1
|
Podany AT, Cramer Y, Imperial M, Rosenkranz SL, Avihingsanon A, Arduino R, Samaneka W, Gelmanova I, Savic R, Swindells S, Dawson R, Luetkemeyer AF. Twice-Daily Dolutegravir-Based Antiretroviral Therapy With 1 Month of Daily Rifapentine and Isoniazid for Tuberculosis Prevention. Clin Infect Dis 2024; 79:983-989. [PMID: 38568956 PMCID: PMC11478809 DOI: 10.1093/cid/ciae183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/16/2024] [Accepted: 03/24/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND One month of daily rifapentine + isoniazid (1HP) is an effective, ultrashort option for tuberculosis prevention in people with human immunodeficiency virus (HIV). However, rifapentine may decrease antiretroviral drug concentrations and increase the risk of virologic failure. AIDS Clinical Trials Group A5372 evaluated the effect of 1HP on the pharmacokinetics of twice-daily dolutegravir. METHODS A5372 was a multicenter, pharmacokinetic study in people with HIV (≥18 years) already on dolutegravir-containing antiretroviral therapy with HIV RNA <50 copies/mL. Participants received daily rifapentine/isoniazid (600 mg/300 mg) for 28 days as part of 1HP. Dolutegravir was increased to 50 mg twice daily during 1HP, and intensive pharmacokinetic sampling was performed on day 0 (before 1HP) and on the final day of 1HP treatment. RESULTS Thirty-two participants (41% female; 66% Black/African; median [Q1, Q3] age, 42 [34, 49] years) were included in the pharmacokinetic analysis; 31 had HIV RNA <50 copies/mL at the end of 1HP dosing. One participant had an HIV RNA of 160 copies/mL at day 28, with HIV RNA <50 copies/mL upon repeat testing on day 42. The median (Q1, Q3) dolutegravir trough concentration was 1751 ng/mL (1195, 2542) on day 0 versus 1987 ng/mL (1331, 2278) on day 28 (day 28:day 0 geometric mean ratio, 1.05 [90% confidence interval, .93-1.2]; P = .43). No serious adverse events were reported. CONCLUSIONS Dolutegravir trough concentrations with 50 mg twice-daily dosing during 1HP treatment were greater than those with standard-dose dolutegravir once daily without 1HP. These pharmacokinetic, virologic, and safety data provide support for twice-daily dolutegravir use in combination with 1HP for tuberculosis prevention. CLINICAL TRIALS REGISTRATION NCT04272242.
Collapse
Affiliation(s)
- Anthony T Podany
- College of Pharmacy, University of Nebraska Medical Center, Omaha
| | - Yoninah Cramer
- Statistical and Data Analysis Center, Harvard School of Public Health, Boston, Massachusetts
| | | | - Susan L Rosenkranz
- Statistical and Data Analysis Center, Harvard School of Public Health, Boston, Massachusetts
| | - Anchalee Avihingsanon
- Thai Red Cross AIDS Research Center and Center of Excellence in Tuberculosis, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Roberto Arduino
- McGovern Medical School, University of Texas Health Science Center at Houston, Texas
| | | | - Irina Gelmanova
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Rada Savic
- College of Pharmacy, University of California, San Francisco
| | - Susan Swindells
- Infectious Diseases, Internal Medicine, University of Nebraska Medical Center, Omaha
| | - Rodney Dawson
- Lung Institute, University of Cape Town, South Africa
| | | |
Collapse
|
2
|
Kadota JL, Musinguzi A, Aschmann HE, Akello L, Welishe F, Nakimuli J, Berger CA, Kiwanuka N, Phillips PP, Katamba A, Dowdy DW, Cattamanchi A, Semitala FC. Adverse events reported during weekly isoniazid-rifapentine (3HP) tuberculosis preventive treatment among people living with HIV in Uganda. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.08.24311632. [PMID: 39148841 PMCID: PMC11326332 DOI: 10.1101/2024.08.08.24311632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background Short-course tuberculosis (TB) prevention regimens, including 12 weeks of isoniazid and rifapentine (3HP), are increasingly used in high TB-burden countries. Despite established safety and tolerability in efficacy trials, 3HP-related adverse events (AE) could differ in routine settings. Real-world data on AE type, frequency, and timing is crucial for health systems considering 3HP programmatic scale-up. Methods We reviewed AEs among people living with HIV (PLHIV) participating in a pragmatic implementation trial of facilitated 3HP taken by directly observed therapy (DOT) or self-administered therapy (SAT) in Kampala, Uganda, and classified them using the Common Terminology Criteria for Adverse Events. We assessed AE timing and summarized related clinical actions including lab tests, diagnoses made, medications prescribed, and treatment interruptions. Results Among 1655 PLHIV treated between July 2020-September 2022, 270 (16.3%) reported 451 events; main issues included general (7%), nervous system (6%), musculoskeletal (5%), gastrointestinal (5%), and dermatologic (3%) disorders. Most (61%) occurred within 6 weeks of initiating 3HP. Among those with events, 211 (78%) required further clinician evaluation, 202 (75%) required laboratory testing, 102 (38%) had medications prescribed, 40 (15%) had treatment paused, and 14 (5%) discontinued 3HP. Women, those multidimensionally impoverished, and DOT recipients were more likely to report an AE. SAT users and later enrollees were more likely to have 3HP interrupted or stopped due to an AE. Conclusions In a routine setting, 3HP was safe with 16% of PLHIV reporting AEs and only 3% requiring temporary or permanent treatment interruption. These findings support 3HP expansion in routine HIV/AIDS care settings for TB prevention.
Collapse
Affiliation(s)
- Jillian L Kadota
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco CA USA
- Center for Tuberculosis, University of California San Francisco, San Francisco CA USA
| | | | - Hélène E Aschmann
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco CA USA
- Center for Tuberculosis, University of California San Francisco, San Francisco CA USA
| | - Lydia Akello
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Fred Welishe
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Jane Nakimuli
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Christopher A Berger
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco CA USA
- Center for Tuberculosis, University of California San Francisco, San Francisco CA USA
| | - Noah Kiwanuka
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Patrick Pj Phillips
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco CA USA
- Center for Tuberculosis, University of California San Francisco, San Francisco CA USA
| | - Achilles Katamba
- Clinical Epidemiology & Biostatistics Unit, Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
- Uganda Tuberculosis Implementation Research Consortium, Kampala, Uganda
| | - David W Dowdy
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Adithya Cattamanchi
- Center for Tuberculosis, University of California San Francisco, San Francisco CA USA
- Division of Pulmonary Diseases and Critical Care Medicine, University of California Irvine, Orange CA USA
| | - Fred C Semitala
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
- Makerere University Joint AIDS Program, Kampala, Uganda
| |
Collapse
|
3
|
Krishnan S, Chaisson RE. US Guidelines Fall Short on Short-Course Tuberculosis-Preventive Therapy. Clin Infect Dis 2024; 78:514-517. [PMID: 37879092 PMCID: PMC10954328 DOI: 10.1093/cid/ciad659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/07/2023] [Accepted: 10/23/2023] [Indexed: 10/27/2023] Open
Abstract
The provision of tuberculosis-preventive therapy (TPT) to vulnerable populations is critical for global control. Shorter-course TPT regimens are highly effective and improve completion rates. Despite incorporation of 1 month of rifapentine and isoniazid into global guidelines, current US TPT guidelines do not include this as a recommended regimen, but should.
Collapse
Affiliation(s)
- Sonya Krishnan
- Center for Tuberculosis Research, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Richard E Chaisson
- Center for Tuberculosis Research, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Semitala FC, Kadota JL, Musinguzi A, Welishe F, Nakitende A, Akello L, Kunihira Tinka L, Nakimuli J, Ritar Kasidi J, Bishop O, Nakasendwa S, Baik Y, Patel D, Sammann A, Nahid P, Belknap R, Kamya MR, Handley MA, Phillips PPJ, Katahoire A, Berger CA, Kiwanuka N, Katamba A, Dowdy DW, Cattamanchi A. Comparison of 3 optimized delivery strategies for completion of isoniazid-rifapentine (3HP) for tuberculosis prevention among people living with HIV in Uganda: A single-center randomized trial. PLoS Med 2024; 21:e1004356. [PMID: 38377166 PMCID: PMC10914279 DOI: 10.1371/journal.pmed.1004356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 03/05/2024] [Accepted: 02/02/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Expanding access to shorter regimens for tuberculosis (TB) prevention, such as once-weekly isoniazid and rifapentine taken for 3 months (3HP), is critical for reducing global TB burden among people living with HIV (PLHIV). Our coprimary hypotheses were that high levels of acceptance and completion of 3HP could be achieved with delivery strategies optimized to overcome well-contextualized barriers and that 3HP acceptance and completion would be highest when PLHIV were provided an informed choice between delivery strategies. METHODS AND FINDINGS In a pragmatic, single-center, 3-arm, parallel-group randomized trial, PLHIV receiving care at a large urban HIV clinic in Kampala, Uganda, were randomly assigned (1:1:1) to receive 3HP by facilitated directly observed therapy (DOT), facilitated self-administered therapy (SAT), or informed choice between facilitated DOT and facilitated SAT using a shared decision-making aid. We assessed the primary outcome of acceptance and completion (≥11 of 12 doses of 3HP) within 16 weeks of treatment initiation using proportions with exact binomial confidence intervals (CIs). We compared proportions between arms using Fisher's exact test (two-sided α = 0.025). Trial investigators were blinded to primary and secondary outcomes by study arm. Between July 13, 2020, and July 8, 2022, 1,656 PLHIV underwent randomization, with equal numbers allocated to each study arm. One participant was erroneously enrolled a second time and was excluded in the primary intention-to-treat analysis. Among the remaining 1,655 participants, the proportion who accepted and completed 3HP exceeded the prespecified 80% target in the DOT (0.94; 97.5% CI [0.91, 0.96] p < 0.001), SAT (0.92; 97.5% CI [0.89, 0.94] p < 0.001), and Choice (0.93; 97.5% CI [0.91, 0.96] p < 0.001) arms. There was no difference in acceptance and completion between any 2 arms overall or in prespecified subgroup analyses based on sex, age, time on antiretroviral therapy, and history of prior treatment for TB or TB infection. Only 14 (0.8%) participants experienced an adverse event prompting discontinuation of 3HP. The main limitation of the study is that it was conducted in a single center. Multicenter studies are now needed to confirm the feasibility and generalizability of the facilitated 3HP delivery strategies in other settings. CONCLUSIONS Short-course TB preventive treatment was widely accepted by PLHIV in Uganda, and very high levels of treatment completion were achieved in a programmatic setting with delivery strategies tailored to address known barriers. TRIAL REGISTRATION ClinicalTrials.gov NCT03934931.
Collapse
Affiliation(s)
- Fred C. Semitala
- Makerere University, Department of Medicine, College of Health Sciences, Kampala, Uganda
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Makerere University Joint AIDS Program, Kampala Uganda
| | - Jillian L. Kadota
- Center for Tuberculosis, University of California San Francisco, San Francisco, California, United States of America
- Division of Pulmonary and Critical Care Medicine, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
| | | | - Fred Welishe
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Anne Nakitende
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Lydia Akello
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Jane Nakimuli
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Opira Bishop
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Suzan Nakasendwa
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Yeonsoo Baik
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Devika Patel
- The Better Lab and Department of Surgery, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
| | - Amanda Sammann
- The Better Lab and Department of Surgery, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
| | - Payam Nahid
- Center for Tuberculosis, University of California San Francisco, San Francisco, California, United States of America
- Division of Pulmonary and Critical Care Medicine, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
| | - Robert Belknap
- Denver Health and Hospital Authority and Division of Infectious Diseases, Department of Medicine, University of Colorado, Denver, Colorado, United States of America
| | - Moses R. Kamya
- Makerere University, Department of Medicine, College of Health Sciences, Kampala, Uganda
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Margaret A. Handley
- Center for Vulnerable Populations, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, United States of America
| | - Patrick PJ Phillips
- Center for Tuberculosis, University of California San Francisco, San Francisco, California, United States of America
- Division of Pulmonary and Critical Care Medicine, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
| | - Anne Katahoire
- Child Health and Development Center, School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Christopher A. Berger
- Center for Tuberculosis, University of California San Francisco, San Francisco, California, United States of America
- Division of Pulmonary and Critical Care Medicine, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
| | - Noah Kiwanuka
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Achilles Katamba
- Clinical Epidemiology & Biostatistics Unit, Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
- Uganda Tuberculosis Implementation Research Consortium, Walimu, Kampala, Uganda
| | - David W. Dowdy
- Uganda Tuberculosis Implementation Research Consortium, Walimu, Kampala, Uganda
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Adithya Cattamanchi
- Center for Tuberculosis, University of California San Francisco, San Francisco, California, United States of America
- Uganda Tuberculosis Implementation Research Consortium, Walimu, Kampala, Uganda
- Division of Pulmonary Diseases and Critical Care Medicine, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
5
|
Asif M, Qusty NF, Alghamdi S. An Overview of Various Rifampicin Analogs against Mycobacterium tuberculosis and their Drug Interactions. Med Chem 2024; 20:268-292. [PMID: 37855280 DOI: 10.2174/0115734064260853230926080134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/14/2023] [Accepted: 08/12/2023] [Indexed: 10/20/2023]
Abstract
The success of the TB control program is hampered by the major issue of drug-resistant tuberculosis (DR-TB). The situation has undoubtedly been made more difficult by the widespread and multidrug-resistant (XDR) strains of TB. The modification of existing anti-TB medications to produce derivatives that can function on resistant TB bacilli is one of the potential techniques to overcome drug resistance affordably and straightforwardly. In comparison to novel pharmaceuticals for drug research and progress, these may have a better half-life and greater bioavailability, be more efficient, and serve as inexpensive alternatives. Mycobacterium tuberculosis, which is drugsusceptible or drug-resistant, is effectively treated by several already prescribed medications and their derivatives. Due to this, the current review attempts to give a brief overview of the rifampicin derivatives that can overcome the parent drug's resistance and could, hence, act as useful substitutes. It has been found that one-third of the global population is affected by M. tuberculosis. The most common cause of infection-related death can range from latent TB to TB illness. Antibiotics in the rifamycin class, including rifampicin or rifampin (RIF), rifapentine (RPT), and others, have a special sterilizing effect on M. tuberculosis. We examine research focused on evaluating the safety, effectiveness, pharmacokinetics, pharmacodynamics, risk of medication interactions, and other characteristics of RIF analogs. Drug interactions are especially difficult with RIF because it must be taken every day for four months to treat latent TB infection. RIF continues to be the gold standard of treatment for drug-sensitive TB illness. RIF's safety profile is well known, and the two medicines' adverse reactions have varying degrees of frequency. The authorized once-weekly RPT regimen is insufficient, but greater dosages of either medication may reduce the amount of time needed to treat TB effectively.
Collapse
Affiliation(s)
- Mohammad Asif
- Department of Pharmaceutical Chemistry, Era College of Pharmacy, Era University, Lucknow, 226003, Uttar Pradesh, India
| | - Naeem F Qusty
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al‒Qura University, Makkah, 21955, Saudi Arabia
| | - Saad Alghamdi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al‒Qura University, Makkah, 21955, Saudi Arabia
| |
Collapse
|
6
|
Brooks KM, Pau AK, Swaim D, Bunn HT, Adeojo L, Peloquin CA, Kumar P, Kovacs JA, George JM. Pharmacokinetics, Safety, and Tolerability of Once-Daily Darunavir With Cobicistat and Weekly Isoniazid/Rifapentine. J Acquir Immune Defic Syndr 2023; 94:468-473. [PMID: 37955446 PMCID: PMC10651166 DOI: 10.1097/qai.0000000000003301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/21/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Once-weekly isoniazid with rifapentine (HP) for 3 months is a recommended treatment for latent tuberculosis infection in persons with HIV. HP reduces exposures of certain antiretroviral medications, resulting in limited options for the concomitant use of these therapies. Here, we examined the pharmacokinetics (PK), safety, and tolerability of darunavir/cobicistat with HP. METHODS This was an open-label, fixed sequence, two-period crossover study in persons without HIV. Participants received darunavir 800 mg/cobicistat 150 mg once-daily alone for 4 days, then continued darunavir/cobicistat once-daily for days 5-19 with HP coadministration on days 5, 12, and 19. Intensive PK assessments were performed on days 4, 14, and 19. PK parameters were determined using noncompartmental methods. Geometric mean ratios with 90% confidence intervals (CIs) were calculated and compared between phases using mixed-effects models. RESULTS Thirteen participants were enrolled. Two withdrew after day 4, and one withdrew after day 14. Of the 3 withdrawals, 2 were attributed to drug-related adverse events. Darunavir area under the concentration-time curve, maximum concentrations (Cmax), and concentrations at 24 hours postdose (C24h) were reduced by 71%, 41%, and 96% ∼48-72 hours after HP administration (day 14), respectively, and 36%, 17%, and 89% with simultaneous HP administration (day 19), respectively. On day 14, 45% of the predose and 73% of C24h concentrations were below the darunavir EC50 (0.055 µg/mL). CONCLUSIONS Darunavir exposures were significantly decreased with HP coadministration. Temporal relationships between HP coadministration and the extent of induction or mixed inhibition/induction of darunavir metabolism were apparent. Coadministration of darunavir/cobicistat with 3HP should be avoided.
Collapse
Affiliation(s)
- Kristina M Brooks
- Clinical Pharmacokinetic Research Laboratory, Clinical Center Pharmacy Department, NIH Clinical Center, Bethesda, MD, USA
- Currently, Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Alice K Pau
- National Institute of Allergy and Infectious Diseases, Bethesda, MD
| | - Doris Swaim
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Haden T Bunn
- Clinical Pharmacokinetic Research Laboratory, Clinical Center Pharmacy Department, NIH Clinical Center, Bethesda, MD, USA
- Currently, Pumas-AI, Inc., Centreville, VA
| | - Lilian Adeojo
- Clinical Pharmacokinetic Research Laboratory, Clinical Center Pharmacy Department, NIH Clinical Center, Bethesda, MD, USA
- Currently, Arcus Biosciences, Hayward, CA
| | - Charles A Peloquin
- University of Florida College of Pharmacy and Emerging Pathogens Institute, Gainesville, FL
| | - Parag Kumar
- Clinical Pharmacokinetic Research Laboratory, Clinical Center Pharmacy Department, NIH Clinical Center, Bethesda, MD, USA
- Currently, Gilead Sciences, Inc., Foster City, CA
| | - Joseph A Kovacs
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, MD; and
| | - Jomy M George
- Clinical Pharmacokinetic Research Laboratory, Clinical Center Pharmacy Department, NIH Clinical Center, Bethesda, MD, USA
- Currently, Office of Regulatory Affairs, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD
| |
Collapse
|
7
|
Mapingure MP, Zech JM, Hirsch-Moverman Y, Msukwa M, Howard AA, Makoni T, Gwanzura C, Apollo T, Sandy C, Musuka GN, Rabkin M. Integrating 3HP-based tuberculosis preventive treatment into Zimbabwe's Fast Track HIV treatment model: experiences from a pilot study. J Int AIDS Soc 2023; 26:e26105. [PMID: 37339341 DOI: 10.1002/jia2.26105] [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: 11/29/2022] [Accepted: 05/02/2023] [Indexed: 06/22/2023] Open
Abstract
INTRODUCTION Tuberculosis (TB) causes one-third of HIV-related deaths worldwide, making TB preventive treatment (TPT) a critical element of HIV programmes. Approximately 16% of people living with HIV (PLHIV) on antiretrovirals in Zimbabwe are enrolled in the Fast Track (FT) differentiated service delivery model, which includes multi-month dispensing of antiretrovirals and quarterly health facility (HF) visits. We assessed the feasibility and acceptability of utilizing FT to deliver 3HP (3 months of once-weekly rifapentine and isoniazid) for TPT by aligning TPT and HIV visits, providing multi-month dispensing of 3HP, and using phone-based monitoring and adherence support. METHODS We recruited a purposive sample of 50 PLHIV enrolled in FT at a high-volume HF in urban Zimbabwe. At enrolment, participants provided written informed consent, completed a baseline survey, and received counselling, education and a 3-month supply of 3HP. A study nurse mentor called participants at weeks 2, 4 and 8 to monitor and support adherence and side effects. When participants returned for their routine 3-month FT visit, they completed another survey, and study staff conducted a structured medical record review. In-depth interviews were conducted with providers who participated in the pilot. RESULTS Participants were enrolled between April and June 2021 and followed through September 2021. Median age = 32 years (IQR 24,41), 50% female, median time in FT 1.8 years (IQR 0.8,2.7). Forty-eight participants (96%) completed 3HP in 13 weeks; one completed in 16 weeks, and one stopped due to jaundice. Most participants (94%) reported "always" or "almost always" taking 3HP correctly. All reported they were very satisfied with the counselling, education, support and quality of care they received from providers and FT service efficiency. Almost all (98%) said they would recommend it to other PLHIV. Challenges reported included pill burden (12%) and tolerability (24%), but none had difficulty with phone-based counselling or wished for additional HF-based visits. DISCUSSION Using FT to deliver 3HP was feasible and acceptable. Some reported tolerability challenges but 98% completed 3HP, and all appreciated the efficiency of aligning TPT and HIV HF visits, multi-month dispensing and phone-based counselling. CONCLUSIONS Scaling up this approach could expand TPT coverage in Zimbabwe.
Collapse
Affiliation(s)
| | | | - Yael Hirsch-Moverman
- ICAP at Columbia University, New York City, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York City, New York, USA
| | | | - Andrea A Howard
- ICAP at Columbia University, New York City, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York City, New York, USA
| | - Tatenda Makoni
- Zimbabwe Network for People Living with HIV (ZNNP+), Harare, Zimbabwe
| | | | - Tsitsi Apollo
- Ministry of Health and Child Care (MoHCC), Harare, Zimbabwe
| | - Charles Sandy
- Ministry of Health and Child Care (MoHCC), Harare, Zimbabwe
| | - Godfrey N Musuka
- International Initiative for Impact Evaluation (3ie), New Delhi, India
| | - Miriam Rabkin
- ICAP at Columbia University, New York City, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York City, New York, USA
| |
Collapse
|
8
|
Aguilar Diaz JM, Abulfathi AA, te Brake LHM, van Ingen J, Kuipers S, Magis-Escurra C, Raaijmakers J, Svensson EM, Boeree MJ. New and Repurposed Drugs for the Treatment of Active Tuberculosis: An Update for Clinicians. Respiration 2023; 102:83-100. [PMID: 36516792 PMCID: PMC9932851 DOI: 10.1159/000528274] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/28/2022] [Indexed: 12/15/2022] Open
Abstract
Although tuberculosis (TB) is preventable and curable, the lengthy treatment (generally 6 months), poor patient adherence, high inter-individual variability in pharmacokinetics (PK), emergence of drug resistance, presence of comorbidities, and adverse drug reactions complicate TB therapy and drive the need for new drugs and/or regimens. Hence, new compounds are being developed, available drugs are repurposed, and the dosing of existing drugs is optimized, resulting in the largest drug development portfolio in TB history. This review highlights a selection of clinically available drug candidates that could be part of future TB regimens, including bedaquiline, delamanid, pretomanid, linezolid, clofazimine, optimized (high dose) rifampicin, rifapentine, and para-aminosalicylic acid. The review covers drug development history, preclinical data, PK, and current clinical development.
Collapse
Affiliation(s)
- Jessica M Aguilar Diaz
- Radboudumc Center for Infectious Diseases, Department of Pulmonary Diseases, TB Expert Center Dekkerswald, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ahmed A Abulfathi
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, Lake Nona (Orlando), University of Florida, Gainesville, Florida, USA,Department of Clinical Pharmacology and Therapeutics, Faculty of Basic Clinical Sciences, College of Medical Sciences, University of Maiduguri, Maiduguri, Nigeria,Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine & Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Lindsey HM te Brake
- Radboudumc Center for Infectious Diseases, Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jakko van Ingen
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Saskia Kuipers
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cecile Magis-Escurra
- Radboudumc Center for Infectious Diseases, Department of Pulmonary Diseases, TB Expert Center Dekkerswald, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jelmer Raaijmakers
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elin M Svensson
- Radboudumc Center for Infectious Diseases, Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands,Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Martin J Boeree
- Radboudumc Center for Infectious Diseases, Department of Pulmonary Diseases, TB Expert Center Dekkerswald, Radboud University Medical Center, Nijmegen, The Netherlands,*Martin J. Boeree,
| |
Collapse
|
9
|
Grañana-Castillo S, Montanha MC, Bearon R, Khoo S, Siccardi M. Evaluation of drug-drug interaction between rilpivirine and rifapentine using PBPK modelling. Front Pharmacol 2022; 13:1076266. [PMID: 36588698 PMCID: PMC9797969 DOI: 10.3389/fphar.2022.1076266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Tuberculosis remains the leading cause of death among people living with HIV. Rifapentine is increasingly used to treat active disease or prevent reactivation, in both cases given either as weekly or daily therapy. However, rifapentine is an inducer of CYP3A4, potentially interacting with antiretrovirals like rilpivirine. This in silico study investigates the drug-drug interaction (DDI) magnitude between daily oral rilpivirine 25 mg with either daily 600 mg or weekly 900 mg rifapentine. A physiologically based pharmacokinetic (PBPK) model was built in Simbiology (Matlab R2018a) to simulate the drug-drug interaction. The simulated PK parameters from the PBPK model were verified against reported clinical data for rilpivirine and rifapentine separately, daily rifapentine with midazolam, and weekly rifapentine with doravirine. The simulations of concomitant administration of rifapentine with rilpivirine at steady-state lead to a maximum decrease on AUC0-24 and Ctrough by 83% and 92% on day 5 for the daily rifapentine regimen and 68% and 92% for the weekly regimen on day 3. In the weekly regimen, prior to the following dose, AUC0-24 and Ctrough were still reduced by 47% and 53%. In both simulations, the induction effect ceased 2 weeks after the interruption of rifapentine's treatment. A daily double dose of rilpivirine after initiating rifapentine 900 mg weekly was simulated but failed to compensate the drug-drug interaction. The drug-drug interaction model suggested a significant decrease on rilpivirine exposure which is unlikely to be corrected by dose increment, thus coadministration should be avoided.
Collapse
Affiliation(s)
- Sandra Grañana-Castillo
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Maiara Camotti Montanha
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Rachel Bearon
- Department of Mathematical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Saye Khoo
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Marco Siccardi
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
10
|
Ammerman NC, Nuermberger EL, Owen A, Rannard SP, Meyers CF, Swindells S. Potential Impact of Long-Acting Products on the Control of Tuberculosis: Preclinical Advancements and Translational Tools in Preventive Treatment. Clin Infect Dis 2022; 75:S510-S516. [PMID: 36410384 PMCID: PMC10200320 DOI: 10.1093/cid/ciac672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A key component of global tuberculosis (TB) control is the treatment of latent TB infection. The use of long-acting technologies to administer TB preventive treatment has the potential to significantly improve the delivery and impact of this important public health intervention. For example, an ideal long-acting treatment could consist of a single dose that could be administered in the clinic (ie, a "1-shot cure" for latent TB). Interest in long-acting formulations for TB preventive therapy has gained considerable traction in recent years. This article presents an overview of the specific considerations and current preclinical advancements relevant for the development of long-acting technologies of TB drugs for treatment of latent infection, including attributes of target product profiles, suitability of drugs for long-acting formulations, ongoing research efforts, and translation to clinical studies.
Collapse
Affiliation(s)
- Nicole C Ammerman
- Department of Medical Microbiology and Infectious Diseases, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Center for Tuberculosis Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eric L Nuermberger
- Center for Tuberculosis Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Owen
- Centre of Excellence for Long-acting Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Steve P Rannard
- Centre of Excellence for Long-acting Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Caren Freel Meyers
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susan Swindells
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
11
|
Care cascade of tuberculosis infection treatment for people living with HIV in the era of antiretroviral therapy scale-up. Sci Rep 2022; 12:16136. [PMID: 36167744 PMCID: PMC9515204 DOI: 10.1038/s41598-022-20394-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 09/13/2022] [Indexed: 11/26/2022] Open
Abstract
Testing and treatment of tuberculosis infection (TBI) are recommended for people living with HIV (PLWH). We aimed to evaluate the care cascade of TBI treatment among PLWH in the era of antiretroviral therapy (ART) scale-up. This retrospective study included adult PLWH undergoing interferon-gamma release assay (IGRA)-based TBI screening during 2019–2021. PLWH testing IGRA-positive were advised to receive directly-observed therapy for TBI after active TB disease was excluded. The care cascade was evaluated to identify barriers to TBI management. Among 7951 PLWH with a median age of 38 years and CD4 count of 616 cells/mm3, 420 (5.3%) tested positive and 38 (0.5%) indeterminate for IGRA. The TBI treatment initiation rate was 73.6% (309/420) and the completion rate was 91.9% (284/309). More than 80% of PLWH concurrently received short-course rifapentine-based regimens and integrase strand transfer inhibitor (InSTI)-containing ART. The main barrier to treatment initiation was physicians’ concerns and patients’ refusal (85.6%). The factors associated with treatment non-completion were older age, female, anti-HCV positivity, and higher plasma HIV RNA. Our observation of a high TBI completion rate among PLWH is mainly related to the introduction of short-course rifapentine-based regimens in the InSTI era, which can be the strategy to improve TBI treatment uptake.
Collapse
|
12
|
Pham MM, Podany AT, Mwelase N, Supparatpinyo K, Mohapi L, Gupta A, Samaneka W, Omoz-Oarhe A, Langat D, Benson CA, Chaisson RE, Swindells S, Fletcher CV. Population Pharmacokinetic Modeling and Simulation of Rifapentine Supports Concomitant Antiretroviral Therapy with Efavirenz and Non-Weight Based Dosing. Antimicrob Agents Chemother 2022; 66:e0238521. [PMID: 35943252 PMCID: PMC9487628 DOI: 10.1128/aac.02385-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/20/2022] [Indexed: 11/20/2022] Open
Abstract
The Brief Rifapentine-Isoniazid Efficacy for TB Prevention/A5279 trial demonstrated a 1-month daily regimen of rifapentine and isoniazid was noninferior to 9 months of isoniazid alone for preventing TB in persons living with HIV (PLWH). Our objective was to evaluate rifapentine pharmacokinetics in trial participants receiving antiretroviral therapy (ART) and perform simulations to compare weight-based rifapentine dosing with a standard, fixed dose. Nonlinear mixed effect modeling was used to estimate rifapentine and 25-desacetyl rifapentine population pharmacokinetic characteristics. The pharmacokinetic model was validated using a nonparametric bootstrap and visual predictive checks. Monte Carlo simulations were performed to compare weight-based and fixed dose regimens. Rifapentine and 25-desacetyl rifapentine concentrations (347 of each; 185 participants) were each described with a one-compartment model with one-way conversion between rifapentine and 25-desacetyl rifapentine. The absorption rate was nearly doubled in fed versus fasting states. Rifapentine clearance was increased 31% in those receiving efavirenz (EFV)-based versus nevirapine-based ART. Metabolite clearance was allometrically scaled with fat-free mass. Simulations showed lower rifapentine exposures with weight-based compared with fixed dosing. With 10 mg/kg weight-based regimens, 26% and 62% of simulated exposures in <35 kg and 35-45 kg weight classes were above target (AUC0 to 24 h of 257 mg*hr/L); 85% of simulated exposures across all weight classes with fixed dosing were above target. These data support fixed dosing with rifapentine 600 mg daily for TB prevention regardless of weight for PLWH 13 years or older receiving the 4-week regimen and no need for dose adjustment when given with EFV-based ART. Clinical Trials Registration. NCT01404312.
Collapse
Affiliation(s)
- Michelle M. Pham
- Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Anthony T. Podany
- Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Noluthando Mwelase
- University of the Witwatersrand Helen Joseph CRS, Johannesburg, South Africa
| | | | - Lerato Mohapi
- Soweto ACTG CRS, Perinatal HIV Research Unit, Johannesburg, South Africa
| | - Amita Gupta
- Johns Hopkins Hospital, Baltimore, Maryland, USA
| | | | | | - Deborah Langat
- Kenya Medical Research Institute/Walter Reed Project Clinical Research Center CRS, Kericho, Kenya
| | | | - Richard E. Chaisson
- Johns Hopkins University School of Medicine, Center for Tuberculosis Research, Baltimore, Maryland, USA
| | - Susan Swindells
- Division of Infectious Diseases, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Courtney V. Fletcher
- Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Division of Infectious Diseases, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
13
|
Podany AT, Pham M, Sizemore E, Martinson N, Samaneka W, Mohapi L, Badal-Faesen S, Dawson R, Johnson JL, Mayanja H, Lalloo U, Whitworth WC, Pettit A, Campbell K, Phillips PPJ, Bryant K, Scott N, Vernon A, Kurbatova EV, Chaisson RE, Dorman SE, Nahid P, Swindells S, Dooley KE, Fletcher CV. Efavirenz Pharmacokinetics and Human Immunodeficiency Virus Type 1 (HIV-1) Viral Suppression Among Patients Receiving Tuberculosis Treatment Containing Daily High-Dose Rifapentine. Clin Infect Dis 2022; 75:560-566. [PMID: 34918028 PMCID: PMC9890454 DOI: 10.1093/cid/ciab1037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND A 4-month regimen containing rifapentine and moxifloxacin has noninferior efficacy compared to the standard 6-month regimen for drug-sensitive tuberculosis. We evaluated the effect of regimens containing daily, high-dose rifapentine on efavirenz pharmacokinetics and viral suppression in patients with human immunodeficiency virus (HIV)-associated tuberculosis (TB). METHODS In the context of a Phase 3 randomized controlled trial, HIV-positive individuals already virally suppressed on efavirenz--containing antiretroviral therapy (ART) (EFV1), or newly initiating efavirenz (EFV2) received TB treatment containing rifapentine (1200 mg), isoniazid, pyrazinamide, and either ethambutol or moxifloxacin. Mid-interval efavirenz concentrations were measured (a) during ART and TB cotreatment (Weeks 4, 8, 12, and 17, different by EFV group) and (b) when ART was taken alone (pre- or post-TB treatment, Weeks 0 and 22). Apparent oral clearance (CL/F) was estimated and compared. Target mid-interval efavirenz concentrations were > 1 mg/L. Co-treatment was considered acceptable if > 80% of participants had mid-interval efavirenz concentrations meeting this target. RESULTS EFV1 and EFV2 included 70 and 41 evaluable participants, respectively. The geometric mean ratio comparing efavirenz CL/F with vs without TB drugs was 0.79 (90% confidence interval [CI] .72-.85) in EFV1 and 0.84 [90% CI .69-.97] in EFV2. The percent of participants with mid-interval efavirenz concentrations > 1mg/L in EFV1 at Weeks 0, 4, 8, and 17 was 96%, 96%, 88%, and 89%, respectively. In EFV2, at approximately 4 and 8 weeks post efavirenz initiation, the value was 98%. CONCLUSIONS TB treatment containing high-dose daily rifapentine modestly decreased (rather than increased) efavirenz clearance and therapeutic targets were met supporting the use of efavirenz with these regimens, without dose adjustment. CLINICAL TRIALS REGISTRATION NCT02410772.
Collapse
Affiliation(s)
| | - Michelle Pham
- University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Erin Sizemore
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Neil Martinson
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg, South Africa
| | | | - Lerato Mohapi
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg, South Africa
| | | | - Rod Dawson
- University of Cape Town Lung Institute, Cape Town, South Africa
| | | | - Harriet Mayanja
- Uganda- Case Western Reserve University Research Collaboration, Kampala, Uganda
| | - Umesh Lalloo
- Durban International Clinical Research Site, Durban, South Africa
| | | | - April Pettit
- Vanderbilt University, Nashville, Tennessee, USA
| | - Kayla Campbell
- University of Nebraska Medical Center, Omaha, Nebraska, USA
- University of Colorado, Denver, Colorado, USA
| | - Patrick P J Phillips
- University of California, San Francisco Center for Tuberculosis, San Francisco, California, USA
| | - Kia Bryant
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nigel Scott
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Andrew Vernon
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | | | - Susan E Dorman
- Medical University of South Carolina, Columbia, South Carolina, USA
| | - Payam Nahid
- University of California, San Francisco Center for Tuberculosis, San Francisco, California, USA
| | | | - Kelly E Dooley
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
14
|
Ntinginya NE, Te Brake L, Sabi I, Chamba N, Kilonzo K, Laizer S, Andia-Biraro I, Kibirige D, Kyazze AP, Ninsiima S, Critchley JA, Romeo R, van de Maat J, Olomi W, Mrema L, Magombola D, Mwayula IH, Sharples K, Hill PC, van Crevel R. Rifapentine and isoniazid for prevention of tuberculosis in people with diabetes (PROTID): protocol for a randomised controlled trial. Trials 2022; 23:480. [PMID: 35689272 PMCID: PMC9186476 DOI: 10.1186/s13063-022-06296-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Diabetes mellitus (DM) increases the risk of tuberculosis (TB) and will hamper global TB control due to the dramatic rise in type 2 DM in TB-endemic settings. In this trial, we will examine the efficacy and safety of TB preventive therapy against the development of TB disease in people with DM who have latent TB infection (LTBI), with a 12-week course of rifapentine and isoniazid (3HP). METHODS The 'Prevention of tuberculosis in diabetes mellitus' (PROTID) consortium will randomise 3000 HIV-negative eligible adults with DM and LTBI, as evidenced by a positive tuberculin skin test or interferon gamma release assay, to 12 weeks of 3HP or placebo. Participants will be recruited through screening adult patients attending DM clinics at referral hospitals in Tanzania and Uganda. Patients with previous TB disease or treatment with a rifamycin medication or isoniazid (INH) in the previous 2 years will be excluded. The primary outcome is the occurrence of definite or probable TB disease; secondary outcome measures include adverse events, all-cause mortality and treatment completion. The primary efficacy analysis will be intention-to-treat; per-protocol analyses will also be carried out. We will estimate the ratio of TB incidence rates in intervention and control groups, adjusting for the study site using Poisson regression. Results will be reported as efficacy estimates (1-rate ratio). Cumulative incidence rates allowing for death as a competing risk will also be reported. Approximately 1000 LTBI-negative, HIV-negative participants will be enrolled consecutively into a parallel cohort study to compare the incidence of TB in people with DM who are LTBI negative vs positive. A number of sub-studies will be conducted among others to examine the prevalence of LTBI and active TB, estimate the population impact and cost-effectiveness of LTBI treatment in people living with DM in these African countries and address gaps in the prevention and therapeutic management of combined TB-DM. DISCUSSION PROTID is anticipated to generate key evidence to guide decisions over the use of TB preventive treatment among people with DM as an important target group for better global TB control. TRIAL REGISTRATION ClinicalTrials.gov NCT04600167 . Registered on 23 October 2020.
Collapse
Affiliation(s)
- Nyanda Elias Ntinginya
- National Institute for Medical Research (NIMR), Mbeya Medical Research Centre, Mbeya, Tanzania.
| | - Lindsey Te Brake
- Departmentt of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), Nijmegen, The Netherlands
| | - Issa Sabi
- National Institute for Medical Research (NIMR), Mbeya Medical Research Centre, Mbeya, Tanzania
| | - Nyasatu Chamba
- The Good Samaritan Foundation (Kilimanjaro Christian Medical Centre GSF KCMC), Moshi, Tanzania
- Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Kajiru Kilonzo
- The Good Samaritan Foundation (Kilimanjaro Christian Medical Centre GSF KCMC), Moshi, Tanzania
- Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Sweetness Laizer
- The Good Samaritan Foundation (Kilimanjaro Christian Medical Centre GSF KCMC), Moshi, Tanzania
- Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Irene Andia-Biraro
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Andrew Peter Kyazze
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Sandra Ninsiima
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | | | - Josephine van de Maat
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Willyhelmina Olomi
- National Institute for Medical Research (NIMR), Mbeya Medical Research Centre, Mbeya, Tanzania
| | - Lucy Mrema
- National Institute for Medical Research (NIMR), Mbeya Medical Research Centre, Mbeya, Tanzania
| | - David Magombola
- National Institute for Medical Research (NIMR), Mbeya Medical Research Centre, Mbeya, Tanzania
| | | | - Katrina Sharples
- Otago Global Health Institute, University of Otago, Dunedin, New Zealand
| | - Philip C Hill
- Otago Global Health Institute, University of Otago, Dunedin, New Zealand
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Ignatius EH, Swindells S. Update on tuberculosis/HIV coinfections: across the spectrum from latent infection through drug-susceptible and drug-resistant disease. TOPICS IN ANTIVIRAL MEDICINE 2022; 30:464-472. [PMID: 36346703 PMCID: PMC9306687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Tuberculosis (TB) remains the leading cause of death among people with HIV, and annual risk of progression from latent TB infection to active disease in this population is 10%. Diagnostic tests for latent and active TB remain suboptimal for people with HIV who have a CD4+ count below 200 cells/μL, and there is an urgent need for assays that predict progression from latent to active disease, monitor treatment response, and test for cure after latent and active TB treatment. Traditional treatment duration for latent infection and active TB disease has been onerous for patients; however, shorter-course regimens are increasingly available across the spectrum of TB, including for drug-resistant TB. Simultaneous treatment of HIV and TB is complicated by drug-drug interactions, although trials are ongoing to better understand the magnitude of these interactions and guide clinicians in how to use short-course regimens, particularly for people with HIV.
Collapse
Affiliation(s)
| | - Susan Swindells
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Marzolini C, Gibbons S, van Oosterhout JJ, Khoo S. Drug-Drug Interaction Potential with Once-Weekly Isoniazid/Rifapentine (3HP) for the Treatment of Latent Tuberculosis Infection. Clin Pharmacokinet 2022; 61:339-346. [PMID: 34905153 DOI: 10.1007/s40262-021-01098-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Catia Marzolini
- Division of Infectious Diseases and Hospital Epidemiology, Departments of Medicine and Clinical Research, University Hospital of Basel, University of Basel, Petersgraben 4, 4031, Basel, Switzerland.
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Sara Gibbons
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Joep J van Oosterhout
- Partners in Hope, Lilongwe, Malawi
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Saye Khoo
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
17
|
Semitala FC, Kadota JL, Musinguzi A, Nabunje J, Welishe F, Nakitende A, Akello L, Bishop O, Patel D, Sammann A, Nahid P, Belknap R, Kamya MR, Handley MA, Phillips PPJ, Katahoire A, Berger CA, Kiwanuka N, Katamba A, Dowdy DW, Cattamanchi A. Completion of isoniazid-rifapentine (3HP) for tuberculosis prevention among people living with HIV: Interim analysis of a hybrid type 3 effectiveness-implementation randomized trial. PLoS Med 2021; 18:e1003875. [PMID: 34914696 PMCID: PMC8726462 DOI: 10.1371/journal.pmed.1003875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/04/2022] [Accepted: 11/25/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Scaling up shorter regimens for tuberculosis (TB) prevention such as once weekly isoniazid-rifapentine (3HP) taken for 3 months is a key priority for achieving targets set forth in the World Health Organization's (WHO) END TB Strategy. However, there are few data on 3HP patient acceptance and completion in the context of routine HIV care in sub-Saharan Africa. METHODS AND FINDINGS The 3HP Options Trial is a pragmatic, parallel type 3 effectiveness-implementation randomized trial comparing 3 optimized strategies for delivering 3HP-facilitated directly observed therapy (DOT), facilitated self-administered therapy (SAT), or informed choice between DOT and SAT using a shared decision-making aid-to people receiving care at a large urban HIV clinic in Kampala, Uganda. Participants and healthcare providers were not blinded to arm assignment due to the nature of the 3HP delivery strategies. We conducted an interim analysis of participants who were enrolled and exited the 3HP treatment period between July 13, 2020 and April 30, 2021. The primary outcome, which was aggregated across trial arms for this interim analysis, was the proportion who accepted and completed 3HP (≥11 of 12 doses within 16 weeks of randomization). We used Bayesian inference analysis to estimate the posterior probability that this proportion would exceed 80% under at least 1 of the 3HP delivery strategies, a coprimary hypothesis of the trial. Through April 2021, 684 participants have been enrolled, and 479 (70%) have exited the treatment period. Of these 479 participants, 309 (65%) were women, mean age was 41.9 years (standard deviation (SD): 9.2), and mean time on antiretroviral therapy (ART) was 7.8 years (SD: 4.3). In total, 445 of them (92.9%, 95% confidence interval (CI): [90.2 to 94.9]) accepted and completed 3HP treatment. There were no differences in treatment acceptance and completion by sex, age, or time on ART. Treatment was discontinued due to a documented adverse event (AE) in 8 (1.7%) patients. The probability that treatment acceptance and completion exceeds 80% under at least 1 of the three 3HP delivery strategies was greater than 99%. The main limitations are that the trial was conducted at a single site, and the interim analysis focused on aggregate outcome data to maintain blinding of investigators to arm-specific outcomes. CONCLUSIONS 3HP was widely accepted by people living with HIV (PLHIV) in Uganda, and very high levels of treatment completion were achieved in a programmatic setting. These findings show that 3HP can enable effective scale-up of tuberculosis preventive therapy (TPT) in high-burden countries, particularly when delivery strategies are tailored to target known barriers to treatment completion. TRIAL REGISTRATION ClinicalTrials.gov NCT03934931.
Collapse
Affiliation(s)
- Fred C. Semitala
- Makerere University, Department of Medicine, College of Health Sciences, Kampala, Uganda
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Makerere University Joint AIDS Program, Kampala, Uganda
| | - Jillian L. Kadota
- UCSF Center for Tuberculosis and Division of Pulmonary and Critical Care Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, California, United States of America
| | | | - Juliet Nabunje
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Fred Welishe
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Anne Nakitende
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Lydia Akello
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Opira Bishop
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Devika Patel
- The Better Lab, Department of Surgery, University of California, San Francisco, San Francisco, California, United States of America
| | - Amanda Sammann
- The Better Lab, Department of Surgery, University of California, San Francisco, San Francisco, California, United States of America
| | - Payam Nahid
- UCSF Center for Tuberculosis and Division of Pulmonary and Critical Care Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, California, United States of America
| | - Robert Belknap
- Denver Health and Hospital Authority and Division of Infectious Diseases, Department of Medicine, University of Colorado, Denver, Colorado, United States of America
| | - Moses R. Kamya
- Makerere University, Department of Medicine, College of Health Sciences, Kampala, Uganda
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Margaret A. Handley
- Center for Vulnerable Populations at Zuckerberg San Francisco General Hospital and Trauma Center, University of California, San Francisco, San Francisco, California, United States of America
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, United States of America
| | - Patrick P. J. Phillips
- UCSF Center for Tuberculosis and Division of Pulmonary and Critical Care Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, California, United States of America
| | - Anne Katahoire
- Child Health and Development Center, School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Christopher A. Berger
- UCSF Center for Tuberculosis and Division of Pulmonary and Critical Care Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, California, United States of America
| | - Noah Kiwanuka
- Clinical Epidemiology & Biostatistics Unit, Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Achilles Katamba
- Clinical Epidemiology & Biostatistics Unit, Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
- Uganda Tuberculosis Implementation Research Consortium, Kampala, Uganda
| | - David W. Dowdy
- Uganda Tuberculosis Implementation Research Consortium, Kampala, Uganda
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Adithya Cattamanchi
- UCSF Center for Tuberculosis and Division of Pulmonary and Critical Care Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, California, United States of America
- Uganda Tuberculosis Implementation Research Consortium, Kampala, Uganda
- * E-mail:
| |
Collapse
|
18
|
Liou BH, Cheng CN, Lin YT, Lin YJ, Chuang YC, Lin KY, Liu WC, Lin SW, Kuo CH, Sun HY, Hung CC. Short-course daily isoniazid and rifapentine for latent tuberculosis infection in people living with HIV who received coformulated bictegravir/emtricitabine/tenofovir alafenamide. J Int AIDS Soc 2021; 24:e25844. [PMID: 34822220 PMCID: PMC8614225 DOI: 10.1002/jia2.25844] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 10/27/2021] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Short-course preventive therapy with 1-month course of daily administration of isoniazid (300-mg) plus rifapentine (600-mg) (1HP) and 3-month course of weekly administration of isoniazid (900-mg) plus rifapentine (900-mg) (3HP) has higher completion rates than 9-month course of daily isoniazid (9H) for individuals with latent tuberculosis infection (LTBI). We aimed to evaluate the effect, safety and tolerability of 1HP in people living with HIV (PLWH) and LTBI who received coformulated bictegravir/emtricitabine/tenofovir alafenamide (BIC/FTC/TAF). METHODS PLWH testing positive by interferon-gamma release assay and having received BIC/FTC/TAF for >2 weeks with plasma HIV RNA load (PVL) <200 copies/ml were enrolled. BIC trough plasma concentrations and cytokine profiles were determined before the first dose (day 1/baseline), 24 h after the 14th (day 15) and 28th (day 29) doses of 1HP. PVL were determined on days 15 and 29 of 1HP and every 3 months subsequently after discontinuation of 1HP. RESULTS From November 2019 to December 2020, 48 PLWH with LTBI were enrolled. One participant (2.1%) discontinued 1HP on day 15 due to fever and generalized rashes with PVL of 72 copies/ml, which was <50 copies/ml in three subsequent determinations while on BIC/FTC/TAF over the 12 months of follow-up. The percentages of BIC trough plasma concentrations above the protein-adjusted 95% effective concentration (paEC95 = 162 ng/ml) were 56.3% and 37.0% on days 15 and 29, respectively. The percentage of PVL <200 copies/ml was 91.7% on day 15, 97.8% on day 29 and 100% at both months 3 and 6. After a median observation of 52 weeks (interquartile range, 51-55), all participants continued BIC/FTC/TAF with a median PVL of 20 copies/ml (range 20-331). Except for the participant who discontinued 1HP because of allergic reactions, none of the participants had relevant symptoms or increases of the cytokine levels assessed between baseline and days 15 and 29 of 1HP. CONCLUSIONS BIC/FTC/TAF in combination with 1HP was well tolerated with a high completion rate. BIC trough plasma concentrations were significantly decreased with concurrent use of 1HP among PLWH with LTBI. While transient viral blips were observed during 1HP without causing subsequent treatment failure, such combination should be applied with caution.
Collapse
Affiliation(s)
- Bo-Huang Liou
- Department of Internal Medicine, Hsinchu MacKay Memorial Hospital, Hsinchu City, Taiwan
| | - Chih-Ning Cheng
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Ya-Ting Lin
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Yu-Jou Lin
- Department of Pharmacy, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Chung Chuang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuan-Yin Lin
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wen-Chun Liu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shu-Wen Lin
- School of Pharmacy, National Taiwan University, Taipei, Taiwan.,Department of Pharmacy, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate Institute of Clinical Pharmacy, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Hua Kuo
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yun Sun
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chien-Ching Hung
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Tropical Medicine and Parasitology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
19
|
Podany AT, Leon-Cruz J, Hakim J, Supparatpinyo K, Omoz-Oarhe A, Langat D, Mwelase N, Kanyama C, Gupta A, Benson CA, Chaisson RE, Swindells S, Fletcher CV. Nevirapine pharmacokinetics in HIV-infected persons receiving rifapentine and isoniazid for TB prevention. J Antimicrob Chemother 2021; 76:718-721. [PMID: 33241266 DOI: 10.1093/jac/dkaa470] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/19/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The use of rifamycin antibiotics for TB prevention carries a risk of detrimental drug-drug interactions with concomitantly used ART. OBJECTIVES To evaluate the interaction of the antiretroviral drug nevirapine in combination with 4 weeks of daily rifapentine and isoniazid for TB prevention in people living with HIV. METHODS Participants were individuals enrolled in the BRIEF-TB study receiving nevirapine and randomized to the rifapentine/isoniazid arm of the study. Participants provided sparse pharmacokinetic (PK) sampling at baseline and weeks 2 and 4 for trough nevirapine determination. Nevirapine apparent oral clearance (CL/F) was estimated and the geometric mean ratio (GMR) of CL/F prior to and during rifapentine/isoniazid was calculated. RESULTS Seventy-eight participants had evaluable PK data: 61 (78%) female, 51 (65%) black non-Hispanic and median (range) age of 40 (13-66) years. Median (IQR) nevirapine trough concentrations were: week 0, 7322 (5266-9302) ng/mL; week 2, 5537 (3552-8462) ng/mL; and week 4, 5388 (3516-8243) ng/mL. Sixty out of 78 participants (77%) had nevirapine concentrations ≥3000 ng/mL at both week 2 and 4. Median (IQR) nevirapine CL/F values were: week 0 pre-rifapentine/isoniazid, 2.03 (1.58-2.58) L/h; and during rifapentine/isoniazid, 2.62 (1.81-3.42) L/h. The GMR (90% CI) for nevirapine CL/F was 1.30 (1.26-1.33). CONCLUSIONS The CL/F of nevirapine significantly increased with concomitant rifapentine/isoniazid. The decrease in nevirapine trough concentrations during rifapentine/isoniazid therapy suggests induction of nevirapine metabolism, consistent with known rifapentine effects. The magnitude of this drug-drug interaction suggests daily rifapentine/isoniazid for TB prevention should not be co-administered with nevirapine-containing ART.
Collapse
Affiliation(s)
- A T Podany
- University of Nebraska Medical Center, Omaha, NE, USA
| | - J Leon-Cruz
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - J Hakim
- Parirenyatwa CRS, Harare, Zimbabwe
| | - K Supparatpinyo
- Chiang Mai University HIV Treatment CRS, Chiang Mai, Thailand
| | - A Omoz-Oarhe
- Molepolole Clinical Research Site, Molepolole, Botswana
| | - D Langat
- Kenya Medical Research Institute/Walter Reed Project Clinical Research Center CRS, Kericho, Kenya
| | - N Mwelase
- University of the Witwatersrand Helen Joseph CRS, Johannesburg, South Africa
| | | | - A Gupta
- Johns Hopkins Hospital, Baltimore, MD, USA
| | - C A Benson
- University of California San Diego, San Diego, CA, USA
| | | | - S Swindells
- University of Nebraska Medical Center, Omaha, NE, USA
| | - C V Fletcher
- University of Nebraska Medical Center, Omaha, NE, USA
| | | |
Collapse
|
20
|
Abstract
Tuberculosis (TB) remains a leading cause of morbidity and mortality among people living with HIV. HIV-associated TB disproportionally affects African countries, particularly vulnerable groups at risk for both TB and HIV. Currently available TB diagnostics perform poorly in people living with HIV; however, new diagnostics such as Xpert Ultra and lateral flow urine lipoarabinomannan assays can greatly facilitate diagnosis of TB in people living with HIV. TB preventive treatment has been underutilized despite its proven benefits independent of antiretroviral therapy (ART). Shorter regimens using rifapentine can support increased availability and scale-up. Mortality is high in people with HIV-associated TB, and timely initiation of ART is critical. Programs should provide decentralized and integrated TB and HIV care in settings with high burden of both diseases to improve access to services that diagnose TB and HIV as early as possible. The new prevention and diagnosis tools recently recommended by WHO offer an immense opportunity to advance our fight against HIV-associated TB. They should be made widely available and scaled up rapidly supported by adequate funding with robust monitoring of the uptake to advance global TB elimination.
Collapse
Affiliation(s)
- Yohhei Hamada
- Centre for International Cooperation and Global TB Information, 46635Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan.,Institute for Global Health, 4919University College London, London, UK
| | - Haileyesus Getahun
- Department of Global Coordination and Partnership on Antimicrobial Resistance, 3489WHO, Geneva, Switzerland
| | - Birkneh Tilahun Tadesse
- Department of Global Coordination and Partnership on Antimicrobial Resistance, 3489WHO, Geneva, Switzerland
| | - Nathan Ford
- Department of Paediatrics, College of Medicine and Health Sciences, 128167Hawassa University, Hawassa, Ethiopia
| |
Collapse
|
21
|
Haas DW, Podany AT, Bao Y, Swindells S, Chaisson RE, Mwelase N, Supparatpinyo K, Mohapi L, Gupta A, Benson CA, Baker P, Fletcher CV. Pharmacogenetic interactions of rifapentine plus isoniazid with efavirenz or nevirapine. Pharmacogenet Genomics 2021; 31:17-27. [PMID: 32815870 PMCID: PMC7655626 DOI: 10.1097/fpc.0000000000000417] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVES The effect of rifapentine plus isoniazid on efavirenz pharmacokinetics was characterized in AIDS Clinical Trials Group protocol A5279 (NCT01404312). The present analyses characterize pharmacogenetic interactions between these drugs, and with nevirapine. METHODS A subset of HIV-positive individuals receiving efavirenz- or nevirapine-containing antiretroviral therapy in A5279 underwent pharmacokinetic evaluations at baseline, and again weeks 2 and 4 after initiating daily rifapentine plus isoniazid. Associations with polymorphisms relevant to efavirenz, nevirapine, isoniazid, and rifapentine pharmacokinetics were assessed. RESULTS Of 128 participants, 101 were evaluable for associations with rifapentine and its active 25-desacetyl metabolite, 87 with efavirenz, and 38 with nevirapine. In multivariable analyses, NAT2 slow acetylators had greater week 4 plasma concentrations of rifapentine (P = 2.6 × 10) and 25-desacetyl rifapentine (P = 7.0 × 10) among all participants, and in efavirenz and nevirapine subgroups. NAT2 slow acetylators also had greater plasma efavirenz and nevirapine concentration increases from baseline to week 4, and greater decreases from baseline in clearance. CYP2B6 poor metabolizers had greater efavirenz concentrations at all weeks and greater nevirapine concentrations at baseline. None of 47 additional polymorphisms in 11 genes were significantly associated with pharmacokinetics. CONCLUSIONS Among HIV-positive individuals receiving efavirenz or nevirapine, and who then initiated rifapentine plus isoniazid in A5279, NAT2 slow acetylators had greater rifapentine and 25-desacetyl rifapentine concentrations, and greater increases from baseline in plasma efavirenz and nevirapine concentrations. These associations are likely mediated by greater isoniazid exposure in NAT2 slow acetylators.
Collapse
Affiliation(s)
- David W Haas
- Department of Medicine, Vanderbilt University School of Medicine
| | - Anthony T Podany
- Department of Pharmacy Practice and Science, Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha
| | - Yajing Bao
- Statistical and Data Analysis Center, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Susan Swindells
- Infectious Diseases, Internal Medicine, University of Nebraska Medical Center, Omaha
| | - Richard E Chaisson
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Noluthando Mwelase
- Helen Joseph Hospital, University of Witwatersrand University, Johannesburg, South Africa
| | - Khuanchai Supparatpinyo
- Department of Medicine, Research Institute for Health Sciences and Faculty of Medicine, Chiang Mai University, Thailand
| | - Lerato Mohapi
- Perinatal HIV Research Unit, University of the Witwatersrand and Chris Hani Baragwanath Hospital, Soweto, South Africa
| | - Amita Gupta
- Department of Medicine, Center for Clinical Global Health Education, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Constance A Benson
- Departments of Medicine and Global Public Health, Antiviral Research Center, University of California, San Diego
| | - Paxton Baker
- Vanderbilt Technologies for Advanced Genomics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Courtney V Fletcher
- Department of Pharmacy Practice and Science, Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha; for the AIDS Clinical Trials Group A5279 Study Team
| |
Collapse
|
22
|
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
|
23
|
Ignatius EH, Swindells S. Are We There Yet? Short-Course Regimens in TB and HIV: From Prevention to Treatment of Latent to XDR TB. Curr HIV/AIDS Rep 2020; 17:589-600. [PMID: 32918195 PMCID: PMC9178518 DOI: 10.1007/s11904-020-00529-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW Despite broad uptake of antiretroviral therapy (ART), tuberculosis (TB) incidence and mortality among people with HIV remain unacceptably high. Short-course regimens for TB, incorporating both novel and established drugs, offer the potential to enhance adherence and completion rates, thereby reducing the global TB burden. This review will outline short-course regimens for TB among patients with HIV. RECENT FINDINGS After many years without new agents, there is now active testing of many novel drugs to treat TB, both for latent infection and active disease. Though not all studies have included patients with HIV, many have, and there are ongoing trials to address key implementation challenges such as potent drug-drug interactions with ART. The goal of short-course regimens for TB is to enhance treatment completion without compromising efficacy. Particularly among patients with HIV, studying these shortened regimens and integrating them into clinical care are of urgent importance. There are now multiple short-course regimens for latent infection and active disease that are safe and effective among patients with HIV.
Collapse
Affiliation(s)
- Elisa H Ignatius
- Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Susan Swindells
- University of Nebraska Medical Center, Omaha, NE, 68198-8106, USA.
| |
Collapse
|
24
|
Jacobs TG, Svensson EM, Musiime V, Rojo P, Dooley KE, McIlleron H, Aarnoutse RE, Burger DM, Turkova A, Colbers A. Pharmacokinetics of antiretroviral and tuberculosis drugs in children with HIV/TB co-infection: a systematic review. J Antimicrob Chemother 2020; 75:3433-3457. [PMID: 32785712 PMCID: PMC7662174 DOI: 10.1093/jac/dkaa328] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/29/2020] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Management of concomitant use of ART and TB drugs is difficult because of the many drug-drug interactions (DDIs) between the medications. This systematic review provides an overview of the current state of knowledge about the pharmacokinetics (PK) of ART and TB treatment in children with HIV/TB co-infection, and identifies knowledge gaps. METHODS We searched Embase and PubMed, and systematically searched abstract books of relevant conferences, following PRISMA guidelines. Studies not reporting PK parameters, investigating medicines that are not available any longer or not including children with HIV/TB co-infection were excluded. All studies were assessed for quality. RESULTS In total, 47 studies met the inclusion criteria. No dose adjustments are necessary for efavirenz during concomitant first-line TB treatment use, but intersubject PK variability was high, especially in children <3 years of age. Super-boosted lopinavir/ritonavir (ratio 1:1) resulted in adequate lopinavir trough concentrations during rifampicin co-administration. Double-dosed raltegravir can be given with rifampicin in children >4 weeks old as well as twice-daily dolutegravir (instead of once daily) in children older than 6 years. Exposure to some TB drugs (ethambutol and rifampicin) was reduced in the setting of HIV infection, regardless of ART use. Only limited PK data of second-line TB drugs with ART in children who are HIV infected have been published. CONCLUSIONS Whereas integrase inhibitors seem favourable in older children, there are limited options for ART in young children (<3 years) receiving rifampicin-based TB therapy. The PK of TB drugs in HIV-infected children warrants further research.
Collapse
Affiliation(s)
- Tom G Jacobs
- Radboud University Medical Center, Radboud Institute for Health Sciences, Department of Pharmacy, Nijmegen, The Netherlands
| | - Elin M Svensson
- Radboud University Medical Center, Radboud Institute for Health Sciences, Department of Pharmacy, Nijmegen, The Netherlands
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Victor Musiime
- Research Department, Joint Clinical Research Centre, Kampala, Uganda
- Department of Paediatrics and Child Health, School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Pablo Rojo
- Pediatric Infectious Diseases Unit. Hospital 12 de Octubre, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Kelly E Dooley
- Divisions of Clinical Pharmacology and Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Helen McIlleron
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Rob E Aarnoutse
- Radboud University Medical Center, Radboud Institute for Health Sciences, Department of Pharmacy, Nijmegen, The Netherlands
| | - David M Burger
- Radboud University Medical Center, Radboud Institute for Health Sciences, Department of Pharmacy, Nijmegen, The Netherlands
| | - Anna Turkova
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, University College London, London, UK
| | - Angela Colbers
- Radboud University Medical Center, Radboud Institute for Health Sciences, Department of Pharmacy, Nijmegen, The Netherlands
| |
Collapse
|
25
|
Kadota JL, Musinguzi A, Nabunje J, Welishe F, Ssemata JL, Bishop O, Berger CA, Patel D, Sammann A, Katahoire A, Nahid P, Belknap R, Phillips PPJ, Namusobya J, Kamya M, Handley MA, Kiwanuka N, Katamba A, Dowdy D, Semitala FC, Cattamanchi A. Protocol for the 3HP Options Trial: a hybrid type 3 implementation-effectiveness randomized trial of delivery strategies for short-course tuberculosis preventive therapy among people living with HIV in Uganda. Implement Sci 2020; 15:65. [PMID: 32787925 PMCID: PMC7425004 DOI: 10.1186/s13012-020-01025-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/27/2020] [Indexed: 12/14/2022] Open
Abstract
Background Recently, a 3-month (12-dose) regimen of weekly isoniazid and rifapentine (3HP) was recommended by the World Health Organization for the prevention of tuberculosis (TB) among people living with HIV (PLHIV) on common antiretroviral therapy regimens. The best approach to delivering 3HP to PLHIV remains uncertain. Methods We developed a three-armed randomized trial assessing optimized strategies for delivering 3HP to PLHIV. The trial will be conducted at the Mulago Immune Suppression Syndrome (i.e., HIV/AIDS) clinic in Kampala, Uganda. We plan to recruit 1656 PLHIV, randomized 1:1 to each of the three arms (552 per arm). Using a hybrid type 3 effectiveness-implementation design, this pragmatic trial aims to (1) compare the acceptance and completion of 3HP among PLHIV under three delivery strategies: directly observed therapy (DOT), self-administered therapy (SAT), and informed patient choice of either DOT or SAT (with the assistance of a decision aid); (2) to identify processes and contextual factors that influence the acceptance and completion of 3HP under each delivery strategy; and (3) to estimate the costs and compare the cost-effectiveness of three strategies for delivering 3HP. The three delivery strategies were each optimized to address key barriers to 3HP completion using a theory-informed approach. We hypothesize that high levels of treatment acceptance and completion can be achieved among PLHIV in sub-Saharan Africa and that offering PLHIV an informed choice between the optimized DOT and SAT delivery strategies will result in greater acceptance and completion of 3HP. The design and planned evaluation of the delivery strategies were guided by the use of implementation science conceptual frameworks. Discussion 3HP—one of the most promising interventions for TB prevention—will not be scaled up unless it can be delivered in a patient-centered fashion. We highlight shared decision-making as a key element of our trial design and theorize that offering PLHIV an informed choice between optimized delivery strategies will facilitate the highest levels of treatment acceptance and completion. Trial registration ClinicalTrials.gov: NCT03934931; Registered 2 May 2019.
Collapse
Affiliation(s)
- Jillian L Kadota
- Division of Pulmonary and Critical Care Medicine and Center for Tuberculosis, University of California San Francisco, San Francisco, CA, USA
| | | | - Juliet Nabunje
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Fred Welishe
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Opira Bishop
- Makerere University Joint AIDS Program, Kampala, Uganda
| | - Christopher A Berger
- Division of Pulmonary and Critical Care Medicine and Center for Tuberculosis, University of California San Francisco, San Francisco, CA, USA
| | - Devika Patel
- Department of Surgery, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Amanda Sammann
- Department of Surgery, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Anne Katahoire
- Child Health and Development Centre, Makerere University, Kampala, Uganda
| | - Payam Nahid
- Division of Pulmonary and Critical Care Medicine and Center for Tuberculosis, University of California San Francisco, San Francisco, CA, USA
| | - Robert Belknap
- Denver Health and Hospital Authority, Denver, CO, USA.,Division of Infectious Diseases, Department of Medicine, University of Colorado, Denver, CO, USA
| | - Patrick P J Phillips
- Division of Pulmonary and Critical Care Medicine and Center for Tuberculosis, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer Namusobya
- University Research Company, Center for Human Services, Department of Defense HIV/AIDS Prevention Program (URC-DHAPP), Kampala, Uganda
| | - Moses Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda.,Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Margaret A Handley
- Center for Vulnerable Populations at Zuckerberg San Francisco General Hospital and Trauma Center, University of California, San Francisco, San Francisco, CA, USA.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Noah Kiwanuka
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University, Kampala, Uganda
| | - Achilles Katamba
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda.,Uganda Tuberculosis Implementation Research Consortium, Kampala, Uganda
| | - David Dowdy
- Uganda Tuberculosis Implementation Research Consortium, Kampala, Uganda.,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Fred C Semitala
- Infectious Diseases Research Collaboration, Kampala, Uganda. .,Makerere University Joint AIDS Program, Kampala, Uganda. .,Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda. .,Mulago- ISS Clinic, Old Mulago Hill Road, New Mulago Hospital Complex, P.O Box 7051, Kampala, Uganda.
| | - Adithya Cattamanchi
- Division of Pulmonary and Critical Care Medicine and Center for Tuberculosis, University of California San Francisco, San Francisco, CA, USA.,Center for Vulnerable Populations at Zuckerberg San Francisco General Hospital and Trauma Center, University of California, San Francisco, San Francisco, CA, USA.,Uganda Tuberculosis Implementation Research Consortium, Kampala, Uganda
| |
Collapse
|
26
|
Hepatocytic transcriptional signatures predict comparative drug interaction potential of rifamycin antibiotics. Sci Rep 2020; 10:12565. [PMID: 32724080 PMCID: PMC7387492 DOI: 10.1038/s41598-020-69228-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
Current strategies to treat tuberculosis (TB) and co-morbidities involve multidrug combination therapies. Rifamycin antibiotics are a key component of TB therapy and a common source of drug–drug interactions (DDIs) due to induction of drug metabolizing enzymes (DMEs). Management of rifamycin DDIs are complex, particularly in patients with co-morbidities, and differences in DDI potential between rifamycin antibiotics are not well established. DME profiles induced in response to tuberculosis antibiotics (rifampin, rifabutin and rifapentine) were compared in primary human hepatocytes. We identified rifamycin induced DMEs, cytochrome P450 (CYP) 2C8/3A4/3A5, SULT2A, and UGT1A4/1A5 and predicted lower DDIs of rifapentine with 58 clinical drugs used to treat co-morbidities in TB patients. Transcriptional networks and upstream regulator analyses showed FOXA3, HNF4α, NR1I2, NR1I3, NR3C1 and RXRα as key transcriptional regulators of rifamycin induced DMEs. Our study findings are an important resource to design effective medication regimens to treat common co-conditions in TB patients.
Collapse
|
27
|
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
|
28
|
Dooley KE, Savic R, Gupte A, Marzinke MA, Zhang N, Edward VA, Wolf L, Sebe M, Likoti M, Fyvie MJ, Shibambo I, Beattie T, Chaisson RE, Churchyard GJ. Once-weekly rifapentine and isoniazid for tuberculosis prevention in patients with HIV taking dolutegravir-based antiretroviral therapy: a phase 1/2 trial. Lancet HIV 2020; 7:e401-e409. [PMID: 32240629 DOI: 10.1016/s2352-3018(20)30032-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Short-course preventive therapy with 12 doses of once-weekly rifapentine (900 mg) plus isoniazid (900 mg) could greatly improve tuberculosis control, especially in areas with high co-endemicity with HIV. However, a small previous trial of such therapy with dolutegravir in healthy, HIV-negative adults was halted early after two of the four patients developed serious adverse events. Because of the potential use of this therapy, and variable safety outcomes of tuberculosis drugs seen in patients with and without HIV, we aimed to characterise safety, pharmacokinetics, and virological suppression in adults who are HIV positive. METHODS DOLPHIN was a phase 1/2, single-arm trial done at The Aurum Institute (Tembisa Clinical Research Site, Tembisa, South Africa), with pharmacokinetic visits done at VxPharma (Pretoria, South Africa). Adults (≥18 years) with HIV infection and undetectable viral load (<40 copies per mL) after at least 8 weeks of efavirenz-based or dolutegravir-based regimens were recruited in three consecutive groups, subject to approval by the independent safety monitoring committee. Participants received 50 mg of daily dolutegravir in place of efavirenz for 8 weeks, then began once-weekly rifapentine (900 mg)-isoniazid (900 mg) for 12 weeks. Groups 1A (n=12) and 1B (n=18) had intensive dolutegravir pharmacokinetic sampling at week 8 (before rifapentine-isoniazid), at week 11 (after the third dose of rifapentine)-isoniazid and at week 16 after the eighth dose. Group 2 (n=30) were treated with the same schedule and had sparse dolutegravir pharmacokinetic sampling at weeks 8, 11, and 16. Participants were followed 4 weeks after completion of prophylactic tuberculosis treatment. HIV viral loads were measured at baseline and at weeks 11 and 24. Primary endpoints were adverse events (grade 3 or higher) and dolutegravir population pharmacokinetics, assessed in participants who began rifapentine-isoniazid. This trial was registered at ClinicalTrials.gov, NCT03435146. FINDINGS Between Jan 24, 2018, and Nov 25, 2018, 61 participants were enrolled into three groups; one participant withdrew (from group 1A). 43 (70%) of 60 participants were women and all participants were black African. Median age was 40 years (IQR 35-48), CD4 cell count was 683 cells per μL (447-935), and body-mass index was 28·9 kg/m2 (24·0-32·9). Three grade 3 adverse events occurred; two elevated creatinine and one hypertension. Rifapentine-isoniazid increased dolutegravir clearance by 36% (relative standard error 13%) resulting in a 26% decrease in dolutegravir area under the curve. Overall geometric mean ratio of trough concentrations with versus without rifapentine-isoniazid was 0·53 (90% CI 0·49-0·56) though this ratio varied by day after rifapentine-isoniazid dose. All but one trough value was above the 90% maximal inhibitory concentration for dolutegravir and HIV viral loads were less than 40 copies per mL in all patients. INTERPRETATION Our results suggest 12 doses of once-weekly rifapentine-isoniazid can be given for tuberculosis prophylaxis to patients with HIV taking dolutegravir-based antiretroviral therapy, without dose adjustments. Further exploration of the pharmacokinetics, safety, and efficacy in children and pharmacodynamics in individuals naive to antiretroviral therapy is needed. FUNDING UNITAID.
Collapse
Affiliation(s)
- Kelly E Dooley
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Radojkam Savic
- University of California San Francisco, San Francisco, CA, USA
| | - Akshay Gupte
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark A Marzinke
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nan Zhang
- University of California San Francisco, San Francisco, CA, USA
| | - Vinodh A Edward
- The Aurum Institute, Johannesburg, South Africa; School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Lisa Wolf
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | - Richard E Chaisson
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gavin J Churchyard
- The Aurum Institute, Johannesburg, South Africa; School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | |
Collapse
|
29
|
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
|
30
|
Cerrone M, Bracchi M, Wasserman S, Pozniak A, Meintjes G, Cohen K, Wilkinson RJ. Safety implications of combined antiretroviral and anti-tuberculosis drugs. Expert Opin Drug Saf 2020; 19:23-41. [PMID: 31809218 PMCID: PMC6938542 DOI: 10.1080/14740338.2020.1694901] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/15/2019] [Indexed: 01/01/2023]
Abstract
Introduction: Antiretroviral and anti-tuberculosis (TB) drugs are often co-administered in people living with HIV (PLWH). Early initiation of antiretroviral therapy (ART) during TB treatment improves survival in patients with advanced HIV disease. However, safety concerns related to clinically significant changes in drug exposure resulting from drug-drug interactions, development of overlapping toxicities and specific challenges related to co-administration during pregnancy represent barriers to successful combined treatment for HIV and TB.Areas covered: Pharmacokinetic interactions of different classes of ART when combined with anti-TB drugs used for sensitive-, drug-resistant (DR) and latent TB are discussed. Overlapping drug toxicities, implications of immune reconstitution inflammatory syndrome (IRIS), safety in pregnancy and research gaps are also explored.Expert opinion: New antiretroviral and anti-tuberculosis drugs have been recently introduced and international guidelines updated. A number of effective molecules and clinical data are now available to build treatment regimens for PLWH with latent or active TB. Adopting a systematic approach that also takes into account the need for individualized variations based on the available evidence is the key to successfully integrate ART and TB treatment and improve treatment outcomes.
Collapse
Affiliation(s)
- Maddalena Cerrone
- Department of Medicine, Imperial College London, W2 1PG, UK
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Observatory 7925, South Africa
- Department of HIV, Chelsea and Westminster Hospital NHS Trust, London, UK
- Francis Crick Institute, London, NW1 1AT, UK
| | - Margherita Bracchi
- Department of HIV, Chelsea and Westminster Hospital NHS Trust, London, UK
| | - Sean Wasserman
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Observatory 7925, South Africa
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Anton Pozniak
- Department of HIV, Chelsea and Westminster Hospital NHS Trust, London, UK
- The London School of Hygiene & Tropical Medicine
| | - Graeme Meintjes
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Karen Cohen
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, South Africa
| | - Robert J Wilkinson
- Department of Medicine, Imperial College London, W2 1PG, UK
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Observatory 7925, South Africa
- Francis Crick Institute, London, NW1 1AT, UK
| |
Collapse
|
31
|
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: 16] [Impact Index Per Article: 4.0] [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
|
32
|
Yu YY, Tsao SM, Yang WT, Huang WC, Lin CH, Chen WW, Yang SF, Chiou HL, Huang YW. Association of Drug Metabolic Enzyme Genetic Polymorphisms and Adverse Drug Reactions in Patients Receiving Rifapentine and Isoniazid Therapy for Latent Tuberculosis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 17:ijerph17010210. [PMID: 31892222 PMCID: PMC6981901 DOI: 10.3390/ijerph17010210] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/24/2019] [Accepted: 12/25/2019] [Indexed: 12/11/2022]
Abstract
Weekly rifapentine and isoniazid therapy (3HP) is the most frequent treatment for latent tuberculosis infection (LTBI). However, the association between major adverse drug reactions (ADRs) and drug metabolic enzyme single-nucleotide polymorphisms (SNPs) remains unclear. In this study, 377 participants who received the 3HP regimen were recruited and examined for genotyping of CYP5A6, CYP2B6, CYP2C19, CYP2E1, and NAT2 SNPs. In our study, 184 participants (48.4%) developed ADRs. Moreover, CYP2C19 rs4986893 (TT vs. CC+CT, odds ratio [OR] [95% CI]: 2.231 [1.015-4.906]), CYP2E1 rs2070676 (CC vs. CG+GG, OR [95% CI]: 1.563 [1.022-2.389]), and CYP2E1 rs2515641 (CC vs. CT+TT, OR [95% CI]: 1.903 [1.250-2.898]) were associated with ADR development. In conclusion, CYP2C19 and CYP2E1 SNPs may provide useful information regarding ADRs in LTBI patients receiving the 3HP regimen.
Collapse
Affiliation(s)
- Ya-Yen Yu
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-Y.Y.); (S.-F.Y.)
- Department of Clinical Laboratory, Changhua Hospital, Changhua 513, Taiwan
| | - Shih-Ming Tsao
- Division of Chest, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung 402, Taiwan;
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung 402, Taiwan
| | - Wen-Ta Yang
- Department of Internal Medicine, Taichung Hospital, Ministry of Health and Welfare, Taichung 403, Taiwan;
| | - Wei-Chang Huang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan;
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung 407, Taiwan
| | - Ching-Hsiung Lin
- Division of Chest, Changhua Christian Hospital, Changhua 500, Taiwan;
| | - Wei-Wen Chen
- Department of Health, Pulmonary and Critical Care Unit, Changhua Hospital, Changhua 500, Taiwan;
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-Y.Y.); (S.-F.Y.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Correspondence: (H.-L.C.); (Y.-W.H.)
| | - Yi-Wen Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-Y.Y.); (S.-F.Y.)
- Department of Health, Pulmonary and Critical Care Unit, Changhua Hospital, Changhua 500, Taiwan;
- Correspondence: (H.-L.C.); (Y.-W.H.)
| |
Collapse
|
33
|
Brooks KM, George JM, Pau AK, Rupert A, Mehaffy C, De P, Dobos KM, Kellogg A, McLaughlin M, McManus M, Alfaro RM, Hadigan C, Kovacs JA, Kumar P. Cytokine-Mediated Systemic Adverse Drug Reactions in a Drug-Drug Interaction Study of Dolutegravir With Once-Weekly Isoniazid and Rifapentine. Clin Infect Dis 2019; 67:193-201. [PMID: 29415190 DOI: 10.1093/cid/ciy082] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/01/2018] [Indexed: 12/18/2022] Open
Abstract
Background Once-weekly isoniazid and rifapentine for 3 months is a treatment option in persons with human immunodeficiency virus and latent tuberculosis infection. This study aimed to examine pharmacokinetic drug-drug interactions between this regimen and dolutegravir, a first-line antiretroviral medication. Methods This was a single-center, open-label, fixed-sequence, drug-drug interaction study in healthy volunteers. Subjects received oral dolutegravir 50 mg once daily alone (days 1-4) and concomitantly with once-weekly isoniazid 900 mg, rifapentine 900 mg, and pyridoxine 50 mg (days 5-19). Dolutegravir concentrations were measured on days 4, 14, and 19, and rifapentine, 25-desacetyl-rifapentine, and isoniazid concentrations were measured on day 19. Cytokines and antidrug antibodies to isoniazid and rifapentine were examined at select time points. Results The study was terminated following the development of flu-like syndrome and elevated aminotransferase levels in 2 of 4 subjects after the third isoniazid-rifapentine dose. Markedly elevated levels of interferon-γ, CXCL10, C-reactive protein, and other cytokines were temporally associated with symptoms. Antidrug antibodies were infrequently detected. Dolutegravir area under the curve (AUC) was decreased by 46% (90% confidence interval, 27-110%; P = .13) on day 14. Rifapentine and 25-desacetyl rifapentine levels on day 19 were comparable to reference data, whereas isoniazid AUCs were approximately 67%-92% higher in the subjects who developed toxicities. Conclusions The combined use of dolutegravir with once-weekly isoniazid-rifapentine resulted in unexpected and serious toxicities that were mediated by endogenous cytokine release. Additional investigations are necessary to examine the safety and efficacy of coadministering these medications. Clinical Trials Registration NCT02771249.
Collapse
Affiliation(s)
- Kristina M Brooks
- Clinical Pharmacokinetics Research Unit, Pharmacy Department, Clinical Center, National Institutes of Health (NIH), Frederick, Maryland
| | - Jomy M George
- Clinical Pharmacokinetics Research Unit, Pharmacy Department, Clinical Center, National Institutes of Health (NIH), Frederick, Maryland
| | - Alice K Pau
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, Frederick, Maryland
| | - Adam Rupert
- AIDS Monitoring Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, Maryland
| | - Carolina Mehaffy
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins
| | - Prithwiraj De
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins
| | - Karen M Dobos
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins
| | - Anela Kellogg
- Clinical Monitoring Research Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, Bethesda, Maryland
| | | | - Maryellen McManus
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland
| | - Raul M Alfaro
- Clinical Pharmacokinetics Research Unit, Pharmacy Department, Clinical Center, National Institutes of Health (NIH), Frederick, Maryland
| | | | - Joseph A Kovacs
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland
| | - Parag Kumar
- Clinical Pharmacokinetics Research Unit, Pharmacy Department, Clinical Center, National Institutes of Health (NIH), Frederick, Maryland
| |
Collapse
|
34
|
Gupta A, Hughes MD, Garcia-Prats AJ, McIntire K, Hesseling AC. Inclusion of key populations in clinical trials of new antituberculosis treatments: Current barriers and recommendations for pregnant and lactating women, children, and HIV-infected persons. PLoS Med 2019; 16:e1002882. [PMID: 31415563 PMCID: PMC6695091 DOI: 10.1371/journal.pmed.1002882] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Amita Gupta and colleagues discuss priorities in clinical research aimed at improving tuberculosis prevention and treatment in pregnant women, children, and people with HIV.
Collapse
Affiliation(s)
- Amita Gupta
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Michael D. Hughes
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Anthony J. Garcia-Prats
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Katherine McIntire
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Anneke C. Hesseling
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
35
|
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: 35] [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
|
36
|
Trinité B, Zhang H, Levy DN. NNRTI-induced HIV-1 protease-mediated cytotoxicity induces rapid death of CD4 T cells during productive infection and latency reversal. Retrovirology 2019; 16:17. [PMID: 31242909 PMCID: PMC6595680 DOI: 10.1186/s12977-019-0479-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/14/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Current efforts towards HIV-1 eradication focus on the reactivation and elimination of the latent viral reservoir, so-called shock and kill therapy. However, work from several groups indicates that infected cell death following virus reactivation is not guaranteed. Thus, it is imperative to develop strategies to foster specific elimination of cells carrying integrated proviruses. It has been shown that some non-nucleoside reverse transcriptase inhibitors (NNRTIs) including efavirenz can induce premature HIV-1 GagPol dimerization in productively infected cells, resulting in intracellular HIV-1 Protease (PR) activation and a reduction in HIV-1 expressing cells. RESULTS Here, we document that NNRTI-induced PR activation triggers apoptotic death of productively infected resting or activated T cells in as little as 2 h via caspase-dependent and independent pathways. Rilpivirine, efavirenz and etravirine were the most potent NNRTIs, whereas nevirapine had almost no effect. NNRTI-induced cell killing was prevented by inhibitors of HIV-1 Protease (PR) activity including indinavir and nelfinavir. HIV-1 transmitter founder viruses induced cell killing similarly to lab-adapted HIV-1 except when NNRTI resistance conferring mutations were present in reverse transcriptase. Mutations in PR that confer PR inhibitor (PI) resistance restore NNRTI-induced killing in the presence of PI. Finally, we show that NNRTIs can rapidly eliminate cells in which latent viruses are stimulated to active expression. CONCLUSIONS This work supports the notion that select NNRTIs might help promote the elimination of HIV-1 producing cells as an adjuvant during shock and kill therapy.
Collapse
Affiliation(s)
- Benjamin Trinité
- Department of Basic Science, New York University College of Dentistry, New York, NY, USA. .,IrsiCaixa AIDS Research Institute, Badalona, Spain.
| | - Hongtao Zhang
- Department of Basic Science, New York University College of Dentistry, New York, NY, USA
| | - David N Levy
- Department of Basic Science, New York University College of Dentistry, New York, NY, USA.
| |
Collapse
|
37
|
Fry SHL, Barnabas SL, Cotton MF. Tuberculosis and HIV-An Update on the "Cursed Duet" in Children. Front Pediatr 2019; 7:159. [PMID: 32211351 PMCID: PMC7073470 DOI: 10.3389/fped.2019.00159] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/04/2019] [Indexed: 11/13/2022] Open
Abstract
HIV and tuberculosis (TB) often occur together with each exacerbating the other. Improvements in vertical transmission prevention has reduced the number of HIV-infected children being born and early antiretroviral therapy (ART) protects against tuberculosis. However, with delayed HIV diagnosis, HIV-infected infants often present with tuberculosis co-infection. The number of HIV exposed uninfected children has increased and these infants have high exposure to TB and may be more immunologically vulnerable due to HIV exposure in utero. Bacillus Calmette-Guérin (BCG) immunization shortly after birth is essential for preventing severe TB in infancy. With early infant HIV diagnosis and ART, disseminated BCG is no longer an issue. TB prevention therapy should be implemented for contacts of a source case and for all HIV-infected individuals over a year of age. Although infection can be identified through skin tests or interferon gamma release assays, the non-availability of these tests should not preclude prevention therapy, once active TB has been excluded. Therapeutic options have moved from isoniazid only for 6-9 months to shorter regimens. Prevention therapy after exposure to a source case with resistant TB should also be implemented, but should not prevent pivotal prevention trials already under way. A microbiological diagnosis for TB remains the gold standard because of increasing drug resistance. Antiretroviral therapy for rifampicin co-treatment requires adaptation for those on lopinavir-ritonavir, which requires super-boosting with additional ritonavir. For those with drug resistant TB, the main problems are identification and overlapping toxicity between antiretroviral and anti-TB therapy. In spite of renewed focus and improved interventions, infants are still vulnerable to TB.
Collapse
Affiliation(s)
| | | | - Mark F. Cotton
- Family Centre for Research with Ubuntu (FAM-CRU), Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
38
|
Churchyard GJ, Swindells S. Controlling latent TB tuberculosis infection in high-burden countries: A neglected strategy to end TB. PLoS Med 2019; 16:e1002787. [PMID: 31013273 PMCID: PMC6478264 DOI: 10.1371/journal.pmed.1002787] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In a Perspective, Gavin Churchyard and Sue Swindells discuss the importance of strategies to target latent tuberculosis infection in high risk populations and thus disrupt a reservoir for new infections in high burden countries.
Collapse
Affiliation(s)
- Gavin J. Churchyard
- Aurum Institute, Parktown, South Africa
- University of Witwatersrand, School of Public Health, Johannesburg, South Africa
- London School of Hygiene and Tropical Medicine, London, United Kingdom
- * E-mail:
| | - Sue Swindells
- University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
39
|
Swindells S, Ramchandani R, Gupta A, Benson CA, Leon-Cruz J, Mwelase N, Jean Juste MA, Lama JR, Valencia J, Omoz-Oarhe A, Supparatpinyo K, Masheto G, Mohapi L, da Silva Escada RO, Mawlana S, Banda P, Severe P, Hakim J, Kanyama C, Langat D, Moran L, Andersen J, Fletcher CV, Nuermberger E, Chaisson RE. One Month of Rifapentine plus Isoniazid to Prevent HIV-Related Tuberculosis. N Engl J Med 2019; 380:1001-1011. [PMID: 30865794 PMCID: PMC6563914 DOI: 10.1056/nejmoa1806808] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Tuberculosis is the leading killer of patients with human immunodeficiency virus (HIV) infection. Preventive therapy is effective, but current regimens are limited by poor implementation and low completion rates. METHODS We conducted a randomized, open-label, phase 3 noninferiority trial comparing the efficacy and safety of a 1-month regimen of daily rifapentine plus isoniazid (1-month group) with 9 months of isoniazid alone (9-month group) in HIV-infected patients who were living in areas of high tuberculosis prevalence or who had evidence of latent tuberculosis infection. The primary end point was the first diagnosis of tuberculosis or death from tuberculosis or an unknown cause. Noninferiority would be shown if the upper limit of the 95% confidence interval for the between-group difference in the number of events per 100 person-years was less than 1.25. RESULTS A total of 3000 patients were enrolled and followed for a median of 3.3 years. Of these patients, 54% were women; the median CD4+ count was 470 cells per cubic millimeter, and half the patients were receiving antiretroviral therapy. The primary end point was reported in 32 of 1488 patients (2%) in the 1-month group and in 33 of 1498 (2%) in the 9-month group, for an incidence rate of 0.65 per 100 person-years and 0.67 per 100 person-years, respectively (rate difference in the 1-month group, -0.02 per 100 person-years; upper limit of the 95% confidence interval, 0.30). Serious adverse events occurred in 6% of the patients in the 1-month group and in 7% of those in the 9-month group (P = 0.07). The percentage of treatment completion was significantly higher in the 1-month group than in the 9-month group (97% vs. 90%, P<0.001). CONCLUSIONS A 1-month regimen of rifapentine plus isoniazid was noninferior to 9 months of isoniazid alone for preventing tuberculosis in HIV-infected patients. The percentage of patients who completed treatment was significantly higher in the 1-month group. (Funded by the National Institute of Allergy and Infectious Diseases; BRIEF TB/A5279 ClinicalTrials.gov number, NCT01404312.).
Collapse
Affiliation(s)
- Susan Swindells
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Ritesh Ramchandani
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Amita Gupta
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Constance A Benson
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Jorge Leon-Cruz
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Noluthando Mwelase
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Marc A Jean Juste
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Javier R Lama
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Javier Valencia
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Ayotunde Omoz-Oarhe
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Khuanchai Supparatpinyo
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Gaerolwe Masheto
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Lerato Mohapi
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Rodrigo O da Silva Escada
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Sajeeda Mawlana
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Peter Banda
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Patrice Severe
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - James Hakim
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Cecilia Kanyama
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Deborah Langat
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Laura Moran
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Janet Andersen
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Courtney V Fletcher
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Eric Nuermberger
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| | - Richard E Chaisson
- From the University of Nebraska Medical Center, Omaha (S.S., C.V.F.); Harvard T.H. Chan School of Public Health, Boston (R.R., J.L.-C., J.A.); Johns Hopkins University School of Medicine, Baltimore (A.G., E.N., R.E.C.), and Social and Scientific Systems, Silver Spring (L. Moran) - both in Maryland; University of California, San Diego, School of Medicine, La Jolla (C.A.B.); GHESKIO, Port-au-Prince, Haiti (M.A.J.J., P.S.); Asociación Civil Impacta Salud y Educación, Lima, Peru (J.R.L., J.V.); Botswana-Harvard AIDS Partnership, Gaborone, Botswana (A.O.-O., G.M.); Chiang Mai University, Chiang Mai, Thailand (K.S.); Helen Joseph Hospital, Johannesburg (N.M.), Perinatal HIV Research Unit, Soweto (L. Mohapi), and the University of Kwa-Zulu Natal, Durban (S.M.) - all in South Africa; Instituto de Pesquisa Clínica Evandro Chagas, Rio de Janeiro (R.O.S.E.); Johns Hopkins-Blantyre Clinical Trials Unit, Blantyre (P.B.), and the University of North Carolina-Lilongwe Clinical Research Site, Lilongwe (C.K.) - both in Malawi; the University of Zimbabwe, Harare (J.H.); and Kenya Medical Research Institute-Walter Reed Clinical Research Site, Nairobi (D.L.)
| |
Collapse
|
40
|
Davy-Mendez T, Shiau R, Okada RC, Moss NJ, Huang S, Murgai N, Chitnis AS. Combining surveillance systems to investigate local trends in tuberculosis-HIV co-infection. AIDS Care 2019; 31:1311-1318. [PMID: 30729804 DOI: 10.1080/09540121.2019.1576845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Alameda County has some of the highest human immunodeficiency virus (HIV) and tuberculosis (TB) case rates of California counties. We identified TB-HIV co-infected patients in 2002-2015 by matching county TB and HIV registries, and assessed trends in TB-HIV case rates and estimated prevalence ratios for HIV co-infection. Of 2054 TB cases reported during 2002-2015, 91 (4%) were HIV co-infected. TB-HIV case rates were 0.29/100,000 and 0.40/100,000 in 2002 and 2015, respectively, with no significant change (P = 0.85). African-American TB case-patients were 9.77 times (95% confidence interval [CI] 5.90-16.17) more likely than Asians to be HIV co-infected, and men 2.74 times (95% CI 1.66-4.51) more likely co-infected than women. HIV co-infection was more likely among TB case-patients with homelessness (6.21, 95% CI 3.49-11.05) and injection drug use (11.75, 95% CI 7.61-18.14), but less common among foreign-born and older case-patients (both P < 0.05). Among foreign-born case-patients, 42% arrived in the U.S. within 5 years of TB diagnosis. TB-HIV case rates were low and stable in Alameda County, and co-infected patients were predominantly young, male, U.S.-born individuals with traditional TB risk factors. Efforts to reduce TB-HIV burden in Alameda County should target persons with traditional TB risk factors and recently arrived foreign-born individuals.
Collapse
Affiliation(s)
- Thibaut Davy-Mendez
- a Gillings School of Global Public Health, University of North Carolina at Chapel Hill , Chapel Hill , NC , USA.,b HIV STD Section, Alameda County Public Health Department , Oakland , CA , USA
| | - Rita Shiau
- c Tuberculosis Control Section, Alameda County Public Health Department , San Leandro , CA , USA
| | - Reiko C Okada
- c Tuberculosis Control Section, Alameda County Public Health Department , San Leandro , CA , USA
| | - Nicholas J Moss
- b HIV STD Section, Alameda County Public Health Department , Oakland , CA , USA
| | - Sandra Huang
- d Acute Communicable Disease Section, Alameda County Public Health Department , Oakland , CA , USA
| | - Neena Murgai
- b HIV STD Section, Alameda County Public Health Department , Oakland , CA , USA
| | - Amit S Chitnis
- c Tuberculosis Control Section, Alameda County Public Health Department , San Leandro , CA , USA
| |
Collapse
|
41
|
Borisov AS, Bamrah Morris S, Njie GJ, Winston CA, Burton D, Goldberg S, Yelk Woodruff R, Allen L, LoBue P, Vernon A. Update of Recommendations for Use of Once-Weekly Isoniazid-Rifapentine Regimen to Treat Latent Mycobacterium tuberculosis Infection. MMWR-MORBIDITY AND MORTALITY WEEKLY REPORT 2018; 67:723-726. [PMID: 29953429 PMCID: PMC6023184 DOI: 10.15585/mmwr.mm6725a5] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Andrey S Borisov
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC
| | - Sapna Bamrah Morris
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC
| | - Gibril J Njie
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC
| | - Carla A Winston
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC
| | - Deron Burton
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC
| | - Stefan Goldberg
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC
| | - Rachel Yelk Woodruff
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC
| | - Leeanna Allen
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC
| | - Philip LoBue
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC
| | - Andrew Vernon
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC
| |
Collapse
|
42
|
Tasillo A, Salomon JA, Trikalinos TA, Horsburgh CR, Marks SM, Linas BP. Cost-effectiveness of Testing and Treatment for Latent Tuberculosis Infection in Residents Born Outside the United States With and Without Medical Comorbidities in a Simulation Model. JAMA Intern Med 2017; 177:1755-1764. [PMID: 29049814 PMCID: PMC5808933 DOI: 10.1001/jamainternmed.2017.3941] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Testing for and treating latent tuberculosis infection (LTBI) is among the main strategies to achieve TB elimination in the United States. The best approach to testing among non-US born residents, particularly those with comorbid conditions, is uncertain. OBJECTIVE To estimate health outcomes, costs, and cost-effectiveness of LTBI testing and treatment among non-US born residents with and without medical comorbidities. DESIGN, SETTING, AND PARTICIPANTS Decision analytic tree and Markov cohort simulation model among non-US born residents with no comorbidities, with diabetes, with HIV infection, or with end-stage renal disease (ESRD) using a health care sector perspective with 3% annual discounting. Strategies compared included no testing, tuberculin skin test (TST), interferon gamma release assay (IGRA), confirm positive (initial TST, IGRA only for TST-positive results; both tests positive indicates LTBI), and confirm negative (initial IGRA, then TST for IGRA-negative; any test positive indicates LTBI). All strategies were coupled to treatment with 3 months of self-administered rifapentine and isoniazid. MAIN OUTCOMES AND MEASURES Number needed to test and treat to prevent 1 case of TB reactivation, discounted quality-adjusted life-years (QALYs), discounted lifetime medical costs, and incremental cost-effectiveness ratios (ICERs). RESULTS Improving health outcomes increased costs, with choice of test dependent on willingness to pay. Strategies ranked by ascending costs and benefits: no testing, confirm positive, TST, IGRA, and confirm negative. The ICERs varied by non-US born patient risk group: patients with no comorbidities, IGRA was likely cost-effective at $83 000/QALY; patients with diabetes, both confirm positive ($53 000/QALY) and IGRA ($120 000/QALY) were likely cost-effective; patients with HIV, confirm negative was clearly preferred ($63 000/QALY); and patients with ESRD, no testing was cost-effective. Increased LTBI prevalence and reduced return for TST reading improved IGRA's relative performance. In 10 000 probabilistic simulations among non-US born patients with no comorbidities, with diabetes, and with HIV, some form of testing was virtually always cost-effective. These simulations highlight the uncertainty of test choice for non-US born patients with no comorbidities and non-US born patients with diabetes, but strategies including IGRA were preferred in over 60% of simulations for all non-US born populations except those with ESRD. CONCLUSIONS AND RELEVANCE Testing for and treating LTBI among non-US born residents with and without selected comorbidities is likely cost-effective except among those with ESRD in whom competing risks of death limit benefits. Strategies including IGRA fell below a $100 000/QALY willingness-to-pay threshold for non-US born patients with no comorbidities, patients with diabetes, and patients with HIV.
Collapse
Affiliation(s)
- Abriana Tasillo
- HIV Epidemiology & Outcomes Unit, Section of Infectious Diseases, Boston Medical Center, Boston, Massachusetts
| | - Joshua A Salomon
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Thomas A Trikalinos
- Center for Evidence-Based Medicine, Brown University, Providence, Rhode Island
| | | | - Suzanne M Marks
- Division of Tuberculosis Elimination, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Benjamin P Linas
- HIV Epidemiology & Outcomes Unit, Section of Infectious Diseases, Boston Medical Center, Boston, Massachusetts.,Boston University School of Public Health, Boston, Massachusetts
| |
Collapse
|
43
|
Zheng C, Hu X, Zhao L, Hu M, Gao F. Clinical and pharmacological hallmarks of rifapentine's use in diabetes patients with active and latent tuberculosis: do we know enough? DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:2957-2968. [PMID: 29066867 PMCID: PMC5644564 DOI: 10.2147/dddt.s146506] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Rifapentine is a rifamycin derivate approved by the US Food and Drug Administration in 1998 for the treatment of active, drug-susceptible tuberculosis (TB). In 2014, rifapentine was approved for the treatment of latent TB infection in patients at high risk of progression to active disease and is currently under evaluation by the European Medicines Agency. Expanding indications of rifapentine largely affect diabetes patients, since about one-third of them harbor latent TB. Clinical consequences of rifapentine use in this population and potentially harmful interactions with hypoglycemic agents are widely underexplored and generally considered similar to the ones of rifampicin. Indeed, rifapentine too may decrease blood levels of many oral antidiabetics and compete with them for protein-binding sites and/or transporters. However, the two drugs differ in protein-binding degree, the magnitude of cytochrome P450 induction and auto-induction, the degree of renal elimination, and so on. Rifapentine seems to be more suitable for use in diabetes patients with renal impairment, owing to the fact that it does not cause renal toxicity, and it is eliminated via kidneys in smaller proportions than rifampicin. On the other hand, there are no data related to rifapentine use in patients >65 years, and hypoalbuminemia associated with diabetic kidney disease may affect a free fraction of rifapentine to a greater extent than that of rifampicin. Until more pharmacokinetic information and information on the safety of rifapentine use in diabetic patients and drug–drug interactions are available, diabetes in TB patients treated with rifapentine should be managed with insulin analogs, and glucose and rifapentine plasma levels should be closely monitored.
Collapse
Affiliation(s)
- Chunlan Zheng
- Department of Internal Medicine - Section 5, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Control Institute)
| | - Xiufen Hu
- Department of Paediatrics, Tongji Hospital
| | - Li Zhao
- Department of Internal Medicine - Section 5, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Control Institute)
| | - Minhui Hu
- Department of Internal Medicine - Section 5, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Control Institute)
| | - Feng Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
44
|
Alfarisi O, Alghamdi WA, Al-Shaer MH, Dooley KE, Peloquin CA. Rifampin vs. rifapentine: what is the preferred rifamycin for tuberculosis? Expert Rev Clin Pharmacol 2017; 10:1027-1036. [PMID: 28803492 DOI: 10.1080/17512433.2017.1366311] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION One-third of the world's population is infected with Mycobacterium tuberculosis (M.tb.). Latent tuberculosis infection (LTBI) can progress to tuberculosis disease, the leading cause of death by infection. Rifamycin antibiotics, like rifampin and rifapentine, have unique sterilizing activity against M.tb. What are the advantages of each for LTBI or tuberculosis treatment? Areas covered: We review studies assessing the pharmacokinetics (PK), pharmacodynamics (PD), drug interaction risk, safety, and efficacy of rifampin and rifapentine and provide basis for comparing them. Expert commentary: Rifampin has shorter half-life, higher MIC against M.tb, lower protein binding, and better distribution into cavitary contents than rifapentine. Drug interactions for the two drugs maybe similar in magnitude. For LTBI, rifapentine is effective as convenient, once-weekly, 12-week course of treatment. Rifampin is also effective for LTBI, but must be given daily for four months, therefore, drug interactions are more problematic. For drug-sensitive tuberculosis disease, rifampin remains the standard of care. Safety profile of rifampin is better-described; adverse events differ somewhat for the two drugs. The registered once-weekly rifapentine regimen is inadequate, but higher doses of either drugs may shorten the treatment duration required for effective management of TB. Results of clinical trials evaluating high-dose rifamycin regimens are eagerly awaited.
Collapse
Affiliation(s)
- Omamah Alfarisi
- a Department of Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Wael A Alghamdi
- b Department of Pharmacotherapy and Translational Research , University of Florida, College of Pharmacy , Gainesville , FL , USA.,c Infectious Disease Pharmacokinetics Laboratory , University of Florida , Gainesville , FL , USA
| | - Mohammad H Al-Shaer
- b Department of Pharmacotherapy and Translational Research , University of Florida, College of Pharmacy , Gainesville , FL , USA.,c Infectious Disease Pharmacokinetics Laboratory , University of Florida , Gainesville , FL , USA
| | - Kelly E Dooley
- a Department of Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Charles A Peloquin
- b Department of Pharmacotherapy and Translational Research , University of Florida, College of Pharmacy , Gainesville , FL , USA.,c Infectious Disease Pharmacokinetics Laboratory , University of Florida , Gainesville , FL , USA
| |
Collapse
|
45
|
Abstract
Tuberculosis (TB) has recently surpassed HIV as the primary infectious disease killer worldwide, but the two diseases continue to display lethal synergy. The burden of TB is disproportionately borne by people living with HIV, particularly where HIV and poverty coexist. The impact of these diseases on one another is bidirectional, with HIV increasing risk of TB infection and disease progression and TB slowing CD4 recovery and increasing progression to AIDS and death among the HIV infected. Both antiretroviral therapy (ART) and latent TB infection (LTBI) treatment mitigate the impact of coinfection, and ART is now recommended for HIV-infected patients independent of CD4 count. LTBI screening should be performed for all HIV-positive people at the time of diagnosis, when their CD4 count rises above 200, and yearly if there is repeated exposure. Tuberculin skin tests (TSTs) may perform better with serial testing than interferon gamma release assays (IGRAs). Any patient with HIV and a TST induration of ≥5 mm should be evaluated for active TB disease and treated for LTBI if active disease is ruled out. Because HIV impairs multiple aspects of immune function, progressive HIV is associated with lower rates of cavitary pulmonary TB and higher rates of disseminated and extrapulmonary disease, so a high index of suspicion is important, and sputum should be obtained for evaluation even if chest radiographs are negative. TB diagnosis is similar in patients with and without TB, relying on smear, culture, and nucleic acid amplification tests, which are the initial tests of choice. TSTs and IGRAs should not be used in the evaluation of active TB disease since these tests are often negative with active disease. Though not always performed in resource-limited settings, drug susceptibility testing should be performed on all TB isolates from HIV-positive patients. Urine lipoarabinomannan testing may also be helpful in HIV-positive patients with disseminated disease. Treatment of TB in HIV-infected patients is similar to that of TB in HIV-negative patients except that daily therapy is required for all coinfected patients, vitamin B6 supplementation should be given to all coinfected patients receiving isoniazid to reduce peripheral neuropathy, and specific attention needs to be paid to drug-drug interactions between rifamycins and many classes of antiretrovirals. In patients requiring ART that contains ritonavir or cobicistat, this can be managed by the use of rifabutin at 150 mg daily in place of rifampin. For newly diagnosed coinfected patients, mortality is lower if treatment is provided in parallel, rather than serially, with treatment initiation within 2 weeks preferred for those with CD4 counts of <50 and within 8 to 12 weeks for those with higher CD4 counts. When TB immune reconstitution inflammatory syndrome occurs, patients can often be treated symptomatically with nonsteroidal anti-inflammatory drugs, but a minority will benefit from steroids. Generally, patients who do not have space-occupying lesions such as occurs in TB meningitis do not require cessation of therapy.
Collapse
|
46
|
Matteelli A, Sulis G, Capone S, D'Ambrosio L, Migliori GB, Getahun H. Tuberculosis elimination and the challenge of latent tuberculosis. Presse Med 2017; 46:e13-e21. [PMID: 28279508 DOI: 10.1016/j.lpm.2017.01.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/17/2017] [Indexed: 12/18/2022] Open
Abstract
Latent tuberculosis infection (LTBI) affects one third to one fourth of the human population and is the reservoir for a significant proportion of emerging active tuberculosis (TB) cases, especially in low incidence countries. The World Health Organization launched in 2015 the END-TB strategy that aims at TB elimination and promotes, for the first time ever, the management of LTBI. The preventive package, basically consisting of testing and treatment for LTBI in groups at high risk of reactivation, is a mainstay of the first pillar of the strategy, alongside prompt diagnosis and early treatment of both drug-susceptible and drug-resistant TB disease. Testing and treatment for LTBI should be pursued with a programmatic perspective. This implies strong political commitment, adequate funding and an effective monitoring and evaluation system. People living with HIV and children under five years of age who are household contact of a contagious TB cases are primarily targeted in all epidemiological setting. In high resource and low incidence setting, additional at risk populations should also be the target for systematic LTBI testing and treatment. Research is urgently needed to develop diagnostic tests with higher predictive value to identify individuals that progress from infection to disease. Similarly, shorter and safer treatment regimens are needed to make the trade-off between potential benefits and harms more favourable for an increasing proportion of infected individuals.
Collapse
Affiliation(s)
- Alberto Matteelli
- University of Brescia, WHO Collaborating Centre for TB/HIV co-infection and TB Elimination, Department of Infectious and Tropical Diseases, Brescia, Italy.
| | - Giorgia Sulis
- University of Brescia, WHO Collaborating Centre for TB/HIV co-infection and TB Elimination, Department of Infectious and Tropical Diseases, Brescia, Italy
| | - Susanna Capone
- University of Brescia, WHO Collaborating Centre for TB/HIV co-infection and TB Elimination, Department of Infectious and Tropical Diseases, Brescia, Italy
| | - Lia D'Ambrosio
- Maugeri Care and Research Institute, WHO Collaborating Centre for Tuberculosis and Lung Diseases, Tradate, Italy; Public Health Consulting Group, Lugano, Switzerland
| | - Giovanni Battista Migliori
- Maugeri Care and Research Institute, WHO Collaborating Centre for Tuberculosis and Lung Diseases, Tradate, Italy
| | - Haileyesus Getahun
- World Health Organization, Global Tuberculosis Programme, Geneva, Switzerland
| |
Collapse
|
47
|
Egelund EF, Dupree L, Huesgen E, Peloquin CA. The pharmacological challenges of treating tuberculosis and HIV coinfections. Expert Rev Clin Pharmacol 2016; 10:213-223. [PMID: 27828731 DOI: 10.1080/17512433.2017.1259066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Tuberculosis (TB) is the most prevalent opportunistic infection among HIV patients, and the leading cause of death among HIV patients worldwide. Simultaneous treatment of both diseases is recommended by current guidelines, but can be challenging due to the potential for drug-drug interactions, overlapping toxicities, difficulty adhering to medications, and an increased risk for immune reconstitution inflammatory syndrome (IRIS). Clinical manifestations of TB can also vary between HIV-infected patients and uninfected patients, which can increase the risk for delayed diagnosis. Areas covered: Topics covered in this review include the following: the inter-related pathophysiology of HIV and TB; clinical manifestations and diagnosis; drug-drug interactions, particularly the rifamycins with the antiretrovirals; IRIS presentation and treatment, as well as a discussion on overlapping toxicity between the two disease states. Expert commentary: The complexity of managing these two disease states simultaneously requires a multidisciplinary approach to care and dedicated resources. If properly funded, TB/HIV co-infection will continue to decline over the coming years.
Collapse
Affiliation(s)
- Eric F Egelund
- a Department of Pharmacotherapy and Translational Research , College of Pharmacy.,b Infectious Disease Pharmacokinetics Laboratory
| | - Lori Dupree
- a Department of Pharmacotherapy and Translational Research , College of Pharmacy
| | - Emily Huesgen
- a Department of Pharmacotherapy and Translational Research , College of Pharmacy
| | - Charles A Peloquin
- a Department of Pharmacotherapy and Translational Research , College of Pharmacy.,b Infectious Disease Pharmacokinetics Laboratory.,c Emerging Pathogens Institute , University of Florida , Gainesville , FL , USA
| |
Collapse
|
48
|
Abstract
Tuberculosis (TB) has been a leading cause of death for more than a century. While effective therapies exist, treatment is long and cumbersome. TB control is complicated by the overlapping problems created by global inadequacy of public health infrastructures, the interaction of the TB and human immunodeficiency virus (HIV) epidemics, and the emergence of drug-resistant TB. After a long period of neglect, there is now significant progress in the development of novel treatment regimens for TB. Focusing on treatment for active disease, we review pathways to TB regimen development and the new and repurposed anti-TB agents in clinical development.
Collapse
Affiliation(s)
- Anthony T Podany
- College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Susan Swindells
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
49
|
Abstract
INTRODUCTION The goal of this article is to review the use of rifapentine in the treatment of latent tuberculosis infection (LTBI). Controlling LTBI is an important part of the global strategy to end the spread of tuberculosis. Rifapentine's potent sterilizing effect against Mycobacterium tuberculosis combined with its long half-life make it an attractive LTBI treatment option. Areas covered: A systematic literature search of Pubmed using the terms 'rifapentine' and 'tuberculosis' was performed. Articles identified were cross-referenced for other relevant publications. The mechanisms of action and resistance, pharmacokinetic and pharmacodynamics, potential drug interactions and side effects are discussed. Expert commentary: Rifapentine in combination with isoniazid for twelve weeks is the best available option for treating latent TB in the majority of patients in the United States due to its favorable safety profile and the increased likelihood of completing therapy. Currently, rifapentine is not registered or available in other countries.
Collapse
Affiliation(s)
- Eric F Egelund
- a Department of Pharmacotherapy and Translational Research, College of Pharmacy , University of Florida , Gainesville , FL , USA.,b Infectious Disease Pharmacokinetics Laboratory , University of Florida , Gainesville , FL , USA
| | - Charles A Peloquin
- a Department of Pharmacotherapy and Translational Research, College of Pharmacy , University of Florida , Gainesville , FL , USA.,b Infectious Disease Pharmacokinetics Laboratory , University of Florida , Gainesville , FL , USA.,c Emerging Pathogens Institute , University of Florida , Gainesville , FL , USA
| |
Collapse
|
50
|
Three months of weekly rifapentine and isoniazid for treatment of Mycobacterium tuberculosis infection in HIV-coinfected persons. AIDS 2016; 30:1607-15. [PMID: 27243774 DOI: 10.1097/qad.0000000000001098] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Compare the effectiveness, tolerability, and safety of 3 months of weekly rifapentine and isoniazid under direct observation (3HP) versus 9 months of daily isoniazid (9H) in HIV-infected persons. DESIGN Prospective, randomized, and open-label noninferiority trial. SETTING The United States , Brazil, Spain, Peru, Canada, and Hong Kong. PARTICIPANTS HIV-infected persons who were tuberculin skin test positive or close contacts of tuberculosis cases. INTERVENTION 3HP versus 9H. MAIN OUTCOME MEASURES The effectiveness endpoint was tuberculosis; the noninferiority margin was 0.75%. The tolerability endpoint was treatment completion; the safety endpoint was drug discontinuation because of adverse drug reaction. RESULTS Median baseline CD4 cell counts were 495 (IQR 389-675) and 538 (IQR 418-729) cells/μl in the 3HP and 9H arms, respectively (P = 0.09). In the modified intention-to-treat analysis, there were two tuberculosis cases among 206 persons [517 person-years (p-y) of follow-up] in the 3HP arm (0.39 per 100 p-y) and six tuberculosis cases among 193 persons (481 p-y of follow-up) in the 9H arm (1.25 per 100 p-y). Cumulative tuberculosis rates were 1.01 versus 3.50% in the 3HP and 9H arms, respectively (rate difference: -2.49%; upper bound of the 95% confidence interval of the difference: 0.60%). Treatment completion was higher with 3HP (89%) than 9H (64%) (P < 0.001), and drug discontinuation because of an adverse drug reaction was similar (3 vs. 4%; P = 0.79) in 3HP and 9H, respectively. CONCLUSION Among HIV-infected persons with median CD4 cell count of approximately 500 cells/μl, 3HP was as effective and safe for treatment of latent Mycobacterium tuberculosis infection as 9H, and better tolerated.
Collapse
|