51
|
Ross JM, Badje A, Rangaka MX, Walker AS, Shapiro AE, Thomas KK, Anglaret X, Eholie S, Gabillard D, Boulle A, Maartens G, Wilkinson RJ, Ford N, Golub JE, Williams BG, Barnabas RV. Isoniazid preventive therapy plus antiretroviral therapy for the prevention of tuberculosis: a systematic review and meta-analysis of individual participant data. Lancet HIV 2021; 8:e8-e15. [PMID: 33387480 PMCID: PMC7875572 DOI: 10.1016/s2352-3018(20)30299-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND Isoniazid preventive therapy prevents active tuberculosis in people with HIV, but previous studies have found no evidence of benefit in people with HIV who had a negative tuberculin skin test, and a non-significant effect on mortality. We aimed to estimate the effect of isoniazid preventive therapy given with antiretroviral therapy (ART) for the prevention of tuberculosis and death among people with HIV across population subgroups. METHODS We searched PubMed, Embase, the Cochrane database, and conference abstracts from database inception to Jan 15, 2019, to identify potentially eligible randomised trials. Eligible studies were trials that enrolled HIV-positive adults (age ≥15 years) taking ART who were randomly assigned to either daily isoniazid preventive therapy plus ART or ART alone and followed up longitudinally for outcomes of incident tuberculosis and mortality. We approached all authors of included trials and requested individual participant data: coprimary outcomes were relative risk of incident tuberculosis and all-cause mortality. We did a single-stage meta-analysis of individual participant data using stratified Cox-proportional hazards models. We did prespecified subgroup analyses by sex, CD4 cell count, and evidence of immune sensitisation to tuberculosis (indicated by tuberculin skin test or interferon-γ release assays [IGRAs]). We also assessed the relative risk of liver injury in an additional prespecified analysis. This study is registered with PROSPERO, CRD42019121400. FINDINGS Of 838 records, we included three trials with data for 2611 participants and 8584·8 person-years of follow-up for the outcome of incident tuberculosis, and a subset of 2362 participants with 8631·6 person-years of follow-up for the coprimary outcome of all-cause mortality. Risk for tuberculosis was lower in participants given isoniazid preventive therapy and ART than participants given ART alone (hazard ratio [HR] 0·68, 95% CI 0·49-0·95, p=0·02). Risk of all-cause mortality was lower in participants given isoniazid preventive therapy and ART than participants given ART alone, but this difference was non-significant (HR 0·69, 95% CI 0·43-1·10, p=0·12). Participants with baseline CD4 counts of less than 500 cells per μL had increased risk of tuberculosis, but there was no significant difference in the benefit of isoniazid preventive therapy with ART by sex, baseline CD4 count, or results of tuberculin skin test or IGRAs. 65 (2·5%) of 2611 participants had raised alanine aminotransferase, but data were insufficient to calculate an HR. INTERPRETATION Isoniazid preventive therapy with ART prevents tuberculosis across demographic and HIV-specific and tuberculosis-specific subgroups, which supports efforts to further increase use of isoniazid preventive therapy with ART broadly among people living with HIV. FUNDING National Institutes of Health and National Institute of Allergy and Infectious Diseases.
Collapse
Affiliation(s)
- Jennifer M Ross
- Department of Global Health, University of Washington, Seattle, WA, USA; Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA.
| | | | | | - A Sarah Walker
- MRC Clinical Trials Unit, University College London, London, UK
| | - Adrienne E Shapiro
- Department of Global Health, University of Washington, Seattle, WA, USA; Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | | | | | - Serge Eholie
- University Félix Houphouet-Boigny, Abidjan, Côte d'Ivoire
| | | | - Andrew Boulle
- Wellcome Centre for Infectious Disease Research in Africa, University of Cape Town, Cape Town, South Africa; Centre for Infectious Disease Epidemiology and Research, University of Cape Town, Cape Town, South Africa; School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Gary Maartens
- Wellcome Centre for Infectious Disease Research in Africa, University of Cape Town, Cape Town, South Africa; Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Disease Research in Africa, University of Cape Town, Cape Town, South Africa; The Francis Crick Institute, London, UK; Department of Infectious Diseases, Imperial College London, London, UK
| | - Nathan Ford
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town, Cape Town, South Africa
| | - Jonathan E Golub
- School of Medicine, Center for Tuberculosis Research, Johns Hopkins University, Baltimore, MD, USA
| | - Brian G Williams
- South African Centre for Epidemiological Modelling and Analysis, Stellenbosch, South Africa
| | - Ruanne V Barnabas
- Department of Global Health, University of Washington, Seattle, WA, USA; Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| |
Collapse
|
52
|
Igari H, Takayanagi S, Yahaba M, Tsuyuzaki M, Taniguchi T, Suzuki K. Prevalence of positive IGRAs and innate immune system in HIV-infected individuals in Japan. J Infect Chemother 2020; 27:592-597. [PMID: 33386257 DOI: 10.1016/j.jiac.2020.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/17/2020] [Accepted: 11/10/2020] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Human immunodeficiency virus (HIV) infected individuals are at increased risk of developing active tuberculosis (TB). TB incidence remains higher than in non-HIV subjects after antiretroviral therapy (ART) initiation. This study was conducted to estimate the prevalence of positive IGRA, reflecting latent tuberculosis infection and/or a history of active TB, in HIV-infected individuals after ART initiation in Japan. METHODS Two IGRAs (Interferon (IFN)-γ release assays), QuantiFERON®-TB Gold Plus (QFT-Plus) and T-Spot®.TB (TSPOT), were used. We also analyzed the TB associated risk factors for the IGRAs results and the role of CD4+ T-cells, CD8+ T-cells and NK cells for producing IFN-γ. We also analyzed the risk factors for positive IGRA responses and the role of CD4+ T-cells, CD8+ T-cells and NK cells for producing IFN-γ. RESULTS One hundred eight-four subjects were prospectively enrolled. Median age was 49 years. The positivity rates of QFT-Plus and TSPOT were 7.6% [95%CI 4.6-12.4] and 2.7% [95%CI 1.2-6.2], respectively, with significant difference. TB-associated risk factors and NK cells ≥300/μL were selected as independently significant factors by multivariate logistic regression. The NK cell count revealed significant linear regression with IFN-γ production responding to TB-specific antigens. CONCLUSIONS The prevalence of positive IGRAs was 2.7%-7.6%. QFT-Plus would be practical for a higher positivity rate and reflect TB risk factors. The innate immune system, referring to IFN-γ production, plays an important role in the immune response to TB-specific antigens even after initiating ART.
Collapse
Affiliation(s)
- Hidetoshi Igari
- Division of Infection Control, Chiba University Hospital, 1-8-1 Inohana Chuo-Ku, Chiba, 260-8677, Japan.
| | - Shin Takayanagi
- Division of Infection Control, Chiba University Hospital, 1-8-1 Inohana Chuo-Ku, Chiba, 260-8677, Japan.
| | - Misuzu Yahaba
- Division of Infection Control, Chiba University Hospital, 1-8-1 Inohana Chuo-Ku, Chiba, 260-8677, Japan.
| | - Mizue Tsuyuzaki
- Chiba Foundation for Health Promotion and Disease Prevention, 32-14 Shin-Minato Mihama-ku, Chiba, 261-0002, Japan.
| | - Toshibumi Taniguchi
- Division of Infection Control, Chiba University Hospital, 1-8-1 Inohana Chuo-Ku, Chiba, 260-8677, Japan.
| | - Kiminori Suzuki
- Chiba Foundation for Health Promotion and Disease Prevention, 32-14 Shin-Minato Mihama-ku, Chiba, 261-0002, Japan.
| |
Collapse
|
53
|
Riccardi N, Villa S, Canetti D, Giacomelli A, Taramasso L, Martini M, Di Biagio A, Bragazzi NL, Brigo F, Sotgiu G, Besozzi G, Codecasa L. Missed opportunities in tb clinical practice: How to bend the curve? A medical, social, economic and ethical point of view. Tuberculosis (Edinb) 2020; 126:102041. [PMID: 33385833 DOI: 10.1016/j.tube.2020.102041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/10/2020] [Accepted: 12/10/2020] [Indexed: 10/22/2022]
Affiliation(s)
- Niccolò Riccardi
- StopTB Italia Onlus, Milan, Italy; Department of Infectious - Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, Verona, Italy
| | - Simone Villa
- StopTB Italia Onlus, Milan, Italy; Centre for Multidisciplinary Research in Health Science, University of Milan, Milan, Italy
| | - Diana Canetti
- StopTB Italia Onlus, Milan, Italy; Department of Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Giacomelli
- StopTB Italia Onlus, Milan, Italy; Department of Biomedical and Clinical Sciences DIBIC L. Sacco, University of Milan, Milan, Italy
| | - Lucia Taramasso
- Infectious Diseases Clinic, Ospedale Policlinico San Martino - IRCCS, Genoa, Italy
| | | | - Antonio Di Biagio
- StopTB Italia Onlus, Milan, Italy; Infectious Diseases Clinic, Ospedale Policlinico San Martino - IRCCS, Genoa, Italy
| | | | - Francesco Brigo
- Department of Neurology, Franz Tappeiner Hospital, Merano, Italy
| | - Giovanni Sotgiu
- StopTB Italia Onlus, Milan, Italy; Clinical Epidemiology and Medical Statistics Unit, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | | | - Luigi Codecasa
- StopTB Italia Onlus, Milan, Italy; Regional TB Reference Centre, Istituto Villa Marelli, Niguarda Hospital, Milan, Italy
| |
Collapse
|
54
|
Cost-effectiveness of newer technologies for the diagnosis of Mycobacterium tuberculosis infection in Brazilian people living with HIV. Sci Rep 2020; 10:21823. [PMID: 33311520 PMCID: PMC7733491 DOI: 10.1038/s41598-020-78737-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Tuberculosis is the leading cause of death among people living with HIV (PLH). Preventive tuberculosis therapy reduces mortality in PLH, especially in those with a positive tuberculin skin test (TST). New, more specific technologies for detecting latent tuberculosis infection (LTBI) are now commercially available. We sought to analyse the cost-effectiveness of four different strategies for the diagnosis of LTBI in PLH in Brazil, from the Brazilian public health care system perspective. We developed a Markov state-transition model comparing four strategies for the diagnosis of LTBI over 20 years. The strategies consisted of TST with the currently used protein purified derivative (PPD RT 23), two novel skin tests using recombinant allergens (Diaskintest [Generium Pharmaceutical, Moscow, Russia] and EC [Zhifei Longcom Biologic Pharmacy Co., Anhui, China]), and the QuantiFERON-TB-Gold-Plus (Qiagen, Hilden, Germany). The main outcome was cost (in 2020 US dollars) per quality-adjusted life years (QALY). For the base case scenario, the Diaskintest was dominant over all other examined strategies. The cost saving estimate per QALY was US $1375. In sensitivity analyses, the Diaskintest and other newer tests remained cost-saving compared to TST. For PLH, TST could be replaced by more specific tests in Brazil, considering the current national recommendations.
Collapse
|
55
|
Nabity SA, Mponda K, Gutreuter S, Surie D, Williams A, Sharma AJ, Schnaubelt ER, Marshall RE, Kirking HL, Zimba SB, Sunguti JL, Chisuwo L, Chiwaula MJ, Gregory JF, da Silva R, Odo M, Jahn A, Kalua T, Nyirenda R, Girma B, Mpunga J, Buono N, Maida A, Kim EJ, Gunde LJ, Mekonnen TF, Auld AF, Muula AS, Oeltmann JE. Protocol for a Case-Control Study to Investigate the Association of Pellagra With Isoniazid Exposure During Tuberculosis Preventive Treatment Scale-Up in Malawi. Front Public Health 2020; 8:551308. [PMID: 33324593 PMCID: PMC7726014 DOI: 10.3389/fpubh.2020.551308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/21/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Pellagra is caused by niacin (vitamin B3) deficiency and manifested by a distinctive dermatitis. Isoniazid is critical for treating tuberculosis globally and is a component of most regimens to prevent tuberculosis. Isoniazid may contribute to pellagra by disrupting intracellular niacin synthesis. In 2017, Malawian clinicians recognized a high incidence of pellagra-like rashes after scale-up of isoniazid preventive treatment (IPT) to people living with HIV (PLHIV). This increase in pellagra incidence among PLHIV coincided with a seasonal period of sustained food insecurity in the region, which obscured epidemiological interpretations. Although isoniazid has been implicated as a secondary cause of pellagra for decades, no hypothesis-driven epidemiological study has assessed this relationship in a population exposed to isoniazid. We developed this case-control protocol to assess the association between large-scale isoniazid distribution and pellagra in Malawi. Methods: We measure the relative odds of having pellagra among isoniazid-exposed people compared to those without exposure while controlling for other pellagra risk factors. Secondary aims include measuring time from isoniazid initiation to onset of dermatitis, comparing niacin metabolites 1-methylnicotinamide (1-MN), and l-methyl-2-pyridone-5-carboxamide (2-PYR) in urine as a proxy for total body niacin status among subpopulations, and describing clinical outcomes after 30-days multi-B vitamin (containing 300 mg nicotinamide daily) therapy and isoniazid cessation (if exposed). We aim to enroll 197 participants with pellagra and 788 age- and sex-matched controls (1:4 ratio) presenting at three dermatology clinics. Four randomly selected community clinics within 3-25 km of designated dermatology clinics will refer persons with pellagra-like symptoms to one of the study enrollment sites for diagnosis. Trained study dermatologists will conduct a detailed exposure questionnaire and perform anthropometric measurements. A subset of enrollees will provide a casual urine specimen for niacin metabolites quantification and/or point-of-care isoniazid detection to confirm whether participants recently ingested isoniazid. We will use conditional logistic regression, matching age and sex, to estimate odds ratios for the primary study aim. Discussion: The results of this study will inform the programmatic scale-up of isoniazid-containing regimens to prevent tuberculosis.
Collapse
Affiliation(s)
- Scott A Nabity
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Kelvin Mponda
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Steve Gutreuter
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Diya Surie
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Anne Williams
- National Center for Chronic Disease Prevention and Health Promotion, U.S. Centers for Disease Control and Prevention, Atlanta, GA, United States.,McKing Consulting Corporation, Atlanta, GA, United States
| | - Andrea J Sharma
- National Center for Chronic Disease Prevention and Health Promotion, U.S. Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Elizabeth R Schnaubelt
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Rebekah E Marshall
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Hannah L Kirking
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Suzgo B Zimba
- Elizabeth Glaser Pediatric AIDS Foundation, Lilongwe, Malawi
| | - Joram L Sunguti
- Elizabeth Glaser Pediatric AIDS Foundation, Lilongwe, Malawi
| | - Laphiod Chisuwo
- National Public Health Reference Laboratory, Public Health Institute of Malawi, Lilongwe, Malawi
| | - Mabvuto J Chiwaula
- National Public Health Reference Laboratory, Public Health Institute of Malawi, Lilongwe, Malawi
| | - Jesse F Gregory
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, United States
| | - Robin da Silva
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, United States
| | - Michael Odo
- Department of HIV and AIDS, Malawi Ministry of Health, Lilongwe, Malawi
| | - Andreas Jahn
- Department of HIV and AIDS, Malawi Ministry of Health, Lilongwe, Malawi
| | - Thokozani Kalua
- Department of HIV and AIDS, Malawi Ministry of Health, Lilongwe, Malawi
| | - Rose Nyirenda
- Department of HIV and AIDS, Malawi Ministry of Health, Lilongwe, Malawi
| | - Belaineh Girma
- National Tuberculosis Control Program, Malawi Ministry of Health, Lilongwe, Malawi
| | - James Mpunga
- National Tuberculosis Control Program, Malawi Ministry of Health, Lilongwe, Malawi
| | - Nicole Buono
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Lilongwe, Malawi
| | - Alice Maida
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Lilongwe, Malawi
| | - Evelyn J Kim
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Lilongwe, Malawi
| | - Laurence J Gunde
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Lilongwe, Malawi
| | - Tigest F Mekonnen
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Lilongwe, Malawi
| | - Andrew F Auld
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Lilongwe, Malawi
| | - Adamson S Muula
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - John E Oeltmann
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Atlanta, GA, United States
| |
Collapse
|
56
|
Campaniço A, Harjivan SG, Warner DF, Moreira R, Lopes F. Addressing Latent Tuberculosis: New Advances in Mimicking the Disease, Discovering Key Targets, and Designing Hit Compounds. Int J Mol Sci 2020; 21:ijms21228854. [PMID: 33238468 PMCID: PMC7700174 DOI: 10.3390/ijms21228854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Despite being discovered and isolated more than one hundred years ago, tuberculosis (TB) remains a global public health concern arch. Our inability to eradicate this bacillus is strongly related with the growing resistance, low compliance to current drugs, and the capacity of the bacteria to coexist in a state of asymptomatic latency. This last state can be sustained for years or even decades, waiting for a breach in the immune system to become active again. Furthermore, most current therapies are not efficacious against this state, failing to completely clear the infection. Over the years, a series of experimental methods have been developed to mimic the latent state, currently used in drug discovery, both in vitro and in vivo. Most of these methods focus in one specific latency inducing factor, with only a few taking into consideration the complexity of the granuloma and the genomic and proteomic consequences of each physiological factor. A series of targets specifically involved in latency have been studied over the years with promising scaffolds being discovered and explored. Taking in account that solving the latency problem is one of the keys to eradicate the disease, herein we compile current therapies and diagnosis techniques, methods to mimic latency and new targets and compounds in the pipeline of drug discovery.
Collapse
Affiliation(s)
- André Campaniço
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Shrika G. Harjivan
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Digby F. Warner
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa;
- Department of Pathology, SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- Welcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch 7701, South Africa
| | - Rui Moreira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Francisca Lopes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
- Correspondence:
| |
Collapse
|
57
|
Kalk E, Heekes A, Mehta U, de Waal R, Jacob N, Cohen K, Myer L, Davies MA, Maartens G, Boulle A. Safety and Effectiveness of Isoniazid Preventive Therapy in Pregnant Women Living with Human Immunodeficiency Virus on Antiretroviral Therapy: An Observational Study Using Linked Population Data. Clin Infect Dis 2020; 71:e351-e358. [PMID: 31900473 PMCID: PMC7643738 DOI: 10.1093/cid/ciz1224] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/31/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Isoniazid preventive therapy (IPT) is widely used to protect against tuberculosis (TB) in people living with human immunodeficiency virus (HIV). Data on the safety and efficacy of IPT in pregnant women living with HIV (PWLHIV) are mixed. We used an individual-level, population-wide health database to examine associations between antenatal IPT exposure and adverse pregnancy outcomes, maternal TB, all-cause mortality, and liver injury during pregnancy through 12 months postpartum. METHODS We used linked routine electronic health data generated in the public sector of the Western Cape, South Africa, to define a cohort of PWLHIV on antiretroviral therapy. Pregnancy outcomes were assessed using logistic regression; for maternal outcomes we applied a proportional hazards model with time-updated IPT exposure. RESULTS Of 43 971 PWLHIV, 16.6% received IPT. Women who received IPT were less likely to experience poor pregnancy outcomes (adjusted odds ratio [aOR], 0.83 [95% confidence interval {CI}, .78-.87]); this association strengthened with IPT started after the first trimester compared with none (aOR, 0.71 [95% CI, .65-.79]) or with first-trimester exposure (aOR, 0.64 [95% CI, .55-.75]). IPT reduced the risk of TB by approximately 30% (aHR, 0.71 [95% CI, .63-.81]; absolute risk difference, 1518/100 000 women). The effect was modified by CD4 cell count with protection conferred if CD4 count was ≤350 cells/μL (aHR, 0.51 [95% CI, .41-.63]) vs 0.93 [95% CI, .76-1.13] for CD4 count >350 cells/µL). CONCLUSIONS This analysis of programmatic data is reassuring regarding the safety of antenatal IPT, with the greatest benefits against TB disease observed in women with CD4 count ≤350 cells/μL.
Collapse
Affiliation(s)
- Emma Kalk
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Alexa Heekes
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
- Health Impact Assessment, Provincial Government of the Western Cape, Cape Town, South Africa
| | - Ushma Mehta
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Renee de Waal
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Nisha Jacob
- Division of Public Health Medicine, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Karen Cohen
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Landon Myer
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Mary-Ann Davies
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
- Health Impact Assessment, Provincial Government of the Western Cape, Cape Town, South Africa
| | - Gary Maartens
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Andrew Boulle
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
- Health Impact Assessment, Provincial Government of the Western Cape, Cape Town, South Africa
| |
Collapse
|
58
|
Ganatra SR, Bucşan AN, Alvarez X, Kumar S, Chatterjee A, Quezada M, Fish A, Singh DK, Singh B, Sharan R, Lee TH, Shanmugasundaram U, Velu V, Khader SA, Mehra S, Rengarajan J, Kaushal D. Antiretroviral therapy does not reduce tuberculosis reactivation in a tuberculosis-HIV coinfection model. J Clin Invest 2020; 130:5171-5179. [PMID: 32544085 PMCID: PMC7524506 DOI: 10.1172/jci136502] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
While the advent of combination antiretroviral therapy (ART) has significantly improved survival, tuberculosis (TB) remains the leading cause of death in the HIV-infected population. We used Mycobacterium tuberculosis/simian immunodeficiency virus-coinfected (M. tuberculosis/SIV-coinfected) macaques to model M. tuberculosis/HIV coinfection and study the impact of ART on TB reactivation due to HIV infection. Although ART significantly reduced viral loads and increased CD4+ T cell counts in blood and bronchoalveolar lavage (BAL) samples, it did not reduce the relative risk of SIV-induced TB reactivation in ART-treated macaques in the early phase of treatment. CD4+ T cells were poorly restored specifically in the lung interstitium, despite their significant restoration in the alveolar compartment of the lung as well as in the periphery. IDO1 induction in myeloid cells in the inducible bronchus-associated lymphoid tissue (iBALT) likely contributed to dysregulated T cell homing and impaired lung immunity. Thus, although ART was indispensable for controlling viral replication, restoring CD4+ T cells, and preventing opportunistic infection, it appeared inadequate in reversing the clinical signs of TB reactivation during the relatively short duration of ART administered in this study. This finding warrants the modeling of concurrent treatment of TB and HIV to potentially reduce the risk of reactivation of TB due to HIV to inform treatment strategies in patients with M. tuberculosis/HIV coinfection.
Collapse
Affiliation(s)
- Shashank R. Ganatra
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Allison N. Bucşan
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Xavier Alvarez
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Shyamesh Kumar
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Ayan Chatterjee
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Melanie Quezada
- Emory Vaccine Center and
- Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Abigail Fish
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Dhiraj K. Singh
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Bindu Singh
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Riti Sharan
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Tae-Hyung Lee
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Uma Shanmugasundaram
- Emory Vaccine Center and
- Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Vijayakumar Velu
- Emory Vaccine Center and
- Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shabaana A. Khader
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Smriti Mehra
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Jyothi Rengarajan
- Emory Vaccine Center and
- Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
59
|
Incidence of and risk factors for tuberculosis among people with HIV on antiretroviral therapy in the United Kingdom. AIDS 2020; 34:1813-1821. [PMID: 32501837 PMCID: PMC8635262 DOI: 10.1097/qad.0000000000002599] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Objective: The United Kingdom has a low tuberculosis incidence and earlier combination antiretroviral therapy (cART) is expected to have reduced incidence among people with HIV. Epidemiological patterns and risk factors for active tuberculosis were analysed over a 20-year period among people accessing HIV care at sites participating in the UK CHIC observational study. Design: Cohort analysis. Methods: Data were included for individuals over 15 years old attending for HIV care between 1996 and 2017 inclusive, with at least 3 months follow-up recorded. Incidence rates of new tuberculosis events were calculated and stratified by ethnicity (white/Black/other) as a proxy for tuberculosis exposure. Poisson regression models were used to determine the associations of calendar year, ethnicity and other potential risk factors after cART initiation. Results: Fifty-eight thousand seven hundred and seventy-six participants (26.3% women; 54.5% white, 32.0% Black, 13.5% other/unknown ethnicity; median (interquartile range) age 34 (29–42) years) were followed for 546 617 person-years. Seven hundred and four were treated for active tuberculosis [rate 1.3; 95% confidence interval (CI) 1.2–1.4/1000 person-years). Tuberculosis incidence decreased from 1.3 (1.2–1.5) to 0.6 (0.4–0.9)/1000 person-years from pre-2004 to 2011–2017. The decline among people of Black ethnicity was less steep than among those of white/other ethnicities, with incidence remaining high among Black participants in the latest period [2.1 (1.4–3.1)/1000 person-years]. Two hundred and eighty-three participants [191 (67%) Black African] had tuberculosis with viral load less than 50 copies/ml. Conclusion: Despite the known protective effect of cART against tuberculosis, a continuing disproportionately high incidence is seen among Black African people. Results support further interventions to prevent tuberculosis in this group.
Collapse
|
60
|
Nel J, Dlamini S, Meintjes G, Burton R, Black JM, Davies NECG, Hefer E, Maartens G, Mangena PM, Mathe MT, Moosa MY, Mulaudzi MB, Moorhouse M, Nash J, Nkonyane TC, Preiser W, Rassool MS, Stead D, van der Plas H, van Vuuren C, Venter WDF, Woods JF. Southern African HIV Clinicians Society guidelines for antiretroviral therapy in adults: 2020 update. South Afr J HIV Med 2020; 21:1115. [PMID: 33101723 PMCID: PMC7564911 DOI: 10.4102/sajhivmed.v21i1.1115] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 06/21/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jeremy Nel
- Helen Joseph Hospital, Department of Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Sipho Dlamini
- Department of Infectious Diseases, Faculty of Medicine, University of Cape Town, Cape Town, South Africa
| | - Graeme Meintjes
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Rosie Burton
- Southern African Medical Unit, Médecins Sans Frontières (MSF), Cape Town, South Africa
| | - John M Black
- Department of Medicine, Division of Infectious Diseases, Livingstone Tertiary Hospital, Port Elizabeth, South Africa
| | | | - Eric Hefer
- Private Practice Medical Adviser, Johannesburg, South Africa
| | - Gary Maartens
- Department of Medicine, Division of Clinical Pharmacology, University of Cape Town, Cape Town, South Africa
| | - Phetho M Mangena
- Department of Internal Medicine, School of Medicine, Pietersburg Hospital, Polokwane, South Africa.,Department of Medicine, School of Medicine, University of Limpopo, Turfloop, South Africa
| | | | - Mahomed-Yunus Moosa
- Department of Infectious Diseases, Division of Internal Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | - Michelle Moorhouse
- Reproductive Health and HIV Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jennifer Nash
- Specialist Family Physician, Amathole District Clinical Specialist Team, East London, South Africa
| | - Thandeka C Nkonyane
- Department of Infectious Diseases, Faculty of Medicine, Sefako Makgatho Health Sciences University, Pretoria, South Africa.,Department of Medicine, Dr George Mokhari Hospital, Pretoria, South Africa
| | - Wolfgang Preiser
- Department of Medical Virology, National Health Laboratory Service, Tygerberg, South Africa.,Department of Pathology, Faculty of Medicine and Health, Stellenbosch University, Cape Town, South Africa
| | - Mohammed S Rassool
- Clinical HIV Research Unit, Wits Health Consortium, Johannesburg, South Africa
| | - David Stead
- Department of Medicine, Faculty of Infectious Diseases, Frere and Cecilia Makiwane Hospitals, East London, South Africa.,Department of Medicine, Faculty of Health Sciences, Walter Sisulu University, Mthatha, South Africa
| | - Helen van der Plas
- Department of Infectious Diseases, Faculty of Medicine, University of Cape Town, Cape Town, South Africa
| | - Cloete van Vuuren
- Department of Internal Medicine, Military Hospital, Bloemfontein, South Africa.,Department of Internal Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Willem D F Venter
- Ezintsha, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Joana F Woods
- Ezintsha, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
61
|
Lesosky M, Rangaka MX, Pienaar C, Coussens AK, Goliath R, Mathee S, Mwansa-Kambafwile J, Maartens G, Wilkinson RJ, Wilkinson KA. Plasma Biomarkers to Detect Prevalent or Predict Progressive Tuberculosis Associated With Human Immunodeficiency Virus-1. Clin Infect Dis 2020; 69:295-305. [PMID: 30256919 PMCID: PMC6603269 DOI: 10.1093/cid/ciy823] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/21/2018] [Indexed: 12/14/2022] Open
Abstract
Background The risk of individuals infected with human immunodeficiency virus (HIV)-1 developing tuberculosis (TB) is high, while both prognostic and diagnostic tools remain insensitive. The potential for plasma biomarkers to predict which HIV-1–infected individuals are likely to progress to active disease is unknown. Methods Thirteen analytes were measured from QuantiFERON Gold in-tube (QFT) plasma samples in 421 HIV-1–infected persons recruited within the screening and enrollment phases of a randomized, controlled trial of isoniazid preventive therapy. Blood for QFT was obtained pre-randomization. Individuals were classified into prevalent TB, incident TB, and control groups. Comparisons between groups, supervised learning methods, and weighted correlation network analyses were applied utilizing the unstimulated and background-corrected plasma analyte concentrations. Results Unstimulated samples showed higher analyte concentrations in the prevalent and incident TB groups compared to the control group. The largest differences were seen for C-X-C motif chemokine 10 (CXCL10), interleukin-2 (IL-2), IL-1α, transforming growth factor-α (TGF-α). A predictive model analysis using unstimulated analytes discriminated best between the control and prevalent TB groups (area under the curve [AUC] = 0.9), reasonably well between the incident and prevalent TB groups (AUC > 0.8), and poorly between the control and incident TB groups. Unstimulated IL-2 and IFN-γ were ranked at or near the top for all comparisons, except the comparison between the control vs incident TB groups. Models using background-adjusted values performed poorly. Conclusions Single plasma biomarkers are unlikely to distinguish between disease states in HIV-1 co-infected individuals, and combinations of biomarkers are required. The ability to detect prevalent TB is potentially important, as no blood test hitherto has been suggested as having the utility to detect prevalent TB amongst HIV-1 co-infected persons.
Collapse
Affiliation(s)
- Maia Lesosky
- Division of Epidemiology & Biostatistics, School of Public Health and Family Medicine.,Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, Observatory, South Africa
| | - Molebogeng X Rangaka
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, Observatory, South Africa.,Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa.,Institute for Global Health, Faculty of Population Health Sciences, University College London, United Kingdom
| | - Cara Pienaar
- Division of Epidemiology & Biostatistics, School of Public Health and Family Medicine
| | - Anna K Coussens
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, Observatory, South Africa.,Department of Pathology, Faculty of Health Sciences, University of Cape Town, Observatory
| | - Rene Goliath
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, Observatory, South Africa
| | - Shaheed Mathee
- Site B Khayelitsha Community Health Centre, Western Cape Department of Health, South Africa
| | - Judith Mwansa-Kambafwile
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, Observatory, South Africa
| | - Gary Maartens
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, Observatory, South Africa.,Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa.,Department of Medicine, Imperial College London, London, United Kingdom.,The Francis Crick Institute, London, United Kingdom
| | - Katalin Andrea Wilkinson
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, Observatory, South Africa.,Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa.,The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
62
|
Swindells S, Hughes M, Chaisson RE. Trials of Tuberculosis-Preventive Therapy in People with HIV Infection. Am J Respir Crit Care Med 2020; 202:304-305. [PMID: 32282226 PMCID: PMC7365362 DOI: 10.1164/rccm.202003-0761le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | - Michael Hughes
- Harvard TH Chan School of Public HealthBoston, Massachusettsand
| | | |
Collapse
|
63
|
Agizew T, Surie D, Oeltmann JE, Letebele M, Pals S, Mathebula U, Mathoma A, Kassa M, Hamda S, Pono P, Rankgoane-Pono G, Boyd R, Auld A, Finlay A. Tuberculosis preventive treatment opportunities at antiretroviral therapy initiation and follow-up visits. Public Health Action 2020; 10:64-69. [PMID: 32639479 DOI: 10.5588/pha.19.0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 02/18/2020] [Indexed: 11/10/2022] Open
Abstract
Setting Twenty-two clinics providing HIV care and treatment in Botswana where tuberculosis (TB) and HIV comorbidity is as high as 49%. Objectives To assess eligibility of TB preventive treatment (TPT) at antiretroviral therapy (ART) initiation and at four follow-up visits (FUVs), and to describe the TB prevalence and associated factors at baseline and yield of TB diagnoses at each FUV. Design A prospective study of routinely collected data on people living with HIV (PLHIV) enrolled into care for the Xpert® MTB/RIF Package Rollout Evaluation Study between 2012 and 2015. Results Of 6041 PLHIV initiating ART, eligibility for TPT was 69% (4177/6041) at baseline and 93% (5408/5815); 95% (5234/5514); 96% (4869/5079); and 97% (3925/4055) at FUV1, FUV2, FUV3, and FUV4, respectively. TB prevalence at baseline was 11% and 2%, 3%, 3% and 6% at each subsequent FUV. At baseline, independent risk factors for prevalent TB were CD4 <200 cells/mm3 (aOR = 1.4, P = 0.030); anemia (aOR = 2.39, P < 0.001); cough (aOR = 11.21, P < 0.001); fever (aOR = 2.15, P = 0.001); and weight loss (aOR = 2.60, P = 0.002). Conclusion Eligibility for TPT initiation is higher at visits post-ART initiation, while most cases of active TB were identified at ART initiation. Missed opportunities for TB further compromises TB control effort among PLHIV in Botswana.
Collapse
Affiliation(s)
- T Agizew
- Centers for Disease Control and Prevention (CDC), Gaborone, Botswana.,Department of Family Medicine and Public Health, Faculty of Medicine, University of Botswana, Gaborone, Botswana.,School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - D Surie
- Division of Global HIV/AIDS and Tuberculosis, CDC, Atlanta, GA, USA
| | - J E Oeltmann
- Division of Global HIV/AIDS and Tuberculosis, CDC, Atlanta, GA, USA
| | - M Letebele
- Centers for Disease Control and Prevention (CDC), Gaborone, Botswana
| | - S Pals
- Division of Global HIV/AIDS and Tuberculosis, CDC, Atlanta, GA, USA
| | - U Mathebula
- Centers for Disease Control and Prevention (CDC), Gaborone, Botswana
| | - A Mathoma
- Centers for Disease Control and Prevention (CDC), Gaborone, Botswana
| | - M Kassa
- Department of Anaesthesia and Critical Care, University of Botswana, Gaborone, Botswana
| | - S Hamda
- Department of Family Medicine and Public Health, Faculty of Medicine, University of Botswana, Gaborone, Botswana
| | - P Pono
- Department of HIV/AIDS Prevention and Care, Ministry of Health and Wellness, Gaborone, Botswana
| | - G Rankgoane-Pono
- National Tuberculosis Control Programme, Ministry of Health and Wellness, Gaborone, Botswana
| | - R Boyd
- Centers for Disease Control and Prevention (CDC), Gaborone, Botswana.,Division of Tuberculosis Elimination, CDC, Atlanta, GA, USA
| | - A Auld
- Division of Global HIV/AIDS and Tuberculosis, CDC, Atlanta, GA, USA
| | - A Finlay
- Centers for Disease Control and Prevention (CDC), Gaborone, Botswana.,Division of Tuberculosis Elimination, CDC, Atlanta, GA, USA
| |
Collapse
|
64
|
|
65
|
Abstract
Abstract
Purpose of Review
Tuberculosis is the number one infectious killer of people with HIV worldwide, but it can be both prevented and treated. Prevention of tuberculosis by screening for and treating latent tuberculosis infection (LTBI), along with the initiation of antiretroviral therapy (ART), is the key component of HIV care.
Recent Findings
While access to ART has increased worldwide, uptake and completion of LTBI treatment regimens among people living with HIV (PWH) are very poor. Concomitant TB-preventive therapy and ART are complex because of drug–drug interactions, but these can be managed. Recent clinical trials of shorter preventive regimens have demonstrated safety and efficacy in PWH with higher completion rates. More research is needed to guide TB-preventive therapy in children and in pregnant women, and for drug-resistant TB (DR-TB).
Summary
Antiretroviral therapy and tuberculosis-preventive treatment regimens can be optimized to avoid drug–drug interactions, decrease pill burden and duration, and minimize side effects in order to increase adherence and treatment completion rates among PWH and LTBI.
Collapse
|
66
|
Tweya H, Feldacker C, Mpunga J, Kanyerere H, Heller T, Ganesh P, Nkosi D, Kalulu M, Sinkala G, Satumba T, Phiri S. The shift in tuberculosis timing among people living with HIV in the course of antiretroviral therapy scale-up in Malawi. J Int AIDS Soc 2020; 22:e25240. [PMID: 31038836 PMCID: PMC6490056 DOI: 10.1002/jia2.25240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 12/17/2018] [Indexed: 01/05/2023] Open
Abstract
Introduction Although the use of antiretroviral therapy (ART) reduces HIV‐associated tuberculosis (TB), patients living with HIV receiving ART remain at a higher risk of developing TB compared to those without HIV. We investigated the incidence of TB and the proportion of HIV‐associated TB cases among patients living with HIV who are receiving ART. Methods The study used TB registration and ART programme data collected between 2008 and 2017 from an integrated, public clinic in urban Lilongwe, Malawi. ART initiation was based on either WHO clinical staging or CD4 cell count. The CD4 thresholds for ART initiation eligibility was initially 250 cells/μL then changed to 350 cells/μL in 2011, 500 cells/μL in 2014 and to universal treatment upon diagnosis from 2016. Using TB registration data, we calculated the proportion of TB/HIV patients who were already on ART when they registered for TB treatment by year of TB registration. ART registration data were used to examine TB incidence by calendar year of ART follow‐up and by time on ART. Results The overall proportion of TB/patients living with HIV who started TB treatment while on ART increased from 21% in 2008 to 81% in 2017 but numbers remained relatively constant at 500 TB cases annually. The overall incidence rate of TB among patients on ART was 1.35/100 person‐years (95% CI 1.28 to 1.42). The incidence of TB by time on ART decreased from 6.4/100 person‐years in the first three months of ART to 0.4/100 person‐years after eight years on ART. TB incidence was highest in the first month on ART. The annual rate of TB among patients on ART rapidly decreased each calendar year and stabilized at 1% after 2013. Although the risk of developing TB decreased with year of ART initiation in univariable analysis, there was no significant association after adjusting for sex, age and reason for ART eligibility. Conclusions The decline in TB incidence over calendar years suggests protective effects of early ART initiation. The high TB incidence within the first month of ART highlights the need for more sensitive tools such as X‐ray and GeneXpert to identify patients living with HIV who have clinical and subclinical TB disease at ART initiation.
Collapse
Affiliation(s)
- Hannock Tweya
- The International Union Against Tuberculosis and Lung Disease, Paris, France.,Lighthouse Trust, Lilongwe, Malawi
| | - Caryl Feldacker
- International Training and Education Center for Health, University of Washington, Seattle, WA, USA.,Department of Global Health, University of Washington, Seattle, WA, USA
| | - James Mpunga
- National Tuberculosis Control Programme, Community Health Science Unit, Lilongwe, Malawi
| | - Henry Kanyerere
- National Tuberculosis Control Programme, Community Health Science Unit, Lilongwe, Malawi
| | | | | | - Dave Nkosi
- Bwaila District Hospital, Lilongwe, Malawi
| | | | | | | | - Sam Phiri
- Lighthouse Trust, Lilongwe, Malawi.,Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA.,Department of Public Health, College of Medicine, School of Public Health and Family Medicine, University of Malawi, Zomba, Malawi
| |
Collapse
|
67
|
Abstract
Treatment of latent tuberculosis infection (LTBI) is an important component of TB control and elimination. LTBI treatment regimens include once-weekly isoniazid plus rifapentine for 3 months, daily rifampin for 4 months, daily isoniazid plus rifampin for 3-4 months, and daily isoniazid for 6-9 months. Isoniazid monotherapy is efficacious in preventing TB disease, but the rifampin- and rifapentine-containing regimens are shorter and have similar efficacy, adequate safety, and higher treatment completion rates. Novel vaccine strategies, host immunity-directed therapies and ultrashort antimicrobial regimens for TB prevention, such as daily isoniazid plus rifapentine for 1 month, are under evaluation.
Collapse
Affiliation(s)
- Moises A Huaman
- Department of Internal Medicine, Division of Infectious Diseases, University of Cincinnati College of Medicine, University of Cincinnati, 200 Albert Sabin Way, Room 3112, Cincinnati, OH 45267, USA; Hamilton County Public Health Tuberculosis Control Program, 184 McMillan Street, Cincinnati, OH 45219, USA; Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, 1161 21st Avenue South, A-2200 Medical Center North, Nashville, TN 37232, USA.
| | - Timothy R Sterling
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, 1161 21st Avenue South, A-2200 Medical Center North, Nashville, TN 37232, USA; Department of Medicine, Division of Infectious Diseases, Vanderbilt University School of Medicine, Vanderbilt University, 1161 21st Avenue South, A-2209 MCN, Nashville, TN 37232, USA
| |
Collapse
|
68
|
Kim HY, Hanrahan CF, Dowdy DW, Martinson NA, Golub JE, Bridges JF. Priorities among HIV-positive individuals for tuberculosis preventive therapies. Int J Tuberc Lung Dis 2020; 24:396-402. [PMID: 32317063 PMCID: PMC7518293 DOI: 10.5588/ijtld.18.0740] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND: There has been slow uptake of isoniazid preventive therapy (IPT) among people living with HIV (PLWH).METHODS: We surveyed adults recently diagnosed with HIV in 14 South African primary health clinics. Based on the literature and qualitative interviews, sixteen potential barriers and facilitators related to preventive therapy among PLWH were selected. Best-worst scaling (BWS) was used to quantify the relative importance of the attributes. BWS scores were calculated based on the frequency of participants' selecting each attribute as the best or worst among six options (across multiple choice sets) and rescaled from 0 (always selected as worst) to 100 (always selected as best) and compared by currently receiving IPT or not.RESULTS: Among 342 patients surveyed, 33% (n = 114) were currently taking IPT. Having the same standard of life as someone without HIV was most highly prioritized (BWS score = 67.3, SE = 0.6), followed by trust in healthcare providers (score, 66.3 ± 0.6). Poor standard of care in public clinics (score, 30.6 ± 0.6) and side effects of medications (score, 33.7 ± 0.6) were least prioritized. BWS scores differed by IPT status for few attributes, but overall ranking was similar (spearman's rho = 0.9).CONCLUSION: Perceived benefits of preventive therapy were high among PLWH. IPT prescription by healthcare providers should be encouraged to enhance IPT uptake among PLWH.
Collapse
Affiliation(s)
- Hae-Young Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Africa Health Research Institute, KwaZulu-Natal, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP)
| | - Colleen F. Hanrahan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - David W. Dowdy
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Neil A. Martinson
- Perinatal HIV Research Unit, University of Witwatersrand, Johannesburg, South Africa
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, MD, USA
| | - Jonathan E. Golub
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, MD, USA
| | - John F.P. Bridges
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
69
|
Sterling TR, Njie G, Zenner D, Cohn DL, Reves R, Ahmed A, Menzies D, Horsburgh CR, Crane CM, Burgos M, LoBue P, Winston CA, Belknap R. Guidelines for the treatment of latent tuberculosis infection: Recommendations from the National Tuberculosis Controllers Association and CDC, 2020. Am J Transplant 2020. [DOI: 10.1111/ajt.15841] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
| | - Gibril Njie
- National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention Division of Tuberculosis Elimination CDC Atlanta Georgia USA
| | - Dominik Zenner
- Institute for Global Health University College London London England
| | - David L. Cohn
- Denver Health and Hospital Authority Denver Colorado USA
| | - Randall Reves
- Denver Health and Hospital Authority Denver Colorado USA
| | - Amina Ahmed
- Levine Children’s Hospital Charlotte North Carolina USA
| | - Dick Menzies
- Montreal Chest Institute and McGill International TB Centre Montreal Canada USA
| | - C. Robert Horsburgh
- Boston University Schools of Public Health and Medicine Boston Massachusetts USA
| | - Charles M. Crane
- National Tuberculosis Controllers Association Smyrna Georgia USA
| | - Marcos Burgos
- National Tuberculosis Controllers Association Smyrna Georgia USA
- New Mexico Department of Health University of New Mexico Health Science Center Albuquerque New Mexico USA
| | - Philip LoBue
- National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention Division of Tuberculosis Elimination CDC Atlanta Georgia USA
| | - Carla A. Winston
- National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention Division of Tuberculosis Elimination CDC Atlanta Georgia USA
| | - Robert Belknap
- Denver Health and Hospital Authority Denver Colorado USA
- National Tuberculosis Controllers Association Smyrna Georgia USA
| |
Collapse
|
70
|
Aemro A, Jember A, Anlay DZ. Incidence and predictors of tuberculosis occurrence among adults on antiretroviral therapy at Debre Markos referral hospital, Northwest Ethiopia: retrospective follow-up study. BMC Infect Dis 2020; 20:245. [PMID: 32216747 PMCID: PMC7098113 DOI: 10.1186/s12879-020-04959-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 03/11/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In resource limited settings, Tuberculosis (TB) is a major cause of morbidity and mortality among patients on antiretroviral treatment. Ethiopia is one of the 30 high TB burden countries. TB causes burden in healthcare system and challenge the effectiveness of HIV care. This study was to assess incidence and predictors of Tuberculosis among adults on antiretroviral therapy at Debre Markos Referral Hospital, Northwest Ethiopia, 2019. METHODS Institution based retrospective follow up study was conducted among adults on ART newly enrolled from 2014 to 2018 at Debre Markos Referral Hospital. Simple random sampling technique was used to select patients chart. Data was entered to EPI- INFO version 7.2.2.6 and analyzed using Stata 14.0. Tuberculosis incidence rate was computed and described using frequency tables. Both bivariable and multivariable Cox proportional hazard models was fitted to identify predictors of TB. RESULTS Out of the 536 patients chart reviewed, 494 patient records were included in the analysis. A total of 62 patients developed new TB cases during the follow up period of 1000.22 Person Years (PY); which gives an overall incidence rate of 6.19 cases per 100 PY (95% CI: 4.83-7.95). The highest rate was seen within the first year of follow up. After adjustment base line Hemoglobin < 10 g/dl (AHR = 5.25; 95% CI: 2.52-10.95), ambulatory/bedridden patients at enrolment (AHR = 2.31; 95% CI: 1.13-4.73), having fair or poor ART adherence (AHR = 3.22; 95% CI: 1.64-6.31) were associated with increased risk of tuberculosis whereas taking Isoniazid Preventive Therapy (IPT) (AHR = 0.33; 95% CI: 0.12-0.85) were protective factors of TB occurrence. CONCLUSION TB incidence was high among adults on ART especially in the first year of enrollment to ART. Low hemoglobin level, ambulatory or bedridden functional status, non-adherence to ART and IPT usage status were found to be independent predictors. Hence, continuous follow up for ART adherence and provision of IPT has a great importance to reduce the risk of TB.
Collapse
Affiliation(s)
- Agazhe Aemro
- Department of Medical Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebaw Jember
- Department of Medical Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Degefaye Zelalem Anlay
- Unit of Community Health Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
71
|
Abubakar I, Cobelens FGJ, Rangaka MX. The Risk of Falsely Declaring Noninferiority of Novel Latent Tuberculosis Treatment in Large Trials. Am J Respir Crit Care Med 2020; 201:511-513. [PMID: 31816247 PMCID: PMC7047458 DOI: 10.1164/rccm.201911-2258ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | - Frank G J Cobelens
- Department of Global Health.,Amsterdam Institute for Global Health and DevelopmentAmsterdam University Medical CentersAmsterdam, the Netherlands
| | - Molebogeng X Rangaka
- UCL Institute for Global HealthLondon, United Kingdom.,Institute of Infectious Disease and Molecular Medicineand.,School of Public HealthUniversity of Cape TownCape Town, South Africa
| |
Collapse
|
72
|
Stout JE, Turner NA, Belknap RW, Horsburgh CR, Sterling TR, Phillips PPJ. Optimizing the Design of Latent Tuberculosis Treatment Trials: Insights from Mathematical Modeling. Am J Respir Crit Care Med 2020; 201:598-605. [PMID: 31711306 PMCID: PMC10797496 DOI: 10.1164/rccm.201908-1606oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/11/2019] [Indexed: 11/16/2022] Open
Abstract
Rationale: Noninferiority trials of treatment for latent tuberculosis infection (LTBI) are challenging because of imperfect LTBI diagnostic tests.Objectives: To assess the effect on study outcomes of different enrollment strategies for a noninferiority trial of LTBI treatment.Methods: We mathematically simulated a two-arm randomized clinical trial of LTBI in which the experimental therapy was 50% efficacious and the control was 80% efficacious, with an absolute 0.75% noninferiority margin. Five enrollment strategies were assessed: 1) enroll based on no LTBI diagnostic test; 2) enroll based on a positive tuberculin skin test (TST); 3) enroll based on a positive IFN-γ release assay (IGRA); 4) enroll based on either a positive TST or IGRA; and 5) enroll regardless of test result, assuming 70% had negative TSTs, 20% positive TSTs, and 10% unknown results.Measurements and Main Results: Under most LTBI prevalence assumptions, enrolling based on a positive IGRA was least likely to result in falsely declaring noninferiority of the experimental regimen. Enrolling based on no test or regardless of test result led to falsely declaring noninferiority unless LTBI prevalence in the underlying population was higher than 45%. Enrolling based on a mix of TST and IGRA substantially reduced the likelihood of falsely declaring noninferiority over enrolling based on TST alone, even if as many as 70% of participants were enrolled based on positive TST.Conclusions: Noninferiority trials of LTBI should enroll based on the most specific diagnostic tests available (i.e., IGRAs) to avoid misclassifying inferior treatment regimens as noninferior.
Collapse
Affiliation(s)
- Jason E. Stout
- Division of Infectious Diseases and International Health, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Nicholas A. Turner
- Division of Infectious Diseases and International Health, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Robert W. Belknap
- Denver Health and Hospital Authority and Division of Infectious Diseases, Department of Medicine, University of Colorado, Denver, Colorado
| | - C. Robert Horsburgh
- Departments of Epidemiology, Biostatistics, Global Health, and Medicine, Boston University Schools of Public Health and Medicine, Boston, Massachusetts
| | - Timothy R. Sterling
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Patrick P. J. Phillips
- Department of Medicine, UCSF Center for Tuberculosis, University of California–San Francisco, San Francisco, California
| |
Collapse
|
73
|
Sterling TR, Njie G, Zenner D, Cohn DL, Reves R, Ahmed A, Menzies D, Horsburgh CR, Crane CM, Burgos M, LoBue P, Winston CA, Belknap R. Guidelines for the Treatment of Latent Tuberculosis Infection: Recommendations from the National Tuberculosis Controllers Association and CDC, 2020. MMWR Recomm Rep 2020; 69:1-11. [PMID: 32053584 PMCID: PMC7041302 DOI: 10.15585/mmwr.rr6901a1] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Comprehensive guidelines for treatment of latent tuberculosis infection (LTBI) among persons living in the United States were last published in 2000 (American Thoracic Society. CDC targeted tuberculin testing and treatment of latent tuberculosis infection. Am J Respir Crit Care Med 2000;161:S221–47). Since then, several new regimens have been evaluated in clinical trials. To update previous guidelines, the National Tuberculosis Controllers Association (NTCA) and CDC convened a committee to conduct a systematic literature review and make new recommendations for the most effective and least toxic regimens for treatment of LTBI among persons who live in the United States. The systematic literature review included clinical trials of regimens to treat LTBI. Quality of evidence (high, moderate, low, or very low) from clinical trial comparisons was appraised using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) criteria. In addition, a network meta-analysis evaluated regimens that had not been compared directly in clinical trials. The effectiveness outcome was tuberculosis disease; the toxicity outcome was hepatotoxicity. Strong GRADE recommendations required at least moderate evidence of effectiveness and that the desirable consequences outweighed the undesirable consequences in the majority of patients. Conditional GRADE recommendations were made when determination of whether desirable consequences outweighed undesirable consequences was uncertain (e.g., with low-quality evidence). These updated 2020 LTBI treatment guidelines include the NTCA- and CDC-recommended treatment regimens that comprise three preferred rifamycin-based regimens and two alternative monotherapy regimens with daily isoniazid. All recommended treatment regimens are intended for persons infected with Mycobacterium tuberculosis that is presumed to be susceptible to isoniazid or rifampin. These updated guidelines do not apply when evidence is available that the infecting M. tuberculosis strain is resistant to both isoniazid and rifampin; recommendations for treating contacts exposed to multidrug-resistant tuberculosis were published in 2019 (Nahid P, Mase SR Migliori GB, et al. Treatment of drug-resistant tuberculosis. An official ATS/CDC/ERS/IDSA clinical practice guideline. Am J Respir Crit Care Med 2019;200:e93–e142). The three rifamycin-based preferred regimens are 3 months of once-weekly isoniazid plus rifapentine, 4 months of daily rifampin, or 3 months of daily isoniazid plus rifampin. Prescribing providers or pharmacists who are unfamiliar with rifampin and rifapentine might confuse the two drugs. They are not interchangeable, and caution should be taken to ensure that patients receive the correct medication for the intended regimen. Preference for these rifamycin-based regimens was made on the basis of effectiveness, safety, and high treatment completion rates. The two alternative treatment regimens are daily isoniazid for 6 or 9 months; isoniazid monotherapy is efficacious but has higher toxicity risk and lower treatment completion rates than shorter rifamycin-based regimens. In summary, short-course (3- to 4-month) rifamycin-based treatment regimens are preferred over longer-course (6–9 month) isoniazid monotherapy for treatment of LTBI. These updated guidelines can be used by clinicians, public health officials, policymakers, health care organizations, and other state and local stakeholders who might need to adapt them to fit individual clinical circumstances.
Collapse
|
74
|
Waters R, Ndengane M, Abrahams MR, Diedrich CR, Wilkinson RJ, Coussens AK. The Mtb-HIV syndemic interaction: why treating M. tuberculosis infection may be crucial for HIV-1 eradication. Future Virol 2020; 15:101-125. [PMID: 32273900 PMCID: PMC7132588 DOI: 10.2217/fvl-2019-0069] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Accelerated tuberculosis and AIDS progression seen in HIV-1 and Mycobacterium tuberculosis (Mtb)-coinfected individuals indicates the important interaction between these syndemic pathogens. The immunological interaction between HIV-1 and Mtb has been largely defined by how the virus exacerbates tuberculosis disease pathogenesis. Understanding of the mechanisms by which pre-existing or subsequent Mtb infection may favor the replication, persistence and progression of HIV, is less characterized. We present a rationale for the critical consideration of ‘latent’ Mtb infection in HIV-1 prevention and cure strategies. In support of this position, we review evidence of the effect of Mtb infection on HIV-1 acquisition, replication and persistence. We propose that ‘latent’ Mtb infection may have considerable impact on HIV-1 pathogenesis and the continuing HIV-1 epidemic in sub-Saharan Africa.
Collapse
Affiliation(s)
- Robyn Waters
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Medicine, University of Cape Town, Observatory 7925, WC, South Africa
| | - Mthawelanga Ndengane
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Pathology, University of Cape Town, Observatory 7925, WC, South Africa
| | - Melissa-Rose Abrahams
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Pathology, University of Cape Town, Observatory 7925, WC, South Africa
| | - Collin R Diedrich
- Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Infectious Diseases, Imperial College London, London W2 1PG, United Kingdom.,The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Anna K Coussens
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Pathology, University of Cape Town, Observatory 7925, WC, South Africa.,Infectious Diseases and Immune Defence Division, The Walter & Eliza Hall Institute of Medical Research, Parkville 3279, VIC, Australia.,Division of Medical Biology, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville 3279, VIC, Australia
| |
Collapse
|
75
|
LaCourse SM, Richardson BA, Kinuthia J, Warr AJ, Maleche-Obimbo E, Matemo D, Cranmer LM, Escudero JN, Hawn TR, John-Stewart GC. Infant TB Infection Prevention Study (iTIPS): a randomised trial protocol evaluating isoniazid to prevent M. tuberculosis infection in HIV-exposed uninfected children. BMJ Open 2020; 10:e034308. [PMID: 31969368 PMCID: PMC7045242 DOI: 10.1136/bmjopen-2019-034308] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/05/2019] [Accepted: 01/07/2020] [Indexed: 12/04/2022] Open
Abstract
INTRODUCTION HIV-exposed uninfected (HEU) infants in tuberculosis (TB) endemic settings are at high risk of Mycobacterium tuberculosis (Mtb) infection and TB disease, even in the absence of known Mtb exposure. Because infancy is a time of rapid progression from primary infection to active TB disease, it is important to define when and how TB preventive interventions exert their effect in order to develop effective prevention strategies in this high-risk population. METHODS AND ANALYSIS We designed a non-blinded randomised controlled trial to determine efficacy of isoniazid (INH) to prevent primary Mtb infection among HEU children. Target sample size is 300 (150 infants in each arm). Children are enrolled at 6 weeks of age from maternal and child health clinics in Kenya and are randomised to receive 12 months of daily INH ~10 mg/kg plus pyridoxine or no INH. The primary endpoint is Mtb infection, assessed by interferon-gamma release assay QuantiFERON-TB Gold Plus (QFT-Plus) or tuberculin skin test after 12 months post-enrolment. Secondary outcomes include severe adverse events, expanded Mtb infection definition using additional QFT-Plus supernatant markers and determining correlates of Mtb infection. Exploratory analyses include a combined outcome of TB infection, disease and mortality, and sensitivity analyses excluding infants with baseline TB-specific responses on flow cytometry. ETHICS AND DISSEMINATION An external and independent Data and Safety Monitoring Board monitors adverse events. Results will be disseminated through peer-reviewed journals, presentations at local and international conferences to national and global policy-makers, the local community and participants. TRIAL REGISTRATION NUMBER NCT02613169; Pre-results.
Collapse
Affiliation(s)
- Sylvia M LaCourse
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Barbra A Richardson
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - John Kinuthia
- Research and Programs, Kenyatta National Hospital, Nairobi, Kenya
- Department of Reproductive Health, Kenyatta National Hospital, Nairobi, Kenya
| | - A J Warr
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | | | - Daniel Matemo
- Research and Programs, Kenyatta National Hospital, Nairobi, Kenya
| | - Lisa M Cranmer
- Department of Pediatrics, Division of Infectious Diseases, Emory University, Atlanta, Georgia, USA
- Children's Healthcare of Atlanta Inc, Atlanta, Georgia, USA
| | - Jaclyn N Escudero
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Thomas R Hawn
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Grace C John-Stewart
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
76
|
Does Isoniazid Preventive Therapy Provide Better Treatment Outcomes in HIV-Infected Individuals in Northern Ethiopia? A Retrospective Cohort Study. AIDS Res Treat 2020; 2020:7025738. [PMID: 32411454 PMCID: PMC7204289 DOI: 10.1155/2020/7025738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/26/2019] [Accepted: 12/09/2019] [Indexed: 11/17/2022] Open
Abstract
Objectives Early antiretroviral therapy (ART), isoniazid preventive therapy (IPT), and isoniazid-rifapentine (3HP) are effective strategies for preventing tuberculosis (TB) among people living with HIV (PLHIV). The study aimed to determine the effect of IPT on the TB incidence, follow-up CD4+ T cells, and all-cause mortality rate. Participants. Eligible patients on ART (n = 1, 863) were categorized into one-to-two ratios of exposed groups to IPT (n = 621) and nonexposed groups to IPT (n = 1, 242). Exposed groups entered the cohort at their first prescription of IPT, and unexposed groups entered into the study at the first prescription of ART and then followed until the occurrence of the outcome or date of administrative censoring (June 30, 2017). The outcome endpoints were TB incidence, follow-up CD4+ T cells, and all-cause mortality rate. Results The follow-up CD4+ T cells for the exposed and nonexposed groups were 405.74 and 366.95 cells/mm (World Health Organization (WHO), 2017), respectively, a statistically significant finding (t 1861 = -3.770, p < 0.0001; Cohen's d = 0.186). Nine percent of the exposed patients (620 incidence of TB per 100,000 person-years (PYs)) and 21.9% of the nonexposed patients (3160 incidence of TB per 100,000 PYs) developed TB. Mortality rate (per 100,000 PYs) was 440 for the exposed and 1490 for the unexposed patients. Statistically significant determinants of the all-cause mortality were unscheduled follow-up (AHR = 1.601; 95% CI: 1.154-2.222) and unable to work properly (AHR = 2.324; 95% CI: 1.643-3.288). Conclusion This study demonstrates the effect of IPT in reducing incidence of TB and all-cause mortality rate and improving follow-up CD4+ T cells. Promoting IPT use can help to achieve the TB eradicating national agenda in Ethiopia.
Collapse
|
77
|
Eriksen J, Carlander C, Albert J, Flamholc L, Gisslén M, Navér L, Svedhem V, Yilmaz A, Sönnerborg A. Antiretroviral treatment for HIV infection: Swedish recommendations 2019. Infect Dis (Lond) 2020; 52:295-329. [PMID: 31928282 DOI: 10.1080/23744235.2019.1707867] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The Swedish Reference Group for Antiviral Therapy (RAV) published recommendations for the treatment of HIV infection in this journal most recently in 2017. An expert group under the guidance of RAV here provides updated recommendations. The most important updates in the present guidelines are the following: (a) The risk of HIV transmission through condomless sex from individuals with fully suppressed HIV viral load is effectively zero. (b) Pre-exposure prophylaxis (PrEP) is recommended for groups with a high risk of HIV infection. (c) Since the last update, two new substances have been registered: bictegravir and doravirine. (d) Dual treatment may be an alternative in selected patients, using lamivudine + dolutegravir or lamivudine + boosted darunavir/atazanavir. As with previous publications, recommendations are evidence-graded in accordance with the Oxford Centre for Evidence Based Medicine. This document does not cover treatment of opportunistic infections and tumours.
Collapse
Affiliation(s)
- Jaran Eriksen
- Unit of Infectious Diseases/Venhälsan, Södersjukhuset, Stockholm, Sweden.,Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Christina Carlander
- Department of Infectious Diseases, Västmanland County Hospital, Västerås, Sweden.,Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jan Albert
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Leo Flamholc
- Department of Infectious Diseases, Skåne University Hospital, Malmö, Sweden
| | - Magnus Gisslén
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lars Navér
- Division of Paediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden.,Department of Paediatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Veronica Svedhem
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Aylin Yilmaz
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Sönnerborg
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden.,Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
78
|
Reddy MM, Thekkur P, Ramya N, Kamath PBT, Shastri SG, Kumar RBN, Chinnakali P, Nirgude AS, Rangaraju C, Somashekar N, Kumar AMV. To start or to complete? - Challenges in implementing tuberculosis preventive therapy among people living with HIV: a mixed-methods study from Karnataka, India. Glob Health Action 2020; 13:1704540. [PMID: 31937200 PMCID: PMC7006687 DOI: 10.1080/16549716.2019.1704540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/05/2019] [Indexed: 01/06/2023] Open
Abstract
Background: Isoniazid preventive therapy (IPT) has been shown to reduce the risk of tuberculosis (TB) among people living with HIV (PLHIV). In 2017, India began a nationwide roll-out of IPT, but there is a lack of evidence on the implementation and the challenges.Objectives: Among PLHIV newly initiated on antiretroviral therapy (ART) from January 2017 to June 2018, to: (i) assess the proportion who started and completed IPT and (ii) explore reasons for non-initiation and non-completion from health-care providers' and patients' perspectives.Methods: An explanatory mixed-methods study was conducted in two selected districts of Karnataka, South India. A quantitative phase (cohort analysis of routinely collected program data) was followed by a qualitative phase involving thematic analysis of in-depth interviews with providers (n = 22) and patients (n = 8).Results: Of the 4020 included PLHIV, 3780 (94%) were eligible for IPT, of whom, 1496 (40%, 95% CI: 38%-41%) were initiated on IPT. Among those initiated, 423 (28.3%) were still on IPT at the time of analysis. Among 1073 patients with declared IPT outcomes 870 (81%, 95% CI: 79%-83%) had completed the six-month course of IPT. The main reason for IPT non-initiation and non-completion was frequent drug stock-outs. This required health-care providers to restrict IPT initiation in selected patient subgroups and earmark six-monthly courses for each patient to ensure that, once started, treatment was not interrupted. The other reasons for non-completion were adverse drug effects and loss to follow-up.Conclusion: The combined picture of 'low IPT initiation and high completion' seen in our study mirrors findings from other countries. Drug stock-out was the key challenge, which obliged health-care providers to prioritize 'IPT completion' over 'IPT initiation'. There is an urgent need to improve the procurement and supply chain management of isoniazid.
Collapse
Affiliation(s)
- Mahendra M. Reddy
- Department of Community Medicine, Sri Devaraj Urs Medical College (SDUMC), Sri Devaraj Urs Academy of Higher Education and Research (SDUAHER), Kolar, India
| | - Pruthu Thekkur
- Centre for Operational Research, International Union Against Tuberculosis and Lung Disease (The Union), Paris, France
- Centre for Operational Research, The Union South-East Asia Office, New Delhi, India
| | - Nagesh Ramya
- Department of Community Medicine, Sri Devaraj Urs Medical College (SDUMC), Sri Devaraj Urs Academy of Higher Education and Research (SDUAHER), Kolar, India
| | - Prasanna B. T. Kamath
- Department of Community Medicine, Sri Devaraj Urs Medical College (SDUMC), Sri Devaraj Urs Academy of Higher Education and Research (SDUAHER), Kolar, India
| | - Suresh G. Shastri
- Department of Health and Family Welfare Services, State Tuberculosis Cell, Bengaluru, India
| | - Ravi B. N. Kumar
- Department of Health and Family Welfare Services, National AIDS Control Organization (NACO), New Delhi, India
- Department of Health and Family Welfare Services, Karnataka AIDS Prevention Society (KSAPS), Bengaluru, India
| | - Palanivel Chinnakali
- Department of Preventive and Social Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Abhay S. Nirgude
- Department of Community Medicine, Yenepoya Medical College, Yenepoya (Deemed to be University), Mangaluru, India
| | | | | | - Ajay M. V. Kumar
- Centre for Operational Research, International Union Against Tuberculosis and Lung Disease (The Union), Paris, France
- Centre for Operational Research, The Union South-East Asia Office, New Delhi, India
- Department of Community Medicine, Yenepoya Medical College, Yenepoya (Deemed to be University), Mangaluru, India
| |
Collapse
|
79
|
Adherence to tuberculosis preventive therapy measured by urine metabolite testing among people with HIV. AIDS 2020; 34:63-71. [PMID: 31567163 DOI: 10.1097/qad.0000000000002380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Tuberculosis preventive therapy for people living with HIV is effective, widely recommended, and increasingly prescribed, but completion rates are less than ideal, and adherence is not typically monitored. We sought to quantify adherence to isoniazid preventive therapy using a urine metabolite assay. DESIGN Two cross-sectional surveys. SETTING Rio de Janeiro, Brazil, 2008-2009; and Northwest Province, South Africa, 2018-2019. PARTICIPANTS Two hundred and three Brazilian and 93 South African patients attending HIV clinics with active prescriptions for isoniazid preventive therapy MAIN OUTCOME MEASURES:: Self-reported isoniazid adherence, paired with semiquantitative measurement of urine isoniazid metabolites. RESULTS By self-report, 90% of patients [95% confidence interval (CI) 86-93%] reported having taken a dose of isoniazid on the day of enrollment or the preceding day, and 91% (95% CI 87-94%) reported missing an average of one dose or fewer per week. By urine testing, only 65% (95% CI 59-70%) of all patients, and 69% (95% CI 63-74%) of those who reported having taken isoniazid on the current or preceding day, had detectable urine metabolites (expected in 95% of patients at 24 h). Longer time since starting preventive therapy was independently associated with a negative urine test for isoniazid metabolites (adjusted prevalence ratio 1.11 per month of isoniazid, 95% CI 1.05-1.18). CONCLUSION Adherence to isoniazid preventive therapy among patients with HIV in Brazil and South Africa is inadequate, is overestimated by self-report, and declines with time on treatment. Shorter regimens for TB preventive therapy may improve adherence and completion, but adherence support for all patients may be necessary.
Collapse
|
80
|
Chaisson LH, Saraceni V, Cohn S, Seabrook D, Cavalcante SC, Chaisson RE, Golub JE, Durovni B. CD4+ cell count stratification to guide tuberculosis preventive therapy for people living with HIV. AIDS 2020; 34:139-147. [PMID: 31634189 PMCID: PMC7112158 DOI: 10.1097/qad.0000000000002398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVES In 2018, Brazilian guidelines changed to recommend tuberculosis (TB) preventive therapy for all people with HIV and a CD4 cell count 350 cells/μl or less, but only for those with a positive tuberculin skin test (TST) if CD4 cell count is than 350 cells/μl. We determined the potential effectiveness of CD4-based guidelines for TB testing and preventive therapy. DESIGN Secondary analysis of the stepped-wedge, cluster-randomized THRio trial for isoniazid preventive therapy (IPT). METHODS We analyzed data from 4114 newly registered patients with HIV in 29 clinics followed until TB diagnosis, death, or administrative censoring. We compared incidence rates of TB and TB/death between CD4, TST, IPT, and antiretroviral therapy categories. RESULTS Initial CD4 cell count was 350 cells/μl or less in 2138 (52%) and more than 350 cells/μl in 1976 (48%) patients. TST was performed for 2922 (71%), of whom 657 (16%) were TST-positive [278 (13%) CD4 ≤ 350 vs. 379 (19%) CD4 > 350]. A total of 619 (15%) received IPT and 2806 (68%) received antiretroviral therapy. For patients with CD4 cell count 350 cells/μl or less who did not receive IPT, the incidence rate of TB was 1.79/100 person-years (pys) and TB/death was 3.89/100 pys. For patients with CD4 cell count more than 350 who did not receive IPT, the incidence rates of TB and TB/death were 0.57/100 and 1.49/100 pys for TST-negatives, and 1.05/100 and 1.64/100 pys for TST-unknowns. CONCLUSION TB incidence was high among all patients who did not receive IPT, including those with CD4 cell count more than 350 cells/μl and negative or unknown TST results. TB preventive therapy should be provided to all people living with HIV in medium burden settings, regardless of CD4 cell count and TST status.
Collapse
Affiliation(s)
- Lelia H Chaisson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA Municipal Health Secretariat, Rio de Janeiro, Brazil Department of Medicine, Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland Linksbridge SPC, Seattle, Washington, USA Instituto Nacional de Infectologia Evandro Chagas Centro de Estudos Estratégicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
81
|
González Fernández L, Casas EC, Singh S, Churchyard GJ, Brigden G, Gotuzzo E, Vandevelde W, Sahu S, Ahmedov S, Kamarulzaman A, Ponce‐de‐León A, Grinsztejn B, Swindells S. New opportunities in tuberculosis prevention: implications for people living with HIV. J Int AIDS Soc 2020; 23:e25438. [PMID: 31913556 PMCID: PMC6947976 DOI: 10.1002/jia2.25438] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/27/2019] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Tuberculosis (TB) is a leading cause of mortality among people living with HIV (PLHIV). An invigorated global END TB Strategy seeks to increase efforts in scaling up TB preventive therapy (TPT) as a central intervention for HIV programmes in an effort to contribute to a 90% reduction in TB incidence and 95% reduction in mortality by 2035. TPT in PLHIV should be part of a comprehensive approach to reduce TB transmission, illness and death that also includes TB active case-finding and prompt, effective and timely initiation of anti-TB therapy among PLHIV. However, the use and implementation of preventive strategies has remained deplorably inadequate and today TB prevention among PLHIV has become an urgent priority globally. DISCUSSION We present a summary of the current and novel TPT regimens, including current evidence of use with antiretroviral regimens (ART). We review challenges and opportunities to scale-up TB prevention within HIV programmes, including the use of differentiated care approaches and demand creation for effective TB/HIV services delivery. TB preventive vaccines and diagnostics, including optimal algorithms, while important topics, are outside of the focus of this commentary. CONCLUSIONS A number of new tools and strategies to make TPT a standard of care in HIV programmes have become available. The new TPT regimens are safe and effective and can be used with current ART, with attention being paid to potential drug-drug interactions between rifamycins and some classes of antiretrovirals. More research and development is needed to optimize TPT for small children, pregnant women and drug-resistant TB (DR-TB). Effective programmatic scale-up can be supported through context-adapted demand creation strategies and the inclusion of TPT in client-centred services, such as differentiated service delivery (DSD) models. Robust collaboration between the HIV and TB programmes represents a unique opportunity to ensure that TB, a preventable and curable condition, is no longer the number one cause of death in PLHIV.
Collapse
Affiliation(s)
| | - Esther C Casas
- Southern Africa Medical UnitMédecins Sans FrontièresCape TownSouth Africa
| | | | - Gavin J Churchyard
- Aurum InstituteParktownSouth Africa
- School of Public HealthUniversity of WitwatersrandJohannesburgSouth Africa
- Advancing Care and Treatment for TB/HIVSouth African Medical Research CouncilParktownSouth Africa
| | - Grania Brigden
- Department of TuberculosisInternational Union Against Tuberculosis and Lung DiseaseGenevaSwitzerland
| | - Eduardo Gotuzzo
- Department of Medicine and Director of the “Alexander von Humboldt” Institute of Tropical Medicine and Infectious DiseasesPeruvian University Cayetano HerediaLimaPeru
| | - Wim Vandevelde
- Global Network of People living with HIV (GNP+)Cape TownSouth Africa
| | | | - Sevim Ahmedov
- Bureau for Global Health, Infectious Diseases, TB DivisionUSAIDWashingtonDCUSA
| | | | - Alfredo Ponce‐de‐León
- Infectious Diseases DepartmentInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | | | | |
Collapse
|
82
|
Salazar-Austin N, Dowdy DW, Chaisson RE, Golub JE. Seventy Years of Tuberculosis Prevention: Efficacy, Effectiveness, Toxicity, Durability, and Duration. Am J Epidemiol 2019; 188:2078-2085. [PMID: 31364692 DOI: 10.1093/aje/kwz172] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 01/12/2023] Open
Abstract
Tuberculosis (TB) has been a leading infectious cause of death worldwide for much of human history, with 1.6 million deaths estimated in 2017. The Department of Epidemiology at the Johns Hopkins Bloomberg School of Public Health has played an important role in understanding and responding to TB, and it has made particularly substantial contributions to prevention of TB with chemoprophylaxis. TB preventive therapy is highly efficacious in the prevention of TB disease, yet it remains underutilized by TB programs worldwide despite strong evidence to support its use in high-risk groups, such as people living with HIV and household contacts, including those under 5 years of age. We review the evidence for TB preventive therapy and discuss the future of TB prevention.
Collapse
|
83
|
Goletti D, Navarra A, Petruccioli E, Cimaglia C, Compagno M, Cuzzi G, De Carli G, Fondaco L, Franzetti F, Giannetti A, Gori A, Lapadula G, Lichtner M, Mastroianni CM, Mazzotta V, Orchi N, Pavone P, Piacentini D, Pirriatore V, Pontali E, Sarmati L, Spolti A, Tacconelli E, Galli M, Antinori A, Calcagno A, Girardi E. Latent tuberculosis infection screening in persons newly-diagnosed with HIV infection in Italy: A multicentre study promoted by the Italian Society of Infectious and Tropical Diseases. Int J Infect Dis 2019; 92:62-68. [PMID: 31887456 DOI: 10.1016/j.ijid.2019.12.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 10/25/2022] Open
Abstract
BACKGROUND The Italian Society of Infectious and Tropical Diseases performed a survey on the application of guidelines for the management of persons living with HIV (PLWH), to evaluate current practice and the yield of screening for latent tuberculosis infection (LTBI) in newly-diagnosed PLWH; in addition, the offer of preventive therapy to LTBI individuals and the completion rate were analysed. MATERIALS AND METHODS Newly-diagnosed PLWH in nine centres were evaluated retrospectively (2016/2017) using binary and multinomial logistic regression to identify factors associated with LTBI diagnostic screening and QuantiFERON (QFT) results. RESULTS Of 801 patients evaluated, 774 were studied after excluding active TB. LTBI tests were performed in 65.5%. Prescription of an LTBI test was associated with being foreign-born (odds ratio (OR) 3.19, p < 0.001), older (for 10-year increments, OR 1.22, p = 0.034), and having a CD4 count <100 cells/mm3 vs ≥500 cells/mm3 (OR 2.30, p = 0.044). LTBI was diagnosed in 6.5% of 495 patients evaluated by QFT. Positive results were associated with being foreign-born (relative risk ratio (RRR) 30.82, p < 0.001), older (for 10-year increments, RRR 1.78, p = 0.003), and having a high CD4 count (for 100 cells/mm3 increments, RRR 1.26, p < 0.003). Sixteen LTBI individuals started TB preventive therapy and eight completed it. CONCLUSIONS LTBI screening is inconsistently performed in newly-diagnosed PLWH. Furthermore, TB preventive therapy is not offered to all LTBI individuals and compliance is poor.
Collapse
Affiliation(s)
- Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases "Lazzaro Spallanzani"- IRCCS, Rome, Italy.
| | - Assunta Navarra
- Clinical Epidemiology Unit, National Institute for Infectious Diseases "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| | - Elisa Petruccioli
- Translational Research Unit, National Institute for Infectious Diseases "Lazzaro Spallanzani"- IRCCS, Rome, Italy
| | - Claudia Cimaglia
- Clinical Epidemiology Unit, National Institute for Infectious Diseases "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| | - Mirko Compagno
- Infectious Diseases Clinic, Università Tor Vergata, Rome, Italy
| | - Gilda Cuzzi
- Translational Research Unit, National Institute for Infectious Diseases "Lazzaro Spallanzani"- IRCCS, Rome, Italy
| | - Gabriella De Carli
- AIDS Reference Centre, National Institute for Infectious Diseases "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| | - Laura Fondaco
- Department of Public Health and Infectious Diseases, Sapienza University, S.M. Goretti Hospital, Latina, Italy
| | - Fabio Franzetti
- Infectious Disease Unit, Department of Biomedical and Clinical Sciences L. Sacco, University of Milano, Italy Infectious Diseases Unit, Ospedale di Busto Arsizio, Varese, Italy
| | - Alberto Giannetti
- Clinical Division of HIV/AIDS, National Institute for Infectious Diseases "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| | - Andrea Gori
- Infectious Diseases, University of Milan, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Miriam Lichtner
- Department of Public Health and Infectious Diseases, Sapienza University, S.M. Goretti Hospital, Latina, Italy
| | - Claudio M Mastroianni
- Department of Public Health and Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico Umberto I, Sapienza University, Rome, Italy
| | - Valentina Mazzotta
- Clinical Division of HIV/AIDS, National Institute for Infectious Diseases "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| | - Nicoletta Orchi
- AIDS Reference Centre, National Institute for Infectious Diseases "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| | - Paolo Pavone
- Department of Public Health and Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico Umberto I, Sapienza University, Rome, Italy
| | - Daniela Piacentini
- Infectious Diseases Department, University Hospital of Verona, Verona, Italy
| | - Veronica Pirriatore
- Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, Turin, Italy
| | - Emanuele Pontali
- Department of Infectious Diseases, Galliera Hospital, Genova, Italy
| | | | - Anna Spolti
- Infectious Diseases Unit, Ospedale San Gerardo, Monza, Italy
| | - Evelina Tacconelli
- Infectious Diseases Department, University Hospital of Verona, Verona, Italy
| | - Massimo Galli
- Infectious Disease Unit, Department of Biomedical and Clinical Sciences L. Sacco, University of Milano, Milan, Italy
| | - Andrea Antinori
- Clinical Division of HIV/AIDS, National Institute for Infectious Diseases "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| | - Andrea Calcagno
- Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, Turin, Italy
| | - Enrico Girardi
- Clinical Epidemiology Unit, National Institute for Infectious Diseases "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| |
Collapse
|
84
|
High incidence of tuberculosis in the first year of antiretroviral therapy in the Botswana National antiretroviral therapy programme between 2011 and 2015. AIDS 2019; 33:2415-2422. [PMID: 31764106 DOI: 10.1097/qad.0000000000002363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Tuberculosis (TB) remains one of the leading causes of mortality and morbidity among people living with HIV. We sought to estimate the incidence of TB in a national database of HIV-infected patients receiving antiretroviral therapy (ART) in Botswana. DESIGN A retrospective analysis of HIV-infected adult patients (≥18years) who initiated ART between 2011 and 2015 in the Botswana ART program. METHODS Multivariable analysis using Cox regression included sex, age, viral load and CD4 T-cell counts. RESULTS Of 45 729 patients, with a median follow-up of 1.7 years Q1 : Q3, 0.5, 3.1), 1791 patients developed TB over a median of 1.5 years (Q1 : Q3, 0.3, 3.1) of follow-up (incidence rate 1.9 per 100 person-years; 95% CI 1.8-2.0). At baseline, the median CD4 T-cell count was 272 cells/μl (Q1, Q3 146, 403). The risk of TB was greatest within the first year of ART (incidence rate 2.9 per 100 person-years; 95% CI 2.7-3.1) and in patients with CD4 T-cell counts below 50 cells/μl (incidence rate 8.3/100 person-years; 95% CI 7.1-9.7). Patients with viral loads above 10 000 copies/ml at 3 months post-ART initiation had two times higher risk of TB, hazard ratio 2.5 (95% CI 1.8-2.3). CONCLUSION We report a high incidence of TB within the first year of ART and in patients with advanced immunodeficiency. Improved screening strategies and virologic monitoring during this early period on ART, coupled with TB preventive treatment, will reduce the burden of TB.
Collapse
|
85
|
Harries AD, Kumar AMV, Satyanarayana S, Takarinda KC, Timire C, Dlodlo RA. Treatment for latent tuberculosis infection in low- and middle-income countries: progress and challenges with implementation and scale-up. Expert Rev Respir Med 2019; 14:195-208. [PMID: 31760848 DOI: 10.1080/17476348.2020.1694907] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Treatment of latent tuberculosis infection (LTBI) is a crucial but neglected component of global tuberculosis control. The 2018 United Nations High-Level Meeting committed world leaders to provide LTBI treatment to at least 30 million people, including 4 million children<5 years, 20 million other household contacts and 6 million HIV-infected people by 2022.Areas covered: This review searched MEDLINE between 1990 and 2019 and discussed: i) high-risk groups to be prioritized for diagnosis and treatment of LTBI; ii) challenges with diagnosing LTBI in programmatic settings; iii) LTBI treatment options including isoniazid monotherapy, shorter regimens (rifampicin-monotherapy, rifampicin-isoniazid and rifapentine-isoniazid) and treatments for contacts of drug-resistant patients; iv) issues with programmatic scale-up of treatment including policy considerations, ruling out active TB, time to start treatment, safety, uninterrupted drug supplies and treatment adherence; and v) recording and reporting.Expert opinion: In 2017, <1.5 million persons were reported to be treated for LTBI. This must rapidly increase to 6 million persons annually. If HIV programs focus on HIV-infected people already accessing or about to start antiretroviral therapy and TB programs focus on household contacts, these targets could be achieved. Isoniazid remains the current treatment of choice although shorter courses of rifapentine-isoniazid are possible alternatives.
Collapse
Affiliation(s)
- Anthony D Harries
- The Centre for Operational Research, International Union Against Tuberculosis and Lung Disease, Paris, France.,Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Ajay M V Kumar
- The Centre for Operational Research, International Union Against Tuberculosis and Lung Disease, Paris, France.,South-East Asia Office, International Union Against Tuberculosis and Lung Disease, New Delhi, India.,Yenepoya Medical College, Yenepoya (Deemed to be University), Mangalore, India
| | - Srinath Satyanarayana
- The Centre for Operational Research, International Union Against Tuberculosis and Lung Disease, Paris, France.,South-East Asia Office, International Union Against Tuberculosis and Lung Disease, New Delhi, India
| | - Kudakwashe C Takarinda
- The Centre for Operational Research, International Union Against Tuberculosis and Lung Disease, Paris, France.,AIDS and TB Department, Ministry of Health and Child Care, Harare, Zimbabwe
| | - Collins Timire
- The Centre for Operational Research, International Union Against Tuberculosis and Lung Disease, Paris, France.,AIDS and TB Department, Ministry of Health and Child Care, Harare, Zimbabwe
| | - Riitta A Dlodlo
- TB Department, International Union Against Tuberculosis and Lung Disease, Paris, France
| |
Collapse
|
86
|
MacPherson P, Webb EL, Variava E, Lala SG, Milovanovic M, Ratsela A, Lebina L, Kinghorn A, Martinson NA. Intensified household contact tracing, prevention and treatment support versus enhanced standard of care for contacts of tuberculosis cases in South Africa: study protocol for a household cluster-randomised trial. BMC Infect Dis 2019; 19:839. [PMID: 31606032 PMCID: PMC6790042 DOI: 10.1186/s12879-019-4502-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 09/24/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Household contact tracing of index TB cases has been advocated as a key part of TB control for many years, but has not been widely implemented in many low-resource setting because of the current dearth of high quality evidence for effectiveness. Innovative strategies for earlier, more effective treatment are particularly important in contexts with hyper-endemic levels of HIV, where levels of TB infection remain extremely high. METHODS We present the design of a household cluster-randomised controlled trial of interventions aimed at improving TB-free survival and reducing childhood prevalence of Mycobacterium tuberculosis infection among household contacts of index TB cases diagnosed in two provinces of South Africa. Households of index TB cases will be randomly allocated in a 1:1 ratio to receive either an intensified home screening and linkage for TB and HIV intervention, or enhanced standard of care. The primary outcome will compare between groups the TB-free survival of household contacts over 15 months. All participants, or their next-of-kin, will provide written informed consent to participate. DISCUSSION Evidence from randomised trials is required to identify cost-effective approaches to TB case-finding that can be applied at scale in sub-Saharan Africa. TRIAL REGISTRATION ISRCTN16006202 (01/02/2017: retrospectively registered) and NHREC4399 (11/04/2016: prospectively registered). Protocol version: 4.0 (date: 18th January 2018).
Collapse
Affiliation(s)
- Peter MacPherson
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Queen Elizabeth Central Hospital, Blantyre, Malawi. .,Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Emily L Webb
- MRC Tropical Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Ebrahim Variava
- Department of Internal Medicine, Klerksdorp Tshepong Hospital Complex and University of the Witwatersrand, Klerksdorp, South Africa.,Perinatal HIV Research Unit (PHRU), SA MRC Soweto Matlosana Collaborating Centre for HIV/AIDS and TB, University of the Witwatersrand, Soweto, South Africa
| | - Sanjay G Lala
- Perinatal HIV Research Unit (PHRU), SA MRC Soweto Matlosana Collaborating Centre for HIV/AIDS and TB, University of the Witwatersrand, Soweto, South Africa.,Department of Paediatrics, Chris Hani Baragwanath Academic Hospital, and University of the Witwatersrand, Soweto, South Africa
| | - Minja Milovanovic
- Perinatal HIV Research Unit (PHRU), SA MRC Soweto Matlosana Collaborating Centre for HIV/AIDS and TB, University of the Witwatersrand, Soweto, South Africa
| | - Andrew Ratsela
- Department of Medicine, Polokwane and Mankweng Hospitals, University of Limpopo, Polokwane, South Africa
| | - Limakatso Lebina
- Perinatal HIV Research Unit (PHRU), SA MRC Soweto Matlosana Collaborating Centre for HIV/AIDS and TB, University of the Witwatersrand, Soweto, South Africa
| | - Anthony Kinghorn
- Perinatal HIV Research Unit (PHRU), SA MRC Soweto Matlosana Collaborating Centre for HIV/AIDS and TB, University of the Witwatersrand, Soweto, South Africa
| | - Neil A Martinson
- Department of Internal Medicine, Klerksdorp Tshepong Hospital Complex and University of the Witwatersrand, Klerksdorp, South Africa.,Perinatal HIV Research Unit (PHRU), SA MRC Soweto Matlosana Collaborating Centre for HIV/AIDS and TB, University of the Witwatersrand, Soweto, South Africa
| |
Collapse
|
87
|
Nyathi S, Dlodlo RA, Satyanarayana S, Takarinda KC, Tweya H, Hove S, Matambo R, Mandewo W, Nyathi K, Sibanda E, Harries AD. Isoniazid preventive therapy: Uptake, incidence of tuberculosis and survival among people living with HIV in Bulawayo, Zimbabwe. PLoS One 2019; 14:e0223076. [PMID: 31581271 PMCID: PMC6776297 DOI: 10.1371/journal.pone.0223076] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 09/12/2019] [Indexed: 01/19/2023] Open
Abstract
SETTING Four primary health care clinics providing tuberculosis (TB) and Human Immunodeficiency Virus care services in Bulawayo, Zimbabwe. OBJECTIVES To assess isoniazid preventive therapy (IPT) initiation and completion, factors associated with IPT uptake and incidence of TB, and TB and antiretroviral treatment (ART) outcomes among people living with HIV (PLHIV). DESIGN This was a cohort study using routine data in the records for PLHIV initiated on ART from October 2013 to March 2014 with 31 December 2017 as the end of the follow-up period. RESULTS A total of 408 PLHIV were eligible for IPT, 214 (52%) were initiated on IPT and 201 (94%) completed IPT. No person in the IPT-initiated group developed Tuberculosis (TB). Six persons with TB were reported among the non-IPT-initiated group leading to an incidence of 9 cases/1,000 person-years of follow-up. About 70% of those who developed and were treated for TB had a successful TB treatment outcome. The survival on ART at four years of follow-up was 88% among the IPT-initiated PLHIV that was significantly higher than the 75% survival in the group not- initiated on IPT. CONCLUSION The study revealed low IPT initiation among eligible PLHIV who, if started on IPT, completed the six month regimen. TB was reported only among the PLHIV not-initiated on IPT and the four year ART survival was higher in the IPT-initiated group than in the non-initiated group. These findings reinforce the need to strengthen IPT uptake among PLHIV in Bulawayo.
Collapse
Affiliation(s)
- Saziso Nyathi
- Health Services Department, City of Bulawayo, Bulawayo, Zimbabwe
| | - Riitta A. Dlodlo
- International Union Against Tuberculosis and Lung Disease (The Union), Paris, France
- International Union Against Tuberculosis and Lung Disease (The Union), Harare, Zimbabwe
| | - Srinath Satyanarayana
- International Union Against Tuberculosis and Lung Disease (The Union), Paris, France
| | - Kudakwashe C. Takarinda
- International Union Against Tuberculosis and Lung Disease (The Union), Paris, France
- International Union Against Tuberculosis and Lung Disease (The Union), Harare, Zimbabwe
- AIDS and TB Directorate, Ministry of Health and Child Care, Harare, Zimbabwe
| | - Hannock Tweya
- Light House Trust, Lilongwe, Malawi
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, England, United Kingdom
| | - Sithokozile Hove
- Health Services Department, City of Bulawayo, Bulawayo, Zimbabwe
| | - Ronnie Matambo
- International Union Against Tuberculosis and Lung Disease (The Union), Harare, Zimbabwe
| | - Winnie Mandewo
- Elizabeth Glaser Peadiatric AIDS Foundation, Harare, Zimbabwe
| | - Khulamuzi Nyathi
- Health Services Department, City of Bulawayo, Bulawayo, Zimbabwe
| | - Edwin Sibanda
- Health Services Department, City of Bulawayo, Bulawayo, Zimbabwe
| | - Anthony D. Harries
- International Union Against Tuberculosis and Lung Disease (The Union), Paris, France
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, England, United Kingdom
| |
Collapse
|
88
|
Gupta A, Montepiedra G, Aaron L, Theron G, McCarthy K, Bradford S, Chipato T, Vhembo T, Stranix-Chibanda L, Onyango-Makumbi C, Masheto GR, Violari A, Mmbaga BT, Aurpibul L, Bhosale R, Mave V, Rouzier V, Hesseling A, Shin K, Zimmer B, Costello D, Sterling TR, Chakhtoura N, Jean-Philippe P, Weinberg A. Isoniazid Preventive Therapy in HIV-Infected Pregnant and Postpartum Women. N Engl J Med 2019; 381:1333-1346. [PMID: 31577875 PMCID: PMC7051859 DOI: 10.1056/nejmoa1813060] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The safety, efficacy, and appropriate timing of isoniazid therapy to prevent tuberculosis in pregnant women with human immunodeficiency virus (HIV) infection who are receiving antiretroviral therapy are unknown. METHODS In this multicenter, double-blind, placebo-controlled, noninferiority trial, we randomly assigned pregnant women with HIV infection to receive isoniazid preventive therapy for 28 weeks, initiated either during pregnancy (immediate group) or at week 12 after delivery (deferred group). Mothers and infants were followed through week 48 after delivery. The primary outcome was a composite of treatment-related maternal adverse events of grade 3 or higher or permanent discontinuation of the trial regimen because of toxic effects. The noninferiority margin was an upper boundary of the 95% confidence interval for the between-group difference in the rate of the primary outcome of less than 5 events per 100 person-years. RESULTS A total of 956 women were enrolled. A primary outcome event occurred in 72 of 477 women (15.1%) in the immediate group and in 73 of 479 (15.2%) in the deferred group (incidence rate, 15.03 and 14.93 events per 100 person-years, respectively; rate difference, 0.10; 95% confidence interval [CI], -4.77 to 4.98, which met the criterion for noninferiority). Two women in the immediate group and 4 women in the deferred group died (incidence rate, 0.40 and 0.78 per 100 person-years, respectively; rate difference, -0.39; 95% CI, -1.33 to 0.56); all deaths occurred during the postpartum period, and 4 were from liver failure (2 of the women who died from liver failure had received isoniazid [1 in each group]). Tuberculosis developed in 6 women (3 in each group); the incidence rate was 0.60 per 100 person-years in the immediate group and 0.59 per 100 person-years in the deferred group (rate difference, 0.01; 95% CI, -0.94 to 0.96). There was a higher incidence in the immediate group than in the deferred group of an event included in the composite adverse pregnancy outcome (stillbirth or spontaneous abortion, low birth weight in an infant, preterm delivery, or congenital anomalies in an infant) (23.6% vs. 17.0%; difference, 6.7 percentage points; 95% CI, 0.8 to 11.9). CONCLUSIONS The risks associated with initiation of isoniazid preventive therapy during pregnancy appeared to be greater than those associated with initiation of therapy during the postpartum period. (Funded by the National Institutes of Health; IMPAACT P1078 TB APPRISE ClinicalTrials.gov number, NCT01494038.).
Collapse
Affiliation(s)
- Amita Gupta
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Grace Montepiedra
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Lisa Aaron
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Gerhard Theron
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Katie McCarthy
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Sarah Bradford
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Tsungai Chipato
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Tichaona Vhembo
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Lynda Stranix-Chibanda
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Carolyne Onyango-Makumbi
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Gaerolwe R Masheto
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Avy Violari
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Blandina T Mmbaga
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Linda Aurpibul
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Ramesh Bhosale
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Vidya Mave
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Vanessa Rouzier
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Anneke Hesseling
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Katherine Shin
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Bonnie Zimmer
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Diane Costello
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Timothy R Sterling
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Nahida Chakhtoura
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Patrick Jean-Philippe
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Adriana Weinberg
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| |
Collapse
|
89
|
Miyahara R, Piyaworawong S, Prachamat P, Wongyai J, Bupachat S, Yamada N, Summanapan S, Yanai H, Mahasirimongkol S. High tuberculosis burden among HIV-infected populations in Thailand due to a low-sensitivity tuberculin skin test. J Infect Public Health 2019; 13:657-660. [PMID: 31563472 DOI: 10.1016/j.jiph.2019.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 08/01/2019] [Accepted: 08/19/2019] [Indexed: 10/25/2022] Open
Abstract
The current Thai guideline recommends that among people living with HIV, isoniazid preventive therapy (IPT) should be given to those with a positive tuberculin skin test (TST). We conducted a case-control study, nested within a cohort study, in Chiang Rai Province in Thailand to determine the role of TST in predicting the development of active tuberculosis (TB) within the following 2 years. Comparison between participants with CD4+ counts <50cells/mm3 to those with CD4+ ≥200cells/mm3 revealed that TST results were less sensitive (7.7% vs 50.0%) and had a lower negative predictive value (73.1% vs 97.3%) in those with a CD4+ count <50cells/mm3. In people with HIV, using a positive TST result as a criterion for initiating IPT inadvertently decreases the benefits of IPT, especially among those with low CD4+ counts.
Collapse
Affiliation(s)
- Reiko Miyahara
- Genome Medical Science Project, National Center for Global Health and Medicine, Tokyo, Japan; Department of Human Genetics, The University of Tokyo, Tokyo, Japan.
| | | | | | | | | | - Norio Yamada
- Research Institute of Tuberculosis (RIT), Japan Anti-Tuberculosis Association (JATA), Tokyo, Japan
| | | | | | | |
Collapse
|
90
|
Takarinda KC, Choto R, Sandy C, Apollo T, Duri C, Dube F, Mpofu A, Timire C, Mugurungi O, Makaza V, Tapera R, Harries AD. How well does the process of screening and diagnosis work for HIV-infected persons identified with presumptive tuberculosis who are attending HIV care and treatment clinics in Harare city, Zimbabwe? Trans R Soc Trop Med Hyg 2019; 112:450-457. [PMID: 30032237 DOI: 10.1093/trstmh/try073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/26/2018] [Indexed: 01/30/2023] Open
Abstract
Background Intensified TB case finding is recommended for all HIV-infected persons regularly attending HIV care and treatment clinics. The authors aimed to determine how well this system worked among HIV-infected patients diagnosed with presumptive TB in 14 health facilities of Harare province, Zimbabwe, between January and December 2016. Methods Retrospective review using routine programme records. Results Of 47 659 HIV-infected persons enrolled in HIV care, 102 were identified with presumptive TB through the programmatic electronic database. Of these, 23% (23/102) were recorded in presumptive TB registers and, of these 65% (15/23) were traced to laboratory registers. Of 79 patients not recorded in presumptive TB registers, 9% (7/79) were traced to laboratory registers. Of 22 patients in the laboratory register, all had negative sputum smears for acid-fast bacilli and 45% (10/22) had Xpert MTB/RIF assays with one positive result. Six patients altogether started anti-tuberculosis treatment, the median time from presumptive tuberculosis diagnosis to treatment being 12 days. The only significant risk factor for loss-to-follow-up between presumptive TB diagnosis and laboratory registration was not being recorded in presumptive TB registers. Conclusions Follow-up mechanisms for presumptive TB cases diagnosed in HIV care clinics in Harare city need strengthening, particularly through improved documentation in presumptive TB registers and better Xpert MTB/RIF use.
Collapse
Affiliation(s)
- Kudakwashe C Takarinda
- Ministry of Health and Child Care, AIDS and TB Department, Harare, Zimbabwe.,International Union Against Tuberculosis and Lung Disease, Paris, France
| | - Regis Choto
- Ministry of Health and Child Care, AIDS and TB Department, Harare, Zimbabwe
| | - Charles Sandy
- Ministry of Health and Child Care, AIDS and TB Department, Harare, Zimbabwe
| | - Tsitsi Apollo
- Ministry of Health and Child Care, AIDS and TB Department, Harare, Zimbabwe
| | | | | | | | - Collins Timire
- Ministry of Health and Child Care, AIDS and TB Department, Harare, Zimbabwe.,International Union Against Tuberculosis and Lung Disease, Paris, France
| | - Owen Mugurungi
- Ministry of Health and Child Care, AIDS and TB Department, Harare, Zimbabwe
| | | | - Roy Tapera
- University of Botswana, Gaborone, Botswana
| | - Anthony D Harries
- International Union Against Tuberculosis and Lung Disease, Paris, France.,London School of Tropical Hygiene & Medicine, London, United Kingdom
| |
Collapse
|
91
|
Kim HY, Hanrahan CF, Martinson N, Golub JE, Dowdy DW. Cost-effectiveness of universal isoniazid preventive therapy among HIV-infected pregnant women in South Africa. Int J Tuberc Lung Dis 2019; 22:1435-1442. [PMID: 30606315 DOI: 10.5588/ijtld.18.0370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE To estimate the incremental cost-effectiveness of universal vs. test-directed treatment of latent tuberculous infection (LTBI) among human immunodeficiency virus (HIV) positive pregnant women in South Africa. METHODS We compared tuberculin skin test (TST) directed isoniazid preventive therapy (IPT) (TST placement with delivery of IPT to women with positive results) against QuantiFERON®-TB Gold In-Tube (QGIT) directed IPT and universal IPT using decision analysis. Costs were measured empirically in six primary care public health clinics in Matlosana, South Africa. The primary outcome was the incremental cost-effectiveness ratio, expressed in 2016 US$ per disability-adjusted life-year (DALY) averted. RESULTS We estimated that 29.2 of every 1000 pregnant women would develop TB over the course of 1 year in the absence of IPT. TST-directed IPT reduced this number to 24.5 vs. 22.6 with QGIT-directed IPT and 21.0 with universal IPT. Universal IPT was estimated to cost $640/DALY averted (95% uncertainty range $44-$3146) relative to TST-directed IPT and was less costly and more effective (i.e., dominant) than QGIT-directed IPT. Cost-effectiveness was most sensitive to the probability of developing TB and LTBI prevalence. CONCLUSION Providing IPT to all eligible women can be a cost-effective strategy to prevent TB among HIV-positive pregnant women in South Africa.
Collapse
Affiliation(s)
- H-Y Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA, Africa Health Research Institute, Durban, School of Nursing & Public Health, University of KwaZulu-Natal, Durban
| | - C F Hanrahan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - N Martinson
- Perinatal HIV Research Unit, University of Witwatersrand, Johannesburg, South Africa, Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland
| | - J E Golub
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA, Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - D W Dowdy
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA, Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
92
|
Stout JE, Sterling TR, Horsburgh CR. One Month of Rifapentine plus Isoniazid to Prevent HIV-Related Tuberculosis. N Engl J Med 2019; 381:e23. [PMID: 31509689 DOI: 10.1056/nejmc1908492] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
93
|
Dravid A, Natarajan K, Medisetty M, Gawali R, Mahajan U, Kulkarni M, Saraf C, Ghanekar C, Kore S, Rathod N, Dravid M. Incidence of tuberculosis among HIV infected individuals on long term antiretroviral therapy in private healthcare sector in Pune, Western India. BMC Infect Dis 2019; 19:714. [PMID: 31409289 PMCID: PMC6692924 DOI: 10.1186/s12879-019-4361-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 08/06/2019] [Indexed: 12/27/2022] Open
Abstract
Background Despite rapid scale up of antiretroviral therapy (ART), Tuberculosis (TB) remains the commonest opportunistic infection and cause of death among HIV infected individuals in resource limited settings like India. Incidence of TB in individuals on ART in private healthcare sector in India is infrequently studied. Methods This retrospective cohort study conducted between 1st March 2009 and 1st March 2017 aimed to evaluate rate of incident TB in individuals initiated on ART at 3 private sector ART clinics in Pune, India. Individuals more than 12 years of age with ART duration of atleast 6 months were included. Patients were classified as having prevalent TB if they had a TB episode within the year prior to ART initiation or if they developed TB within 6 months of starting ART. Individuals who were diagnosed with TB after 6 months of starting ART were classified as incident TB cases. A recurrent episode of TB after treatment completion or cure of prevalent TB was also regarded as incident TB. Patients were classified as definitive TB if Mycobacterium tuberculosis was grown in culture from a biological sample or a positive rapid molecular test. Patients were classified as probable TB if there was radiologic evidence of TB in absence of confirmatory culture or PCR. Results 1904 patients with a median duration of follow up on ART of 57 (IQR = 32.0, 84.0) months were included. Of these, 182 developed incident TB (22% definitive TB, 38% recurrent cases). TB incidence at 6–12 months, 13–24 months, 25–60 months and > 60 months of ART was 24.32, 5.46, 2.54 and 0.75 cases per 100 person years respectively. Current time updated CD4 count < 500 cells/mm3 (p < 0.0001), virologic failure on ART (adjusted Hazard Ratio (aHR): 3.05 (95% CI: 2.094, 4.454), p < 0.0001) and receipt of ART without IPT (aHR: 8.24 (95% CI, 3.358, 20.204), p < 0.0001) were associated with higher risk of incident TB. Conclusion Starting ART early in treatment naïve individuals, prompt detection of virologic failure on ART and providing IPT along with ART will be useful in reducing incident TB. Efforts from private sector are crucial in achieving Sustainable Development Goals set by Government of India and attaining the vision of a TB free India.
Collapse
Affiliation(s)
- Ameet Dravid
- Department of Medicine, Ruby Hall Clinic, Pune, Maharashtra, India. .,Department of Medicine, Poona Hospital and Research Centre, Pune, Maharashtra, India. .,Department of Medicine, Noble hospital, Pune, Maharashtra, India.
| | - Kartik Natarajan
- Department of Medicine, Poona Hospital and Research Centre, Pune, Maharashtra, India
| | | | - Raviraj Gawali
- Department of Medicine, Poona Hospital and Research Centre, Pune, Maharashtra, India
| | - Uma Mahajan
- Department of Biostatistics, Precision Diagnostics and Biosciences, Pune, Maharashtra, India
| | - Milind Kulkarni
- Department of Medicine, Ruby Hall Clinic, Pune, Maharashtra, India
| | - Chinmay Saraf
- Department of Pathology, Precision Diagnostics and Biosciences, Pune, Maharashtra, India
| | - Charuta Ghanekar
- Department of Microbiology, Poona Hospital and Research Centre, Pune, Maharashtra, India
| | - Sachin Kore
- Department of Dermatology, Ashwini Sahakari Rugnalaya and Research Centre, Solapur, Maharashtra, India
| | - Niranjan Rathod
- Department of Medicine, Apex hospital, Kolhapur, Maharashtra, India
| | - Mrudula Dravid
- Department of Microbiology, Infectious Disease Clinic, Dhule, Maharashtra, India
| |
Collapse
|
94
|
Pathmanathan I, Ahmedov S, Pevzner E, Anyalechi G, Modi S, Kirking H, Cavanaugh JS. TB preventive therapy for people living with HIV: key considerations for scale-up in resource-limited settings. Int J Tuberc Lung Dis 2019; 22:596-605. [PMID: 29862942 DOI: 10.5588/ijtld.17.0758] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Tuberculosis (TB) is the leading cause of death for persons living with the human immunodeficiency virus (PLHIV). TB preventive therapy (TPT) works synergistically with, and independently of, antiretroviral therapy to reduce TB morbidity, mortality and incidence among PLHIV. However, although TPT is a crucial and cost-effective component of HIV care for adults and children and has been recommended as an international standard of care for over a decade, it remains highly underutilized. If we are to end the global TB epidemic, we must address the significant reservoir of tuberculous infection, especially in those, such as PLHIV, who are most likely to progress to TB disease. To do so, we must confront the pervasive perception that barriers to TPT scale-up are insurmountable in resource-limited settings. Here we review available evidence to address several commonly stated obstacles to TPT scale-up, including the need for the tuberculin skin test, limited diagnostic capacity to reliably exclude TB disease, concerns about creating drug resistance, suboptimal patient adherence to therapy, inability to monitor for and prevent adverse events, a 'one size fits all' option for TPT regimen and duration, and uncertainty about TPT use in children, adolescents, and pregnant women. We also discuss TPT delivery in the era of differentiated care for PLHIV, how best to tackle advanced planning for drug procurement and supply chain management, and how to create an enabling environment for TPT scale-up success.
Collapse
Affiliation(s)
- I Pathmanathan
- Division of Global HIV and TB, US Centers for Disease Control and Prevention, Atlanta, Georgia
| | - S Ahmedov
- Bureau for Global Health, United States Agency for International Development, Washington, DC
| | - E Pevzner
- Division of Global HIV and TB, US Centers for Disease Control and Prevention, Atlanta, Georgia
| | - G Anyalechi
- Division of Global HIV and TB, US Centers for Disease Control and Prevention, Atlanta, Georgia
| | - S Modi
- Division of Global HIV and TB, US Centers for Disease Control and Prevention, Atlanta, Georgia
| | - H Kirking
- Division of Global HIV and TB, US Centers for Disease Control and Prevention, Atlanta, Georgia
| | - J S Cavanaugh
- Office of the Global AIDS Coordinator, Washington, DC, USA
| |
Collapse
|
95
|
Abstract
PURPOSE OF REVIEW In the past few years, tuberculosis (TB) has overtaken HIV as the infectious disease with the highest global mortality. Successful management of this syndemic will require improved diagnostic tests, shorter preventive therapies, and more effective treatments, particularly in light of drug-resistant TB. RECENT FINDINGS Results from several major studies have been published or presented recently, including the development of a more sensitive rapid, molecular assay for TB; several new symptom-based screening tools; use of a 1-month regimen for TB prevention; the results of early vs. delayed TB preventive therapy for pregnant women; newer drugs and regimens for multidrug-resistant tuberculosis; and pharmacokinetic, safety, and efficacy studies of new HIV drugs in combination with TB treatment. We reviewed each of these topic areas and summarize relevant findings for the management of TB and HIV co-infection. SUMMARY Moving forward, as new treatment regimes for HIV or TB are developed, consideration of the HIV-TB co-infected patient must figure prominently, both when determining the diagnostic tests employed and to assess properly the drug-drug and drug-disease interactions that influence dosing, safety, and response.
Collapse
|
96
|
Abstract
PURPOSE OF REVIEW Tuberculosis (TB) remains the leading cause of death in people living with HIV (PLHIV) despite the achievements in antiretroviral therapy coverage. TB preventive therapy (TPT) has proved efficacy but has been neglected and poorly implemented. We reviewed recent publications and guidelines about TPT in PLHIV. RECENT FINDINGS High-quality studies showed that TPT has a durable effect, over 5 years, preventing TB and all-cause mortality. There is new evidence showing the noninferiority of shorter, rifamycin-based regimens of TPT increasing the options for treatment. Recent studies describing robust implementation in different settings showed promising results for feasibility, tolerance, retention, and cost-effectiveness. New WHO recommendations, unifying previous versions, have been released to guide countries implementation. SUMMARY New evidence support the scale up of TPT for PLHIV globally, further studies are needed to bring more evidence for specific populations, like pregnant women and for drug-drug interactions with antiretroviral agents.
Collapse
|
97
|
Pathmanathan I, Pevzner E, Cavanaugh J, Nelson L. Addressing tuberculosis in differentiated care provision for people living with HIV. Bull World Health Organ 2019; 95:3. [PMID: 28053356 PMCID: PMC5180337 DOI: 10.2471/blt.16.187021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Ishani Pathmanathan
- Division of Global HIV and TB, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, Mailbox E-04, GA, 30329, United States of America (USA)
| | - Eric Pevzner
- Division of Global HIV and TB, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, Mailbox E-04, GA, 30329, United States of America (USA)
| | - Joseph Cavanaugh
- Division of Global HIV and TB, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, Mailbox E-04, GA, 30329, United States of America (USA)
| | - Lisa Nelson
- Office of the Global AIDS Coordinator, Washington, USA
| |
Collapse
|
98
|
Thindwa D, MacPherson P, Choko AT, Khundi M, Sambakunsi R, Ngwira LG, Kalua T, Webb EL, Corbett EL. Completion of isoniazid preventive therapy among human immunodeficiency virus positive adults in urban Malawi. Int J Tuberc Lung Dis 2019; 22:273-279. [PMID: 29471904 PMCID: PMC5824849 DOI: 10.5588/ijtld.17.0370] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
SETTING: Despite worldwide scale-up of human immunodeficiency virus (HIV) care services, relatively few countries have implemented isoniazid preventive therapy (IPT). Among other programmatic concerns, IPT completion tends to be low, especially when not fully integrated into HIV care clinics. OBJECTIVE: To estimate non-completion of 6-month IPT and its predictors among HIV-positive adults aged ⩾16 years. DESIGN: A prospective cohort study nested within a cluster-randomised trial of TB prevention was conducted between February 2012 and June 2014. IPT for 6 months was provided with pyridoxine at study clinics. Non-completion was defined as loss to follow-up (LTFU), death, active/presumptive TB or stopping IPT for any other reason. Random-effects logistic regression was used to determine predictors of non-completion. RESULTS: Of 1284 HIV-positive adults initiated on IPT, 885/1280 (69.1%) were female; the median CD4 count was 337 cells/μl (IQR 199–511); 320 (24.9%) did not complete IPT. After controlling for antiretroviral treatment status, IPT initiation year, age and sex, non-completion of IPT was associated with World Health Organization stage 3/4 (aOR 1.76, 95%CI 1.22–2.55), CD4 count 100–349 cells/μl (aOR 1.93, 95%CI 1.10–3.38) and any reported side effects (aOR 22.00, 95%CI 9.45–46.71). CONCLUSION: Completion of IPT was suboptimal. Interventions to further improve retention should target immunosuppressed HIV-positive adults and address side effects.
Collapse
Affiliation(s)
- D Thindwa
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi, Department of Infectious Disease Epidemiology, Imperial College London, London
| | - P MacPherson
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, Department of Public Health and Policy, University of Liverpool, Liverpool
| | - A T Choko
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi, Infectious Disease Epidemiology Department, London School of Hygiene & Tropical Medicine (LSHTM), London, UK
| | - M Khundi
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - R Sambakunsi
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - L G Ngwira
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - T Kalua
- Department of HIV/AIDS, Ministry of Health, Lilongwe, Malawi
| | - E L Webb
- Department of HIV/AIDS, Ministry of Health, Lilongwe, Malawi
| | - E L Corbett
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi, Clinical Research Department, LSHTM, London, UK
| |
Collapse
|
99
|
Dhungana GP, Thekkur P, Chinnakali P, Bhatta U, Pandey B, Zhang WH. Initiation and completion rates of isoniazid preventive therapy among people living with HIV in Far-Western Region of Nepal: a retrospective cohort study. BMJ Open 2019; 9:e029058. [PMID: 31147370 PMCID: PMC6549711 DOI: 10.1136/bmjopen-2019-029058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/28/2019] [Accepted: 04/30/2019] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES Isoniazid preventive therapy (IPT), for people living with HIV (PLHIV) is the proven and recommended intervention to avert tuberculosis (TB). In 2015, Nepal implemented 6 months of IPT for all PLHIV registered for HIV care in antiretroviral therapy (ART) centres. After programmatic implementation, there has been no systematic assessment of IPT initiation and completion rates among PLHIV. We aimed to assess IPT initiation and completion rates in the Far-Western Region (FWR) of Nepal. DESIGN We conducted a retrospective cohort study using secondary data extracted from registers maintained at ART centres. SETTING All 11 ART centres in the FWR of Nepal. PARTICIPANTS All PLHIV registered for care between January 2016 and December 2017 in 11 ART centres. PRIMARY OUTCOME MEASURES IPT initiation and completion rates were summarised as percentages with 95% CI. Independent association between patient characteristics and non-initiation of IPT was assessed using cluster-adjusted generalised linear model (log binomial regression) and adjusted relative risk (RR) with 95% CI was calculated. RESULT Of the 492 PLHIV included, 477 (97.0%) did not have active TB at registration. Among 477 without active TB, 141 (29.8%, 95% CI 25.7% to 34.1%) had been initiated on IPT and 85 (17.8%) were initiated within 3 months of registration. Of 141 initiated on IPT, 133 (94.3%, 95% CI 89.1% to 97.5%) had completed 6 months of IPT. Being more than 60 years of age (RR-1.3, 95% CI 1.1 to 1.7), migrant worker (RR-1.3, 95% CI 1.1 to 1.4) and not being initiated on ART (RR-1.4, 95% CI 1.1 to 1.8) were significantly associated with IPT initiation. CONCLUSIONS In FWR of Nepal, three out of 10 eligible PLHIV had received IPT. Among those who have received IPT, the completion rate was good. The HIV care programme needs to explore the potential reasons for this low coverage and take context specific corrective action to fix this gap.
Collapse
Affiliation(s)
| | - Pruthu Thekkur
- Centre for Operational Research, International Union Against Tuberculosis and Lung Disease, Paris, France
- Centre for Operational Research, The Union South-East Asia Office, New Delhi, India
| | - Palanivel Chinnakali
- Department of Preventive and Social Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Usha Bhatta
- National Center for AIDS and STD Control, Kathmandu, Nepal
| | - Basudev Pandey
- Sukraraj Tropical and Infectious Disease Hospital, Kathmandu, Nepal
| | - Wei-Hong Zhang
- International Centre for Reproductive Health (ICRH), Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, Gent, Belgium
- Research Laboratory for Human Reproduction, Faculty of Medicine, School of Public Health, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| |
Collapse
|
100
|
The protective effect of isoniazid preventive therapy on tuberculosis incidence among HIV positive patients receiving ART in Ethiopian settings: a meta-analysis. BMC Infect Dis 2019; 19:405. [PMID: 31077133 PMCID: PMC6511123 DOI: 10.1186/s12879-019-4031-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 04/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background Tuberculosis (TB) and HIV makeup a deadly synergy of infectious disease, and the combined effect is apparent in resource limited countries like Ethiopia. Previous studies have demonstrated inconsistent results about the protective effect of isoniazid preventive therapy (IPT) on active TB incidence among HIV positive patients receiving ART. Therefore, the aim of this meta-analysis was, first, to determine the protective effect of IPT on active tuberculosis incidence, and second, to assess the pooled incidence of active TB among HIV positive patients taking ART with and without IPT intervention in Ethiopia. Methods PubMed, Google scholar and Cochran library databases were searched from April 1 to 30, 2018. Two independent authors explored and assessed studies for eligibility, and extracted data based on predefined criteria. Studies that reported TB incidence among HIV positive patients taking ART in Ethiopia with and without IPT concomitant intervention, and with a clear stratified data on the incidence of TB based on the duration of IPT intervention were selected. A random effects model was used to estimate risk ratios and the pooled incident TB with the respective 95% confidence intervals. Results We identified 7 suitable studies in this analysis. Accordingly, IPT reduced the risk of TB incidence by 74%, risk ratio (RR) 0.26 (95% CI; 0.16–0.43%), compared to no IPT group. Moreover, IPT for 12 months reduced incident TB by 91% (RR: 0.09, 95% CI: 0.04 to 0.21), whereas 6 months IPT averted TB incidence by 63% (RR: 0.37, 95% CI: 0.26 to 0.52). The overall pooled incident TB among HIV infected patients receiving ART was 10.30% (95% CI; 7.57–13.02%). Specifically, incident TB among study cohorts with and without IPT was 3.79% (95% CI; 2.03–5.55%) and 16.32% (95% CI; 11.57–21.06%) respectively. Conclusion IPT reduced the risk of incident TB among HIV positive patients receiving ART in Ethiopian settings. Moreover, the duration of IPT intervention has effect on its protective role. Thus, scaling up the isoniazid preventive therapy program and its strict compliance is necessary to avert HIV fueled tuberculosis. Study protocol registration CRD42018090804. Electronic supplementary material The online version of this article (10.1186/s12879-019-4031-2) contains supplementary material, which is available to authorized users.
Collapse
|