including extrapulmonary forms [27].

In clinical practice, the risk of TB development must be balanced against the potential toxicity of TPT. Table 2 summarises the risk factors for developing TB in infected individuals, classified as high, intermediate, or low risk [89]. In this context, screening for TBI should be performed in individuals at high-risk, prioritising people living with HIV, as well as the following groups: patients starting treatment with TNF inhibitors, those with end-stage renal disease, solid-organ or haematological transplant recipients, and individuals with silicosis, where the benefit of TPT clearly outweighs the associated risks [7,90,91]. It has also been suggested that the presence of fibrotic pulmonary lesions2cm2 in infected individuals with no previous TB treatment should be an indication for TPT [92–95]. TBI testing may also be considered on a case-by-case basis in individuals receiving biological agents such as IL-6R or JAK inhibitors, where the risk of developing TB disease is not negligible [96,97].

Table 2.

Risk of developing tuberculosis among individuals infected with M. tuberculosis.

Risk factor  Relative risk compared to the general population 
High risk (tuberculosis infection tracing)
HIV  10–100 
Solid organ or blood transplant  20–70 
Close contacts  15 
Silicosis  2.8 
Chronic kidney failure requiring dialysis  6.9–52.5 
Apical fibronodular changes on X-ray  6–19 
Treatment with TNF inhibitors  1.6–25.1 
Moderate risk
Treatment with corticosteroids  2.8–7.7 
Diabetes mellitus  1.6–7.83 
Immigrant from high-TB-prevalence countries  2.9–5.3 
Treatment with biologics:a   
IL-6R inhibitors (tocilizumab, sarilumab 
JAK inhibitors (baricitinib, upadacitinib, tofacitinib)   
Low risk
Body mass index20kg/m2  2–3 
Smoker (20cigarettes/day)  2–3.4 
Lung granuloma on chest X-ray 
Treatment with biologics:a   
CTL4Ig (abatacept), IL-12/23 inhibitor (ustekinumab), IL-23 inhibitors (guselkumab, risankizumab), IL-17 inhibitors (secukinumab, ixekizumab)   
a

Picchianti-Diamanti A, Aiello A, De Lorenzo C, Migliori GB, Goletti D. Management of tuberculosis risk, screening and preventive therapy in patients with chronic autoimmune arthritis undergoing biotechnological and targeted immunosuppressive agents. Front Immunol. 2025 Feb 3;16:1494283.

Adapted from Ai JW, Ruan QL, Liu QH, Zhang WH. Updates on the risk factors for latent tuberculosis reactivation and their managements. Emerg Microbes Infect. 2016 Feb 3;5 [2]: e10.

In other scenarios, such as patients with diabetes, gastrectomy, alcohol misuse, solid-organ or haematological malignancies, or long-term corticosteroid therapy, the benefits of TPT are less certain; therefore, TBI screening and treatment should be considered on an individual basis [98,99]. In individuals diagnosed with TBI during contact tracing of a patient with pulmonary TB who decline TPT, clinical and radiological follow-up should be undertaken every 6–12 months for two years (the period of highest risk of progression to TB disease after infection). [100,101]

In frontline healthcare workers, people deprived of liberty, migrants from high TB-prevalence countries (particularly minors and those who have arrived within the past five years), people experiencing homelessness and individuals with substance use disorders may also be considered for TPT on a case-by-case basis [7,38,102]. The decision to recommend TPT in patients previously treated for TB or TBI should likewise be individualised. In such situations, TBI tests are usually positive and cannot distinguish whether reinfection has occurred. It should also be noted that individuals with a prior episode of TB are at increased risk of developing disease again if reinfected with M. tuberculosis[103,104].

Algorithm to rule out tuberculosis disease

Before starting preventive treatment, it is essential to rule out TB disease.

In immunocompetent individuals without symptoms suggestive of TB (cough, haemoptysis, fever/low-grade fever, weight loss, night sweats), chest radiography has a sensitivity exceeding 90–95%; therefore, the absence of lesions can be reasonably used to rule out pulmonary TB [105].

In people living with HIV, symptoms have a sensitivity of 83%, but low specificity (38%), which is not improved by radiology (20%). However, when combined with C-reactive protein levels>5mg/L, specificity increases to 76%, and to 90% in outpatients on antiretroviral therapy [105,106].

Fig. 1 shows an algorithm for ruling out TB disease in infected individuals. It highlights the performance of a chest X-ray to exclude TB. Rapid molecular techniques (real-time PCR and others) should be considered in individuals with symptoms and/or radiological findings suggestive of TB [27].

Fig. 1.

Algorithm to rule out tuberculosis in asymptomatic individuals. * Consider diagnostic evaluation in patients with severe immunosuppression, or if C-reactive protein>5mg/L in patients living with human immunodeficiency virus.

Preventive treatment options for tuberculosis infection

The first evidence suggesting that TPT could be effective was published more than 50 years ago, demonstrating the efficacy of long-term H monotherapy [92,107–117]. At that time, H was the only drug with both strong activity and good tolerability against the M. tuberculosis complex, and all studies conducted over the following three to four decades were based on H. Numerous clinical trials and publications consistently showed its efficacy when administered for six to nine months.

Although maximum efficacy was achieved with 9–10-month regimens provided adherence was good [118,119], field studies demonstrated that 6 months was similarly effective, as longer regimens of 9–12 months were associated with higher treatment discontinuation rates [92,118,119]. Based on this evidence and some subsequently published meta-analyses [120], the tendency for decades has been to recommend a 6-month regimen with H [121–125].

Nonetheless, it has been known for decades that the main activity of H lies in its bactericidal properties [126–128], acting mainly against continuously replicating bacilli, while it has limited sterilising effect against bacilli in a latent or semi-latent state. Given this, H cannot be considered the best drug for TPT, since its efficacy depends on prolonged administration over many months (to allow the bacilli in a latent/semi-latent state to replicate).

An ideal TPT drug should have sterilising activity, since this allows treatment durations to be shortened [126–128]. In recent years, numerous randomised clinical trials have evaluated such drugs, mainly rifamycins, particularly P and R [113,115,120,129–139]. These studies have demonstrated that the efficacy of 3-month daily R combined with H (3HR) [113,115,129–132], 4-month daily R [113,120,133,134], 3-month weekly HP [135–138] and 1-month daily HP [139,140] is similar to 6–9 months of H monotherapy, with the major advantage of shortening treatment duration, which improves adherence. However, these regimens may be associated with drug-drug interactions, as rifamycins are potent inducers of cytochrome P450.

Table 3 lists the TPT regimens that have been shown to be effective and safe, together with the recommended doses by age group, with some comments on their indication [7,113,115,120,129–139].

Table 3.

TPT regimens shown to be effective. Doses and observations.

Regimen  Doses  Observations 
1 HP  H: 300mg/dayP: 600mg/dayFixed dose (regardless of body weight)  Not currently recommended for individuals <14 years oldCheck for drug interactionsa 
3 HP, weekly  H: 900mg/doseP: 900mg/dose  12 doses in total.Check for drug interactionsa 
3 HR  H:- >10 years old: 5mg/kg/day- <10 years old: 10mg/kg/day (range, 7–15mg)Maximum dose: 300mg/dayR:- >10 years old: 10mg/kg/day- <10 years old: 15mg/kg/day (range, 10–20mg)Maximum dose: 600mg/day  Check for drug interactionsa 
R:-> 10 years old: 10mg/kg/day- <10 years old: 15mg/kg/day (range, 10–20mg)Maximum dose: 600mg/day  Check for drug interactionsa 
6–9 H  H:-> 10 years old: 5mg/kg/day- <10 years old: 10mg/kg/day (range, 7–15mg)Maximum dose: 300mg/day   
6 Lfx  Levofloxacin:- >14 years old, based on body weight:- <50kg, 500mg/day;- ≥50kg, 750mg/day- ≤15 years old (range, approx. 15–20mg/kg/day):- 6–10kg: 150mg/day;- 10-15kg: 200g/day- 15–25kg: 250-300mg/day- 25–45kg: 500mg/day  Although there are no comparative data, moxifloxacin can be used if levofloxacin cannot be used. 
a

Rifamycins (rifampicin –R– and rifapentine –P–) are potent inducers of some enzymes of cytochrome P450, and accelerate the metabolism of some drugs, reducing their effect. These include some antiretrovirals, antiepileptics, antiarrhythmics, quinines, oral anticoagulants, antifungals, medical contraceptives, corticosteroids, ciclosporin, mycophenolate, quinolones, and other antimicrobials, antihyperglycemic agents, methadone and tricyclic antidepressants.

Based on its shorter duration, the 1HP regimen (daily dosing) would be the preferred option, particularly for people living with HIV. At present, however, it is not recommended for individuals under 14 years of age, as this age group has not been included in clinical trials, and therefore no scientific evidence is available, although it is reasonable to assume that it may also be effective [7,139,141,142]. The second option for TPT would be the 3HP regimen (weekly dosing), which requires only 12 doses in total, a feature that may facilitate adherence and supervised administration when deemed necessary. Although the WHO advises against the use of P during pregnancy due to the lack of evidence regarding its non-teratogenicity [7], some studies have reported a safety profile comparable to that of other TPT regimens [143], and its use in pregnant women may therefore be considered [144]. At the time of writing this report, P is not available in Spain or many other European countries, making the recommendation of 1HP or 3HP regimens unfeasible. Accordingly, the regimen of choice in Spain remains 3HR, with 6-9H reserved as an alternative in cases where R cannot be used due to toxicity, drug interactions, or mono- resistance to R. A study in Spain found the 3HR regimen to be the most cost-effective [145].

In individuals who have been in contact with TB patients with mono- or polyresistance to H, or intolerance to it, the treatment of choice for TPT is 4R [7,113,120,133,134]. In contacts of patients with multidrug-resistant TB (MDR-TB, resistant to at least H and R), 6 months of levofloxacin is recommended [7,146–148]. In this regard, there is no evidence for the efficacy of any TPT for contacts of patients with pre-extensively drug-resistant TB (MDR-TB that is also resistant to at least fluoroquinolones), and therefore these individuals should be managed on a case-by-case basis. Delamanid may be a good option given its mechanism of action [128].

The effectiveness of TPT in preventing TB disease is directly related to adherence. It is therefore essential to encourage each patient to fully adhere to the prescribed treatment throughout the diagnosis and treatment processes, especially among vulnerable populations [149]. Specialised TB units have achieved high rates of TPT adherence [150–153].

Preventive treatment-related adverse reactions and drug interactions

As with any other drug, TPT entails the risk of adverse effects, which may be serious; therefore, patients should be monitored through clinical follow-up and laboratory testing as deemed appropriate.

Serious adverse effects are uncommon, appearing in fewer than 5% of cases in most series [7,154,155], the most frequent being liver toxicity and dyspepsia. Factors such as older age, alcohol misuse, baseline liver disease, immunosuppressive medication, and malnutrition may increase the risk of toxicity [156–160]. The 6–9-month regimens with H are associated with slightly higher rates of hepatotoxicity than shorter treatments with R, P or H and R [134,135,139,161–163]. On the other hand, H and P containing regimens are associated with higher rates of cutaneous, flu-like or systemic adverse effects that may occasionally require discontinuation of treatment [161,164–166].

Although some researchers do not recommend systematic blood testing before TPT in patients at low risk of developing toxicity [7], in our setting, we recommend baseline laboratory testing, as well as monitoring during treatment, including at least liver function tests. Patients should be advised that in the event of signs of liver toxicity, they must contact a healthcare professional, who will assess the need for additional blood tests (including liver function tests). Warning signs and symptoms include severe dyspepsia, vomiting, fever, and jaundice of the skin or mucous membranes. Fig. 2 presents an action plan for suspected toxicity and/or the presence of compatible symptoms. In healthy children, adverse drug effects are rare; therefore, only a baseline test is recommended, with repeat testing indicated only if signs or symptoms suggestive of toxicity appear.

Fig. 2.

Algorithm for the management of suspected hepatotoxicity due to antituberculosis drugs. ULN: upper limit of normal value.

Other relatively common adverse effects are listed in Table 4.

Table 4.

Adverse effects of drugs used in tuberculosis preventive treatment.

Drug  Common  Uncommon 
Isoniazid  Elevated liver enzymes in asymptomatic patientsHepatitisPeripheral neuropathySkin rashSleepiness, lethargy  SeizuresJoint painLupus-like syndromeAcnePsychological disturbances Pancreatitis 
Rifampicin  DyspepsiaElevated liver enzymes in asymptomatic patientsHepatitisSkin rashThrombocytopeniaDiscolouration of urine and/or other bodily fluids  Acute kidney failureHaemolytic anaemiaFlu-like signs and symptomsHypoprothrombinaemia 
Rifapentine  DyspepsiaFlu-like signs and symptomsSkin rashHepatitisDiscolouration of urine and/or other bodily fluids  Low blood pressureLeucopeniaAnaemiaAnorexiaHypoprothrombinaemia 
Levofloxacin  DiarrhoeaDyspepsiaNausea, vomitingJoint pain  TendinopathyHepatitisMuscle painPeripheral neuropathyInsomniaProlonged QTc intervalAltered taste and smell 
Ref: Adapted from WHO operational handbook on tuberculosis. Module 1: prevention – tuberculosis preventive treatment, second edition. Geneva: World Health Organization; 2024 (https://iris.who.int/handle/10665/378535).

As a general rule, any medication associated with a previous serious adverse effect in a given patient should not be reintroduced; instead, an alternative treatment should be considered [7,167,168].

Conclusions

As stated in these recommendations, a comprehensive approach to TBI is a key strategy in the pathway towards TB elimination.

In Spain, testing for TBI should be performed before indicating TPT, with IGRA preferred over TST, although newer antigen-specific skin tests may play a relevant role in the near future. All individuals at high risk of developing TB should undergo diagnostic TBI testing as described in this document. Once TBI has been diagnosed and TB excluded, TPT should be initiated. At present, the preferred regimen is 3HR, although 1HP and weekly 3HP are expected to become options once rifapentine becomes available in our setting. Ensuring and promoting adherence to TPT is essential.

In addition to the correct and early diagnosis and treatment of TB and TBI, effective control of the disease requires thorough and appropriate management of cases, their contacts, and groups at high risk of developing TB disease. For this purpose, it is essential to notify all cases, ensure accurate contact tracing, and implement appropriate screening actions for both TB and TBI, within a multidisciplinary approach under the framework of a TB control programme.

Ethical statement

This work is part of the Agreement between the Directorate General of Public Health and Health Equity of the Ministry of Health, the Spanish Society of Pulmonology and Thoracic Surgery, and the Spanish Society of Infectious Diseases and Clinical Microbiology, for the development of activities within the framework of addressing tuberculosis (Resolution of November 25, 2024; Official State Gazette, December 21, 2024).

Declaration of generative AI and AI-assisted technologies in the manuscript preparation process

During the preparation of this work, the authors used ChatGPT in order to improve readability and language, and NapkinAI to enhance the Visual Abstract. After using these tools, the authors reviewed and edited the content as needed and take full responsibility for the content of the published article.

Funding

There is no private funding.

Conflicts of interest

The authors declare that they have no conflicts of interest that could be considered to influence, directly or indirectly, the content of the manuscript.

Acknowledgments

To the individuals who have contributed with comments and suggestions in the preparation of this document from the following organisations: Division of HIV, STI, Viral Hepatitis, and Tuberculosis Control of the Ministry of Health; Tuberculosis Working Group of the Spanish Society of Pediatric Infectious Diseases (SEIP); National Centre for Epidemiology; SEPAR Document Management Committee; members of the Mycobacterial Infections Study Group (GEIM) of SEIMC; Directorate-General of Public Health of the Galician Health Service; Public Health Agency of Catalonia (ASPCAT); Coordination Centre for Health Alerts and Emergencies (CCAES); and the. Public Health Centre of Castellón de la Plana.

References
[1]
P.K. Drain, K.L. Bajema, D. Dowdy, K. Dheda, K. Naidoo, S.G. Schumacher, et al.
Incipient and subclinical tuberculosis: a clinical review of early stages and progression of infection.
Clin Microbiol Rev, 31 (2018),
[2]
J.M. Achkar, E.R. Jenny-Avital.
Incipient and subclinical tuberculosis: defining early disease states in the context of host immune response.
J Infect Dis, 204 (2011), pp. S1179-S1186
[3]
A.K. Coussens, S.M.A. Zaidi, B.W. Allwood, P.K. Dewan, G. Gray, M. Kohli, et al.
Classification of early tuberculosis states to guide research for improved care and prevention: an international Delphi consensus exercise.
Lancet Respir Med, 12 (2024), pp. 484-498
[4]
A.L. Garcia-Basteiro, J. Ehrlich, M. Bonnet, M. Calnan, S.M. Graham, S. Hermans, et al.
A global tuberculosis dictionary: unified terms and definitions for the field of tuberculosis.
Lancet Glob Health, 12 (2024), pp. e737-e739
[5]
Report of the WHO consultation on asymptomatic tuberculosis, Geneva, Switzerland, 14–15 October 2024. Geneva: World Health Organization; 2025. Licence: CC BY-NC-SA 3.0 IGO.
[6]
J. Ehrlich, C. Suñer, G. Churchyard, F. Cobelens, M. Hatherill, S.C. Mendelsohn, et al.
Unveiling a hidden phenotype of early tuberculosis.
[7]
WHO., operational handbook on tuberculosis.
Module 1: prevention – tuberculosis preventive treatment second edition.
World Health Organization, (2024),
[8]
WHO.
Global Tuberculosis Report 2024.
Licence: CC BY-NC-SA 3.0 IGO ed, (2024),
[9]
R.M. Houben, P.J. Dodd.
The global burden of latent tuberculosis infection: a re-estimation using mathematical modelling.
[10]
C. Dye, C.J. Watt, D.M. Bleed, S.M. Hosseini, M.C. Raviglione.
Evolution of tuberculosis control and prospects for reducing tuberculosis incidence, prevalence, and deaths globally.
JAMA, 293 (2005), pp. 2767-2775
[11]
E. Ongoz, O. Aycicek, Y. Bülbül, F. Öztuna, T. Özlü.
Retrospective evaluation of active tuberculosis cases in terms of chemoprophylaxis indication: a cross-sectional study.
Medicine (Baltimore), 104 (2025),
[12]
Centro Nacional de Epidemiología.
Instituto de Salud Carlos III. Informe epidemiológico sobre la situación de la tuberculosis en España.
(2023),
[13]
J. Mahon, S. Beale, H. Holmes, M. Arber, V. Nikolayevskyy, R. Alagna, et al.
A systematic review of cost-utility analyses of screening methods in latent tuberculosis infection in high-risk populations.
BMC Pulm Med, 22 (2022), pp. 375
[14]
R.K. Gupta, C.J. Calderwood, A. Yavlinsky, M. Krutikov, M. Quartagno, M.C. Aichelburg, et al.
Discovery and validation of a personalized risk predictor for incident tuberculosis in low transmission settings.
Nat Med, 26 (2020), pp. 1941-1949
[15]
J. González-Martín, J.M. García-García, L. Anibarro, R. Vidal, J. Esteban, R. Blanquer, et al.
Consensus document on the diagnosis, treatment and prevention of tuberculosis.
Arch Bronconeumol, 46 (2010), pp. 255-274
[16]
M. Santin, J.M. García-García, D. Rigau, N. Altet, L. Anibarro, I. Casas, et al.
Executive summary of the guidelines for the use of interferon-γ release assays in the diagnosis of tuberculosis infection.
Enferm Infecc Microbiol Clin, 34 (2016), pp. 304-308
[17]
H. Getahun, A. Matteelli, R.E. Chaisson, M. Raviglione.
Latent Mycobacterium tuberculosis infection.
Engl N J Med, 372 (2015), pp. 2127-2135
[18]
S.A. Joosten, K.E. van Meijgaarden, S.M. Arend, C. Prins, F. Oftung, G.E. Korsvold, et al.
Mycobacterial growth inhibition is associated with trained innate immunity.
J Clin Invest, 128 (2018), pp. 1837-1851
[19]
J.L. Flynn, J. Chan.
Immune cell interactions in tuberculosis.
Cell, 185 (2022), pp. 4682-4702
[20]
I.M. Orme.
A new unifying theory of the pathogenesis of tuberculosis.
Tuberculosis (Edinb), 94 (2014), pp. 8-14
[21]
M.F. Goldberg, N.K. Saini, S.A. Porcelli.
Evasion of innate and adaptive immunity by Mycobacterium tuberculosis.
[22]
P.L. Lin, M. Rodgers, L. Smith, M. Bigbee, A. Myers, C. Bigbee, et al.
Quantitative comparison of active and latent tuberculosis in the cynomolgus macaque model.
Infect Immun, 77 (2009), pp. 4631-4642
[23]
J.M. Davis, L. Ramakrishnan.
The role of the granuloma in expansion and dissemination of early tuberculous infection.
[24]
M. Coppola, R. Villar-Hernández, K.E. van Meijgaarden, I. Latorre, B. Muriel Moreno, E. Garcia-Garcia, et al.
Cell-mediated immune responses to in vivo-expressed and stage-specific Mycobacterium tuberculosis antigens in latent and active tuberculosis across different age groups.
Front Immunol, 11 (2020), pp. 103
[25]
M.A. Behr, P.H. Edelstein, L. Ramakrishnan.
Revisiting the timetable of tuberculosis.
BMJ, 362 (2018), pp. k2738
[26]
S. Moreno Guillén, F.J. Rodríguez-Artalejo, J. Ruiz-Galiana, R. Cantón, P. De Lucas Ramos, A. García-Botella, et al.
Tuberculosis in Spain: an opinion paper.
Rev Esp Quimioter, 36 (2023), pp. 562-583
[27]
Grupo de trabajo Plan Prevención, Control de la Tuberculosis.
Plan para la prevención y control de la tuberculosis en España. Comisión de Salud Pública del Consejo Interterritorial del Sistema Nacionalde Salud. Ministerio de Sanidad.
Consumo y Bienestar Social, (2019),
[28]
A. Cohen, V.D. Mathiasen, T. Schön, C. Wejse.
The global prevalence of latent tuberculosis: a systematic review and meta-analysis.
[29]
C. Dye, S. Scheele, P. Dolin, V. Pathania, M.C. Raviglione.
Consensus statement. Global burden of tuberculosis: estimated incidence, prevalence, and mortality by country. WHO Global Surveillance and Monitoring Project.
JAMA, 282 (1999), pp. 677-686
[30]
C. Ding, M. Hu, W. Guo, W. Hu, X. Li, S. Wang, et al.
Prevalence trends of latent tuberculosis infection at the global, regional, and country levels from 1990–2019.
Int J Infect Dis, 122 (2022), pp. 46-62
[31]
K. Styblo.
The relationship between the risk of tuberculous infection and the risk of developing infectious tuberculosis.
Bull Int Union Tuberc Lung Dis, 60 (1985), pp. 117-119
[32]
F. van Leth, M.J. van der Werf, M.W. Borgdorff.
Prevalence of tuberculous infection and incidence of tuberculosis: a re-assessment of the Styblo rule.
Bull World Health Organ, 86 (2008), pp. 20-26
[33]
Registro regional de casos de tuberculosis de la comunidad de Madrid.
Informe del año 2023 SG Vigilancia en Salud Pública. Dirección General de Salud Pública.
Consejería de Sanidad-Comunidad de Madrid, (2023),
[34]
Informe da tuberculose en Galicia. Características dos casos de tuberculose de Galicia no ano 2023. Evolución do período 1996–2023. Available from: http://www.sergas.es/Saude-publica/Vixilancia-epidemioloxica-da-TB-en-Galicia [accessed 11.1.25].
[35]
J.P. Millet, A. Orcau, C. López-Muley, J. Molero, A. Artigas, I. Avellanés, et al.
La Tuberculosi a Barcelona. Informe 2022.
Agència de Salut Pública de Barcelona, (2024),
[36]
N. Serre-Delcor, B. Treviño-Maruri, M.T. Tórtola, M. Fernández-Quevedo, A. Soriano-Arandes, I. Oliveira-Souto, et al.
Sequential strategy for the LTBI screening of newly-arrived immigrants in vulnerable social situations.
Enferm Infecc Microbiol Clin (Engl Ed), 36 (2018), pp. 550-554
[37]
J. Salas-Coronas, M.T. Cabezas-Fernández, A.B. Lozano-Serrano, M.J. Soriano-Pérez, J. Vázquez-Villegas, J. Cuenca-Gómez.
Newly arrived African migrants to Spain: epidemiology and burden of disease.
Am J Trop Med Hyg, 98 (2018), pp. 319-325
[38]
S. Godoy, M. Alsedà, I. Parrón, J.P. Millet, J.A. Caylà, N. Follia, et al.
Exposure time to a tuberculosis index case as a marker of infection in immigrant populations.
[39]
J. García-Guerrero, A. Marco Mouriño, P. Sáiz de la Hoya Zamácola, E.J. Vera-Remartínez, G.de.P.d. Prisiones.
Multi-centre study of the prevalence of latent tuberculosis infection amongst inmates in Spanish prisons.
Rev Esp Sanid Penit, 12 (2010), pp. 79-85
[40]
M.E. López de Goicoechea-Saiz, F. Sternberg, J. Portilla-Sogorb.
Prevalence and associated risk factors of latent tuberculosis infection in a Spanish prison.
Rev Esp Sanid Penit, 20 (2018), pp. 4-10
[41]
R. Baca, M.J. Vivancos-Gallego.
Interferon-gamma release assay for the diagnosis of latent tuberculosis infection in the prison population with a positive tuberculin test: a descriptive study in a prison (Burgos Spain).
Rev Esp Sanid Penit, 25 (2023), pp. 104-111
[42]
J. Sol Vidiella, T. Catalán Gómez, C. Callau Casanova, M. Lejeune.
Latent tuberculosis infection in healthcare personnel at a primary level general hospital in Catalonia Spain.
Arch Prev Riesgos Labor, 17 (2014), pp. 97-101
[43]
I. Casas, M. Esteve, R. Guerola, I. García-Olivé, J. Roldán-Merino, C. Martinez-Rivera, et al.
Incidence of tuberculosis infection among healthcare workers: risk factors and 20-year evolution.
Respir Med, 107 (2013), pp. 601-607
[44]
A. Arnedo-Pena, J.V. Juan-Cerdán, M. Romeu-Garcia, S. Sorribes-Segura, M. Tirado-Balaguer, M. Gil-Fortuño, et al.
Vitamin D status and latent tuberculosis infection: conversion in nursing homes, Spain.
Int J Tuberc Lung Dis, 24 (2020), pp. 278-286
[45]
T.M. Perez-Porcuna, A. Noguera-Julian, M.T. Riera-Bosch, E. Macià-Rieradevall, J. Santos-Santiago, M. Rifà Pujol, et al.
Tuberculosis among children visiting friends & relatives.
J Travel Med, 31 (2024),
[46]
N. Busquets-Pérez, A. Ponce, V. Ortiz-Santamaria, J. de Agustín de Oro, Y.L. Hernández-Rico, I. Vidal, et al.
How many patients with rheumatic diseases and TNF inhibitors treatment have latent tuberculosis?.
Reumatol Clin, 13 (2017), pp. 282-286
[47]
J. Calzada-Hernández, J. Anton, J. Martín de Carpi, B. López-Montesinos, I. Calvo, E. Donat, et al.
Dual latent tuberculosis screening with tuberculin skin tests and QuantiFERON-TB assays before TNF-α inhibitor initiation in children in Spain.
Eur J Pediatr, 182 (2023), pp. 307-317
[48]
S. Quirós, D. de la Rosa, A. Uranga, R. Madero, R. Amaro, N. Bruguera, et al.
Screening for latent tuberculosis infection in patients who are candidate for biological therapies in Spain? A multidisciplinary survey.
Arch Bronconeumol (Engl Ed), 54 (2018), pp. 510-517
[49]
L.S. Li, L. Yang, L. Zhuang, Z.Y. Ye, W.G. Zhao, W.P. Gong.
From immunology to artificial intelligence: revolutionizing latent tuberculosis infection diagnosis with machine learning.
Mil Med Res, 10 (2023), pp. 58
[50]
L. Muñoz, L. Gonzalez, L. Soldevila, J. Dorca, F. Alcaide, M. Santin.
QuantiFERON®-TB Gold In-Tube for contact screening in BCG-vaccinated adults: a longitudinal cohort study.
[51]
QuantiFERON®-TB Gold Plus (QFT®-Plus) ELISA package insert.
QIAGEN, (2019),
[52]
E. Ortiz-Brizuela, L. Apriani, T. Mukherjee, S. Lachapelle-Chisholm, M. Miedy, Z. Lan, et al.
Assessing the diagnostic performance of new commercial interferon-γ release assays for Mycobacterium tuberculosis infection: a systematic review and meta-analysis.
Clin Infect Dis, 76 (2023), pp. 1989-1999
[53]
M. Fernández-Huerta, C. Moreto, N. Vila-Olmo, E.I. García de Cara, M.C. Basaez, M. Santín, et al.
Evaluation of the fully automated chemiluminescence analyzer liaison XL for the performance of the quantiFERON-TB gold plus assay in an area with a low incidence of tuberculosis.
J Clin Microbiol, 59 (2021),
[54]
T-SPOT.TB package insert.
Oxford Immunotec, (2017),
[55]
Y. Kobashi.
Current status and future landscape of diagnosing tuberculosis infection.
Respir Investig, 61 (2023), pp. 563-578
[56]
G. Zhou, Q. Luo, S. Luo, J. He, N. Chen, Y. Zhang, et al.
Positive rates of interferon-γ release assay and tuberculin skin test in detection of latent tuberculosis infection: a systematic review and meta-analysis of 200,000 head-to-head comparative tests.
[57]
S.H. Wong, Q. Gao, K.K. Tsoi, W.K. Wu, L.S. Tam, N. Lee, et al.
Effect of immunosuppressive therapy on interferon γ release assay for latent tuberculosis screening in patients with autoimmune diseases: a systematic review and meta-analysis.
[58]
Z. Ozsoy, A. Ozdemir, M. Ekici, E. Bilgin, L. Kılıc, S. Kiraz, et al.
Do the drug doses of conventional synthetic DMARDs used for the treatment of biologic/targeted-synthetic DMARDs naive rheumatoid arthritis patients affect QuantiFERON-TB Gold Plus test results?.
Rheumatol Int, 43 (2023), pp. 1445-1451
[59]
R.M. Jasmer, P. Nahid, P.C. Hopewell.
Clinical practice Latent tuberculosis infection.
N Engl J Med, 347 (2002), pp. 1860-1866
[60]
European Centre for Disease Prevention and Control.
Handbook on tuberculosis laboratory diagnostic methods in the European Union – Updated 2022.
ECDC, (2023),
[61]
M. Ruhwald, H. Aggerbeck, R.V. Gallardo, S.T. Hoff, J.I. Villate, B. Borregaard, et al.
Safety and efficacy of the C-Tb skin test to diagnose Mycobacterium tuberculosis infection, compared with an interferon γ release assay and the tuberculin skin test: a phase 3, double-blind, randomised, controlled trial.
Lancet Respir Med, 5 (2017), pp. 259-268
[62]
Y. He, L. Shen, J. Du, X. Cao, B. Zhang, D. Wang, et al.
Agreement between Mycobacterium tuberculosis antigen-based skin test and interferon-gamma release assay in elderly individuals aged ≥65 years in China.
Clin Microbiol Infect, 31 (2025),
[63]
L. Peng, W. Ma, L. Zhong, J. Yang, H. Wu, L. Zhu, et al.
Diagnostic accuracy of Mycobacterium tuberculosis antigen-based skin tests (TBSTs) for tuberculosis infection compared with TST and IGRA: a network meta-analysis.
[64]
J. Chen, L. Zhao, X. Zhou, Y. Mo, L. Kong, F. Ma, et al.
Safety and diagnostic performance of recombinant fusion protein ESAT6-CPF10 skin test in a large population: a phase IV clinical trial.
[65]
WHO operational handbook on tuberculosis.
Module 3: diagnosis Tests for tuberculosis infection.
World Health Organization, (2022),
[66]
O. Estévez, L. Anibarro, E. Garet, Á. Pallares, L. Barcia, L. Calviño, et al.
An RNA-seq based machine learning approach identifies latent tuberculosis patients with an active tuberculosis profile.
Front Immunol, 11 (2020), pp. 1470
[67]
B.M.F. Nogueira, F. Rangel, A.M.S. Andrade, E.A. Daniel, M.C. Figueiredo, C. Staats, et al.
Predictive markers of incident tuberculosis in close contacts in Brazil and India.
[68]
S.C. Mendelsohn, B.B. Andrade, S.K. Mbandi, A.M.S. Andrade, V.M. Muwanga, M.C. Figueiredo, et al.
Transcriptomic signatures of progression to tuberculosis disease among close contacts in Brazil.
J Infect Dis, 230 (2024), pp. e1355-e1365
[69]
T. Ayers, A.N. Hill, J. Raykin, S. Mohanty, R.W. Belknap, R. Brostrom, et al.
Comparison of tuberculin skin testing and interferon-γ release assays in predicting tuberculosis disease.
[70]
Y. Zhang, G. Zhou, W. Shi, M. Hu, D. Kong, R. Long, et al.
Comparing the diagnostic performance of QuantiFERON-TB Gold Plus with QFT-GIT T-SPOT.TB and TST: a systematic review and meta-analysis.
BMC Infect Dis, 23 (2023), pp. 40
[71]
A. Fernández-Blázquez, P. Argüelles Menéndez, C. Sabater-Cabrera, J.M. García-García, V. Asensi Álvarez, J.J. Palacios Gutiérrez, et al.
Diagnosis of tuberculous infection in immunosuppressed patients and/or candidates for biologics using a combination of 2 IGRA tests: T-SPOT.TB/QuantiFERON TB gold in-tube vs T-SPOT.TB/QuantiFERON TB gold plus.
Arch Bronconeumol, 58 (2022), pp. 305-310
[72]
L. Muñoz, M. Santin, F. Alcaide, M.J. Ruiz-Serrano, P. Gijon, E. Bermudez, et al.
QuantiFERON-TB gold in-tube as a confirmatory test for tuberculin skin test in tuberculosis contact tracing: a noninferiority clinical trial.
Clin Infect Dis, 66 (2018), pp. 396-403
[73]
C.K. Chan, C.C. Leung, S.S. Huang, S.N.M. Lee, L.B. Tai.
Combining tuberculin skin test with follow-on interferon gamma release assay markedly improves screening of household contacts.
[74]
G. Zhou, Q. Luo, S. Luo, Z. Teng, Z. Ji, J. Yang, et al.
Interferon-γ release assays or tuberculin skin test for detection and management of latent tuberculosis infection: a systematic review and meta-analysis.
Lancet Infect Dis, 20 (2020), pp. 1457-1469
[75]
C.H. Park, J.H. Park, Y.S. Jung.
Impact of immunosuppressive therapy on the performance of latent tuberculosis screening tests in patients with inflammatory bowel disease: a systematic review and meta-analysis.
J Pers Med, 12 (2022), pp. 507
[76]
G. Woodfield, B. Passey-Heaton, A. Chakrabarti, A. Pozniak, M.R. Loebinger.
An evaluation of the use of a negative interferon-γ release assay for tuberculosis screening before TNF antagonist therapy.
Eur Respir J, 44 (2014), pp. 1369-1372
[77]
L. Anibarro, M. Trigo, C. Villaverde, A. Pena, S. Cortizo, D. Sande, et al.
Interferon-gamma release assays in tuberculosis contacts: is there a window period?.
Eur Respir J, 37 (2011), pp. 215-217
[78]
W.M. Kyaw, J.Y. Tay, L.K. Lim, D.H.L. Ng.
Time interval for QuantiFERON-TB Gold Plus conversion after last exposure with tuberculosis.
[79]
L. Muñoz, S. Casas, X. Juanola, X. Bordas, C. Martinez, M. Santin, et al.
Prevention of anti-tumor necrosis factor-associated tuberculosis: a 10-year longitudinal cohort study.
Clin Infect Dis, 60 (2015), pp. 349-356
[80]
P. Lu, Q. Liu, Y. Zhou, L. Martinez, W. Kong, X. Ding, et al.
Predictors of discordant tuberculin skin test and QuantiFERON-TB gold in-tube results in Eastern China: a population-based, cohort study.
Clin Infect Dis, 72 (2021), pp. 2006-2015
[81]
Y. Hamada, R.K. Gupta, M. Quartagno, A. Izzard, C. Acuna-Villaorduna, N. Altet, et al.
Predictive performance of interferon-gamma release assays and the tuberculin skin test for incident tuberculosis: an individual participant data meta-analysis.
[82]
F. Baquero-Artigao, T. Del Rosal, L. Falcón-Neyra, L. Ferreras-Antolín, D. Gómez-Pastrana, A. Hernanz-Lobo, et al.
Update on the diagnosis and treatment of tuberculosis.
An Pediatr (Engl Ed), 98 (2023), pp. 460-469
[83]
S. Pérez-Recio, M.D. Grijota-Camino, J. Guardiola, X. Juanola, J. Notario, X. Solanich, et al.
Prevention of tuberculosis in patients treated with biological therapies: twenty years’ experience in a specialised tuberculosis clinic in a low-prevalence country.
Clin Infect Dis, (2025),
[84]
Guidelines on the management of latent tuberculosis infection (WHO/HTM/TB/2015.01).
World Health Organization, (2015),
[85]
J.M. Trauer, N. Moyo, E.L. Tay, K. Dale, R. Ragonnet, E.S. McBryde, et al.
Risk of active tuberculosis in the five years following infection. 15%?.
Chest, 149 (2016), pp. 516-525
[86]
R. Sloot, M.F. Schim van der Loeff, P.M. Kouw, M.W. Borgdorff.
Risk of tuberculosis after recent exposure. A 10-year follow-up study of contacts in Amsterdam.
Am J Respir Crit Care Med, 190 (2014), pp. 1044-1052
[87]
L. Anibarro, M. Trigo, C. Villaverde, A. Pena, A. González-Fernández.
Tuberculin skin test and interferon-γ release assay show better correlation after the tuberculin ‘window period’ in tuberculosis contacts.
Scand J Infect Dis, 43 (2011), pp. 424-429
[88]
J.W. Kim, J. Nazareth, J. Lee, H. Patel, G. Woltmann, R. Verma, et al.
Interferon-gamma release assay conversion after Mycobacterium tuberculosis exposure specifically associates with greater risk of progression to tuberculosis: a prospective cohort study in Leicester, UK.
Int J Infect Dis, 141 (2024),
[89]
J.W. Ai, Q.L. Ruan, Q.H. Liu, W.H. Zhang.
Updates on the risk factors for latent tuberculosis reactivation and their managements.
Emerg Microbes Infect, 5 (2016),
[90]
H. Getahun, A. Matteelli, I. Abubakar, M.A. Aziz, A. Baddeley, D. Barreira, et al.
Management of latent Mycobacterium tuberculosis infection: WHO guidelines for low tuberculosis burden countries.
Eur Respir J, 46 (2015), pp. 1563-1576
[91]
G.J. Churchyard, K.L. Fielding, A.D. Grant.
A trial of mass isoniazid preventive therapy for tuberculosis control.
N Engl J Med, 370 (2014), pp. 1662-1663
[92]
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, 60 (1982), pp. 555-564
[93]
J. Solsona Peiro, M.L. de Souza Galvao, M.N. Altet Gomez.
Inactive fibrotic lesions versus pulmonary tuberculosis with negative bacteriology.
Arch Bronconeumol, 50 (2014), pp. 484-489
[94]
J. Komukai, K. Matsumoto, W. Fukushima, S. Kudoh.
Pulmonary tuberculosis incidence among interferon-gamma release assay-positive individuals with latent tuberculosis infection and fibrotic lesions in a vulnerable urban population in Osaka City, Japan, 2015–2021.
Jpn J Infect Dis, 77 (2024), pp. 21-24
[95]
L. Shen, Y. Zhang, H. Xin, X. Cao, J. Du, Y. Di, et al.
Association of inactive pulmonary tuberculosis lesions with the risk of active disease development in the elderly: a population-based retrospective study.
ERJ Open Res, 11 (2025), pp. 01231-2024
[96]
A. Picchianti-Diamanti, A. Aiello, C. De Lorenzo, G.B. Migliori, D. Goletti.
Management of tuberculosis risk, screening and preventive therapy in patients with chronic autoimmune arthritis undergoing biotechnological and targeted immunosuppressive agents.
[97]
D.K. Jha, R. Kakadiya, A. Sharma, S. Naidu, D. De, V. Sharma.
Assessment and management for latent tuberculosis before advanced therapies for immune-mediated inflammatory diseases: a comprehensive review.
[98]
H. Li, C.B.E. Chee, T. Geng, A. Pan, W.P. Koh.
Joint associations of multiple lifestyle factors with risk of active tuberculosis in the population: the Singapore Chinese Health Study.
Clin Infect Dis, 75 (2022), pp. 213-220
[99]
Y. Li, S.M. Marks, G.R. Beeler Asay, C.A. Winston, D. Pepin, S. McClure, et al.
Effectiveness and cost-effectiveness of expanded targeted testing and treatment of latent tuberculosis infection among the medicare population in 2022.
[100]
A. Sanchez-Montalva, J.A. Caminero, M.R. Guna, T.R. Sanz, R. Rabuñal, J.P. Millet, et al.
Executive Summary: Clinical Practice Guidelines on the Management of Resistant Tuberculosis of the Spanish Society of Pulmonology and Thoracic Surgery (SEPAR) and the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC).
Arch Bronconeumol, 60 (2024), pp. 759-767
[101]
C.G. Erkens, M. Kamphorst, I. Abubakar, G.H. Bothamley, D. Chemtob, W. Haas, et al.
Tuberculosis contact investigation in low prevalence countries: a European consensus.
Eur Respir J, 36 (2010), pp. 925-949
[102]
M. D’Ottavi, P. Godfrey-Faussett, C.S. Merle, M.T. Sofonea, D. Laureillard, P. Vickerman, et al.
Tuberculosis and people who use drugs: why focus on this overlooked population is important and why adapted interventions are necessary.
Lancet Glob Health, 13 (2025), pp. e593-e598
[103]
S. Verver, R.M. Warren, N. Beyers, M. Richardson, G.D. van der Spuy, M.W. Borgdorff, et al.
Rate of reinfection tuberculosis after successful treatment is higher than rate of new tuberculosis.
Am J Respir Crit Care Med, 171 (2005), pp. 1430-1435
[104]
F.M. Marx, A.C. Hesseling, N. Martinson, G. Theron, T. Cohen.
National survey in South Africa reveals high tuberculosis prevalence among previously treated people.
Lancet Infect Dis, 22 (2022), pp. 1273
[105]
WHO consolidated guidelines on tuberculosis.
Module 2: screening – systematic screening for tuberculosis disease.
World Health Organization, (2021),
[106]
R. Crowder, B. Thangakunam, A. Andama, D.J. Christopher, V. Dalay, W. Nwamba, et al.
Diagnostic accuracy of TB screening tests in a prospective multinational cohort: Chest-X-ray with computer-aided detection Xpert TB host response, and C-reactive protein.
Clin Infect Dis, (2024),
[107]
S.H. Ferebee, F.W. Mount.
Tuberculosis morbidity in a controlled trial of the prophylactic use of isoniazid among household contacts.
Am Rev Respir Dis, 85 (1962), pp. 490-510
[108]
G.W. Comstock, S.H. Ferebee, L.M. Hammes.
A controlled trial of community-wide isoniazid prophylaxis in Alaska.
Am Rev Respir Dis, 95 (1967), pp. 935-943
[109]
S.H. Ferebee.
Controlled chemoprophylaxis trials in tuberculosis. A general review.
Bibl Tuberc, 26 (1970), pp. 28-106
[110]
L.B. Edwards, B. Doster, V.T. Livesay, S.H. Ferebee.
Risk of tuberculosis among persons with “not active – not treated” lesions.
Bull Int Union Tuberc, 47 (1972), pp. 151-156
[111]
A. Falk, G.F. Fuchs.
Prophylaxis with isoniazid in inactive tuberculosis. A Veterans administration cooperative study XII.
Chest, 73 (1978), pp. 44-48
[112]
G.W. Comstock, C. Baum, D.E. Snider Jr..
Isoniazid prophylaxis among Alaskan Eskimos: a final report of the bethel isoniazid studies.
Am Rev Respir Dis, 119 (1979), pp. 827-830
[113]
A double-blind placebo-controlled clinical trial of three antituberculosis chemoprophylaxis regimens in patients with silicosis in Hong Kong.
Hong Kong Chest Service/Tuberculosis Research Centre Madras/British Medical Research Council.
Am Rev Respir Dis, 145 (1992), pp. 36-41
[114]
J.W. Pape, S.S. Jean, J.L. Ho, A. Hafner, W.D. Johnson Jr..
Effect of isoniazid prophylaxis on incidence of active tuberculosis and progression of HIV infection.
[115]
C.C. Whalen, J.L. Johnson, A. Okwera, D.L. Hom, R. Huebner, P. Mugyenyi, 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, 337 (1997), pp. 801-808
[116]
F.M. Gordin, J.P. Matts, C. Miller, L.S. Brown, R. Hafner, S.L. John, et al.
A controlled trial of isoniazid in persons with anergy and human immunodeficiency virus infection who are at high risk for tuberculosis Terry Beirn Community Programs for Clinical Research on AIDS.
N Engl J Med, 337 (1997), pp. 315-320
[117]
M.P. Hawken, H.K. Meme, L.C. Elliott, J.M. Chakaya, J.S. Morris, W.A. Githui, et al.
Isoniazid preventive therapy for tuberculosis in HIV-1-infected adults: results of a randomized controlled trial.
[118]
G.W. Comstock, S.H. Ferebee.
How much isoniazid is needed for prophylaxis?.
Am Rev Respir Dis, 101 (1970), pp. 780-782
[119]
G.W. Comstock.
How much isoniazid is needed for prevention of tuberculosis among immunocompetent adults?.
Int J Tuberc Lung Dis, 3 (1999), pp. 847-850
[120]
D. Zenner, N. Beer, R.J. Harris, M.C. Lipman, H.R. Stagg, M.J. van der Werf.
Treatment of latent tuberculosis infection: an updated network meta-analysis.
Ann Intern Med, 167 (2017), pp. 248-255
[121]
Preventive therapy of tuberculous infection.
Am Rev Respir Dis, 110 (1974), pp. 371-374
[122]
American Thoracic Society. Medical Section of the American Lung Association: treatment of tuberculosis, tuberculosis infection in adults, children.
Am Rev Respir Dis, 134 (1986), pp. 355-363
[123]
J.B. Bass Jr., L.S. Farer, P.C. Hopewell, R. O’Brien, R.F. Jacobs, F. Ruben, et al.
Treatment of tuberculosis and tuberculosis infection in adults and children American Thoracic Society and The Centers for Disease Control and Prevention.
Am J Respir Crit Care Med, 149 (1994), pp. 1359-1374
[124]
Control, prevention of tuberculosis in the United Kingdom: Code of Practice 1994.
Joint tuberculosis committee of the British Thoracic Society.
Thorax, 49 (1994), pp. 1193-1200
[125]
Targeted tuberculin testing, treatment of latent tuberculosis infection.
Am J Respir Crit Care Med, 161 (2000), pp. S221-S247
[126]
J.A. Caminero, A. Scardigli.
Classification of antituberculosis drugs: a new proposal based on the most recent evidence.
Eur Respir J, 2015 (2015), pp. 887-893
[127]
J.A. Caminero, J.M. García-García, J.A. Caylà, F.J. García-Pérez, J.J. Palacios, J. Ruiz-Manzano.
Update of SEPAR guideline “Diagnosis and Treatment of Drug-Resistant Tuberculosis”.
Arch Bronconeumol (Engl Ed), 56 (2020), pp. 514-521
[128]
J.A. Caminero, R. Singla, A. Scardigli, A. Amitesh, G. Pérez-Mendoza, A. Mendoza-Ticona.
The basis of tuberculosis treatment: fundamental concepts before treating a patient.
The challenge of tuberculosis in the 21st century (ERS Monograph), pp. 104-116 http://dx.doi.org/10.1183/2312508X.10024522
[129]
E. Martínez Alfaro, J. Solera, E. Serna, D. Cuenca, M.L. Castillejos, A. Espinosa, et al.
Compliance, tolerance and effectiveness of a short chemoprophylaxis regimen for the treatment of tuberculosis.
Med Clin (Barc), 111 (1998), pp. 401-404
[130]
E.M. Martínez Alfaro, F. Cuadra, J. Solera, M.A. Maciá, P. Geijo, P.A. Sánchez Martínez, et al.
Evaluation of 2 tuberculosis chemoprophylaxis regimens in patients infected with human immunodeficiency virus. The GECMEI Group.
Med Clin (Barc), 115 (2000), pp. 161-165
[131]
A. Rivero, L. López-Cortés, R. Castillo, F. Lozano, M.A. García, F. Díez, et al.
Randomized trial of three regimens to prevent tuberculosis in HIV-infected patients with anergy.
Enferm Infecc Microbiol Clin, 21 (2003), pp. 287-292
[132]
N.P. Spyridis, P.G. Spyridis, A. Gelesme, V. Sypsa, M. Valianatou, F. Metsou, et al.
The effectiveness of a 9-month regimen of isoniazid alone versus 3- and 4-month regimens of isoniazid plus rifampin for treatment of latent tuberculosis infection in children: results of an 11-year randomized study.
Clin Infect Dis, 45 (2007), pp. 715-722
[133]
A. Aspler, R. Long, A. Trajman, M.J. Dion, K. Khan, K. Schwartzman, et al.
Impact of treatment completion, intolerance and adverse events on health system costs in a randomised trial of 4 months rifampin or 9 months isoniazid for latent TB.
Thorax, 65 (2010), pp. 582-587
[134]
D. Menzies, M. Adjobimey, R. Ruslami, A. Trajman, O. Sow, H. Kim, et al.
Four months of rifampin or nine months of isoniazid for latent tuberculosis in adults.
N Engl J Med, 379 (2018), pp. 440-453
[135]
T.R. Sterling, M.E. Villarino, A.S. Borisov, N. Shang, F. Gordin, E. Bliven-Sizemore, et al.
Three months of rifapentine and isoniazid for latent tuberculosis infection.
N Engl J Med, 365 (2011), pp. 2155-2166
[136]
T.R. Sterling, N.A. Scott, J.M. Miro, G. Calvet, A. La Rosa, R. Infante, et al.
Three months of weekly rifapentine and isoniazid for treatment of Mycobacterium tuberculosis infection in HIV-coinfected persons.
[137]
M.E. Villarino, N.A. Scott, S.E. Weis, M. Weiner, M.B. Conde, B. Jones, et al.
Treatment for preventing tuberculosis in children and adolescents: a randomized clinical trial of a 3-month, 12-dose regimen of a combination of rifapentine and isoniazid.
JAMA Pediatr, 169 (2015), pp. 247-255
[138]
G.J. Njie, S.B. Morris, R.Y. Woodruff, R.N. Moro, A.A. Vernon, A.S. Borisov.
Isoniazid-rifapentine for latent tuberculosis infection: a systematic review and meta-analysis.
Am J Prev Med, 55 (2018), pp. 244-252
[139]
S. Swindells, R. Ramchandani, A. Gupta, C.A. Benson, J. Leon-Cruz, N. Mwelase, et al.
One month of rifapentine plus isoniazid to prevent HIV-related tuberculosis.
N Engl J Med, 380 (2019), pp. 1001-1011
[140]
H.L. Huang, M.R. Lee, C.H. Lee, M.H. Cheng, P.L. Lu, C.C. Sheu, et al.
One-month daily and three-month weekly rifapentine plus isoniazid are comparable in completion rate and safety for latent tuberculosis infection in non-HIV Population: a randomized controlled trial.
Clin Microbiol Infect, 30 (2024), pp. 1410-1417
[141]
W. Liu, N. Xu, W. Li, W.Y. Mak, T. He, H. Qin, et al.
Pharmacokinetics and safety of rifapentine in children: dosing for latent tuberculosis infection.
J Antimicrob Chemother, 80 (2025), pp. 1022-1030
[142]
A. Noguera-Julián, C. Latre, M. Ruiz, C. García-Rebollo, M. Peralta, C. Fortuny.
Rifapentine-based treatment of tuberculosis infection in Spanish adolescents.
Enferm Infecc Microbiol Clin, (2025),
[143]
R.N. Moro, N.A. Scott, A. Vernon, N.K. Tepper, S.V. Goldberg, K. Schwartzman, et al.
Exposure to latent tuberculosis treatment during pregnancy. The PREVENT TB and the iAdhere trials.
Ann Am Thorac Soc, 15 (2018), pp. 570-580
[144]
V. Chihota, Z. Waggie, V. Cardenas, N. Martinson, G. Yimer, A.L. Garcia-Basteiro, et al.
OA07-638-19. Safety of short-course weekly rifapentine and isoniazid (3HP) for TB preventive treatment during pregnancy.
Int J Tuberc Lung Dis, 25 (2021), pp. S61-S62
[145]
J.A. Gullón-Blanco, T. Rodrigo-Sanz, E. Tabernero-Huguet, J. Sabría-Mestres, L. Anibarro, M. Villanueva-Montes, et al.
Tuberculosis contacts tracing in Spain: cost analysis.
Arch Bronconeumol, 58 (2022), pp. 448-450
[146]
G.J. Fox, N.V. Nhung, N. Cam Binh, N.B. Hoa, F.L. Garden, A. Benedetti, et al.
Levofloxacin for the prevention of multidrug-resistant tuberculosis in Vietnam.
N Engl J Med, 391 (2024), pp. 2304-2314
[147]
A.C. Hesseling, S.E. Purchase, N.A. Martinson, L. Fairlie, H.S. Schaaf, J. Brigden, et al.
Levofloxacin preventive treatment in children exposed to MDR tuberculosis.
N Engl J Med, 391 (2024), pp. 2315-2326
[148]
T. Duong, J. Brigden, H. Simon Schaaf, F. Garden, B.J. Marais, T. Anh Nguyen, et al.
A meta-analysis of levofloxacin for contacts of multidrug-resistant tuberculosis.
[149]
Tuberculosis among populations at high risk and people in vulnerable situations: policy brief.
World Health Organization, (2025),
[150]
L. Anibarro, S. Casas, J. Paz-Esquete, L. Gonzalez, A. Pena, M.R. Guerra, et al.
Treatment completion in latent tuberculosis infection at specialist tuberculosis units in Spain.
Int J Tuberc Lung Dis, 14 (2010), pp. 701-707
[151]
M.D. Grijota-Camino, S. Pérez-Recio, C. Trapero, M.J. Luque, M. Casellas, N. Sabé-Fernández, et al.
Identifying gaps in the cascade of care for latent TB infection in a low-incidence setting.
Int J Tuberc Lung Dis, 27 (2023), pp. 315-321
[152]
N. Ortiz Laza, I. Lopez Aranaga, J. Toral Andres, B. Toja Uriarte, B. Santos Zorrozua, L. Altube Urrengoechea, et al.
Latent tuberculosis infection treatment completion in Biscay: differences between regimens and monitoring approaches.
Front Med (Lausanne), 10 (2023),
[153]
M.A. Jiménez-Fuentes, C.M. Augé, M.N. Gómez, J.S. Peiró, M.L. de Souza Galvao, J. Maldonado, et al.
Screening for active tuberculosis in high-risk groups.
Int J Tuberc Lung Dis, 18 (2014), pp. 1459-1465
[154]
H.R. Stagg, D. Zenner, R.J. Harris, L. Muñoz, M.C. Lipman, I. Abubakar.
Treatment of latent tuberculosis infection: a network meta-analysis.
Ann Intern Med, 161 (2014), pp. 419-428
[155]
C. Pease, B. Hutton, F. Yazdi, D. Wolfe, C. Hamel, P. Barbeau, et al.
A systematic review of adverse events of rifapentine and isoniazid compared to other treatments for latent tuberculosis infection.
Pharmacoepidemiol Drug Saf, 27 (2018), pp. 557-566
[156]
Y.S. Jung, S.Y. Jung, J.E. Lee, K. Lee, J.C. Choi.
Adverse drug reactions following treatment of latent tuberculosis infection: a linked national tuberculosis surveillance with claims database.
Korean J Intern Med, 39 (2024), pp. 979-988
[157]
J.D. Puyana Ortiz, A.C. Garcés Rodríguez, M.L. Aznar, J. Espinosa Pereiro, A. Sánchez-Montalvá, J. Martínez-Campreciós, et al.
Adherence and toxicity during the treatment of latent tuberculous infection in a referral center in Spain.
Trop Med Infect Dis, 8 (2023),
[158]
C. Chung, Y.J. Kim, K.W. Jo, T.S. Shim.
Safety of latent tuberculosis infection treatment in older patients with immune-mediated inflammatory diseases.
Clin Rheumatol, 41 (2022), pp. 889-897
[159]
J.R. Campbell, A. Trajman, V.J. Cook, J.C. Johnston, M. Adjobimey, R. Ruslami, 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, 20 (2020), pp. 318-329
[160]
J.A. Hahn, C. Ngabirano, R. Fatch, N.I. Emenyonu, D.M. Cheng, J. Adong, et al.
Safety and tolerability of isoniazid preventive therapy for tuberculosis for persons with HIV with and without alcohol use.
AIDS, 37 (2023), pp. 1535-1543
[161]
L. Melnychuk, S. Perlman-Arrow, M. Lisboa Bastos, D. Menzies.
A systematic review and meta-analysis of tuberculous preventative therapy adverse events.
Clin Infect Dis, 77 (2023), pp. 287-294
[162]
L.A. Ronald, J.M. FitzGerald, G. Bartlett-Esquilant, K. Schwartzman, A. Benedetti, J.F. Boivin, et al.
Treatment with isoniazid or rifampin for latent tuberculosis infection: population-based study of hepatotoxicity, completion and costs.
[163]
E.E. Bliven-Sizemore, T.R. Sterling, N. Shang, D. Benator, K. Schwartzman, R. Reves, et al.
Three months of weekly rifapentine plus isoniazid is less hepatotoxic than nine months of daily isoniazid for LTBI.
Int J Tuberc Lung Dis, 19 (2015), pp. 1039-1044
[164]
H.L. Huang, M.R. Lee, C.H. Lee, J.Y. Wang.
‘One-month daily and three-month weekly rifapentine plus isoniazid are comparable in completion rate and safety for latent tuberculosis infection in non-HIV Population: a randomized controlled trial’ – author's response.
Clin Microbiol Infect, 30 (2024), pp. 1482-1483
[165]
J.L. Kadota, A. Musinguzi, H.E. Aschmann, L. Akello, F. Welishe, J. Nakimuli, et al.
Adverse events reported during weekly isoniazid-rifapentine (3HP) tuberculosis preventive treatment among people with human immunodeficiency virus in Uganda.
Open Forum Infect Dis, 11 (2024),
[166]
A.T. Cruz, L.H. Cameron, J.R. Starke.
Completion safety and tolerability of once-weekly isoniazid and rifapentine for tuberculosis infection by children and adolescents.
[167]
J.J. Saukkonen, D.L. Cohn, R.M. Jasmer, S. Schenker, J.A. Jereb, C.M. Nolan, et al.
An official ATS statement: hepatotoxicity of antituberculosis therapy.
Am J Respir Crit Care Med, 174 (2006), pp. 935-952
[168]
K.P. Singh, A.C.C. Carvalho, R.D. Centis, L. Ambrosio, G.B. Migliori, S.G. Mpagama, et al.
Clinical standards for the management of adverse effects during treatment for TB.
Int J Tuberc Lung Dis, 27 (2023), pp. 506-519

This paper was jointly developed by Archivos de Bronconeumología, Enfermedades Infecciosas y Microbiología Clínica, Enfermedades Infecciosas y Microbiología Clínica (English ed.) and jointly published by Elsevier España S.L.U. The articles are identical except for minor stylistic and spelling differences in keeping with each journal's style. Either citation can be used when citing this article.

Joint first authors.

Co-senior.

Copyright © 2025. Sociedad Española de Neumología y Cirugía Torácica (SEPAR), Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica, Sociedad Española de Neumología y Cirugía Torácica
Archivos de Bronconeumología
Article options
Tools