1
|
Grant-McAuley W, Morgenlander WR, Ruczinski I, Kammers K, Laeyendecker O, Hudelson SE, Thakar M, Piwowar-Manning E, Clarke W, Breaud A, Ayles H, Bock P, Moore A, Kosloff B, Shanaube K, Meehan SA, van Deventer A, Fidler S, Hayes R, Larman HB, Eshleman SH, for the HPTN 071 (PopART) Study Team. Identification of antibody targets associated with lower HIV viral load and viremic control. PLoS One 2024; 19:e0305976. [PMID: 39288118 PMCID: PMC11407625 DOI: 10.1371/journal.pone.0305976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/09/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND High HIV viral loads (VL) are associated with increased morbidity, mortality, and on-going transmission. HIV controllers maintain low VLs in the absence of antiretroviral therapy (ART). We previously used a massively multiplexed antibody profiling assay (VirScan) to compare antibody profiles in HIV controllers and persons living with HIV (PWH) who were virally suppressed on ART. In this report, we used VirScan to evaluate whether antibody reactivity to specific HIV targets and broad reactivity across the HIV genome was associated with VL and controller status 1-2 years after infection. METHODS Samples were obtained from participants who acquired HIV infection in a community-randomized trial in Africa that evaluated an integrated strategy for HIV prevention (HPTN 071 PopART). Controller status was determined using VL and antiretroviral (ARV) drug data obtained at the seroconversion visit and 1 year later. Viremic controllers had VLs <2,000 copies/mL at both visits; non-controllers had VLs >2,000 copies/mL at both visits. Both groups had no ARV drugs detected at either visit. VirScan testing was performed at the second HIV-positive visit (1-2 years after HIV infection). RESULTS The study cohort included 13 viremic controllers and 64 non-controllers. We identified ten clusters of homologous peptides that had high levels of antibody reactivity (three in gag, three in env, two in integrase, one in protease, and one in vpu). Reactivity to 43 peptides (eight unique epitopes) in six of these clusters was associated with lower VL; reactivity to six of the eight epitopes was associated with HIV controller status. Higher aggregate antibody reactivity across the eight epitopes (more epitopes targeted, higher mean reactivity across all epitopes) and across the HIV genome was also associated with lower VL and controller status. CONCLUSIONS We identified HIV antibody targets associated with lower VL and HIV controller status 1-2 years after infection. Robust aggregate responses to these targets and broad antibody reactivity across the HIV genome were also associated with lower VL and controller status. These findings provide novel insights into the relationship between humoral immunity and viral containment that could help inform the design of antibody-based approaches for reducing HIV VL.
Collapse
Affiliation(s)
- Wendy Grant-McAuley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William R. Morgenlander
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Kai Kammers
- Quantitative Sciences Division, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Oliver Laeyendecker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Sarah E. Hudelson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Manjusha Thakar
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Estelle Piwowar-Manning
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Helen Ayles
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Peter Bock
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - Ayana Moore
- FHI 360, Durham, North Carolina, United States of America
| | - Barry Kosloff
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Kwame Shanaube
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
| | - Sue-Ann Meehan
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - Anneen van Deventer
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Richard Hayes
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - H. Benjamin Larman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | | |
Collapse
|
2
|
Jia W, Ouyang Y, Zhang S, Du X, Zhang P, Huang S. Nanopore Signatures of Nucleoside Drugs. NANO LETTERS 2023; 23:9437-9444. [PMID: 37818841 DOI: 10.1021/acs.nanolett.3c02872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Nucleoside drugs, which are analogues of natural nucleosides, have been widely applied in the clinical treatment of viral infections and cancers. The development of nucleoside drugs, repurposing of existing drugs, and combined use of multiple drug types have made the rapid sensing of nucleoside drugs urgently needed. Nanopores are emerging single-molecule sensors that have high resolution to resolve even minor structural differences between chemical compounds. Here, an engineered Mycobacterium smegmatis porin A hetero-octamer was used to perform general nucleoside drug analysis. Ten nucleoside drugs were simultaneously detected and fully discriminated. An accuracy of >99.9% was consequently reported. This sensing capacity was further demonstrated in direct nanopore analysis of ribavirin buccal tablets, confirming its sensing reliability against complex samples and environments. No sample separation is needed, however, significantly minimizing the complexity of the measurement. This technique may inspire nanopore applications in pharmaceutical production and pharmacokinetics measurements.
Collapse
Affiliation(s)
- Wendong Jia
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023 Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023 Nanjing, China
| | - Yusheng Ouyang
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023 Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023 Nanjing, China
| | - Shanyu Zhang
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023 Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023 Nanjing, China
| | - Xiaoyu Du
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023 Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023 Nanjing, China
| | - Panke Zhang
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023 Nanjing, China
| | - Shuo Huang
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023 Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023 Nanjing, China
| |
Collapse
|
3
|
Grant‐McAuley W, Piwowar‐Manning E, Clarke W, Breaud A, Zewdie KB, Moore A, Ayles HM, Kosloff B, Shanaube K, Bock P, Meehan S, Maarman G, Fidler S, Hayes R, Donnell D, Eshleman SH, for the HPTN 071 (PopART) Study Team. Population-level analysis of natural control of HIV infection in Zambia and South Africa: HPTN 071 (PopART). J Int AIDS Soc 2023; 26:e26179. [PMID: 37886843 PMCID: PMC10603557 DOI: 10.1002/jia2.26179] [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: 01/10/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
INTRODUCTION HIV controllers have low viral loads (VL) without antiretroviral treatment (ART). We evaluated viraemic control in a community-randomized trial conducted in Zambia and South Africa that evaluated the impact of a combination prevention intervention on HIV incidence (HPTN 071 [PopART]; 2013-2018). METHODS VL and antiretroviral (ARV) drug testing were performed using plasma samples collected 2 years after enrolment for 4072 participants who were HIV positive at the start of the study intervention. ARV drug use was assessed using a qualitative laboratory assay that detects 22 ARV drugs in five drug classes. Participants were classified as non-controllers if they had a VL ≥2000 copies/ml with no ARV drugs detected at this visit. Additional VL and ARV drug testing was performed at a second annual study visit to confirm controller status. Participants were classified as controllers if they had VLs <2000 with no ARV drugs detected at both visits. Non-controllers who had ARV drugs detected at either visit were excluded from the analysis to minimize potential confounders associated with ARV drug access and uptake. RESULTS The final cohort included 126 viraemic controllers and 766 non-controllers who had no ARV drugs detected. The prevalence of controllers among the 4072 persons assessed was 3.1% (95% confidence interval [CI]: 2.6%, 3.6%). This should be considered a minimum estimate, since high rates of ARV drug use in the parent study limited the ability to identify controllers. Among the 892 participants in the final cohort, controller status was associated with biological sex (female > male, p = 0.027). There was no significant association between controller status and age, study country or herpes simplex virus type 2 (HSV-2) status at study enrolment. CONCLUSIONS To our knowledge, this report presents the first large-scale, population-level study evaluating the prevalence of viraemic control and associated factors in Africa. A key advantage of this study was that a biomedical assessment was used to assess ARV drug use (vs. self-reported data). This study identified a large cohort of HIV controllers and non-controllers not taking ARV drugs, providing a unique repository of longitudinal samples for additional research. This cohort may be useful for further studies investigating the mechanisms of virologic control.
Collapse
Affiliation(s)
- Wendy Grant‐McAuley
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | | | - William Clarke
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Autumn Breaud
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | | | | | - Helen Mary Ayles
- ZambartUniversity of Zambia School of Public HealthLusakaZambia
- Clinical Research DepartmentLondon School of Hygiene and Tropical MedicineLondonUK
| | - Barry Kosloff
- ZambartUniversity of Zambia School of Public HealthLusakaZambia
- Clinical Research DepartmentLondon School of Hygiene and Tropical MedicineLondonUK
| | - Kwame Shanaube
- ZambartUniversity of Zambia School of Public HealthLusakaZambia
| | - Peter Bock
- Desmond Tutu TB CenterDepartment of Paediatrics and Child HealthStellenbosch UniversityWestern CapeSouth Africa
| | - Sue‐Ann Meehan
- Desmond Tutu TB CenterDepartment of Paediatrics and Child HealthStellenbosch UniversityWestern CapeSouth Africa
| | - Gerald Maarman
- Centre for Cardio‐Metabolic Research in AfricaDivision of Medical PhysiologyFaculty of Medicine and Health SciencesStellenbosch UniversityWestern CapeSouth Africa
| | - Sarah Fidler
- Department of Infectious DiseaseImperial College LondonLondonUK
| | - Richard Hayes
- Department of Infectious Disease EpidemiologyLondon School of Hygiene and Tropical MedicineLondonUK
| | | | - Susan H. Eshleman
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | | |
Collapse
|
4
|
Grant-McAuley W, Morgenlander W, Hudelson SE, Thakar M, Piwowar-Manning E, Clarke W, Breaud A, Blankson J, Wilson E, Ayles H, Bock P, Moore A, Kosloff B, Shanaube K, Meehan SA, van Deventer A, Fidler S, Hayes R, Ruczinski I, Kammers K, Laeyendecker O, Larman HB, Eshleman SH. Comprehensive profiling of pre-infection antibodies identifies HIV targets associated with viremic control and viral load. Front Immunol 2023; 14:1178520. [PMID: 37744365 PMCID: PMC10512082 DOI: 10.3389/fimmu.2023.1178520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 08/15/2023] [Indexed: 09/26/2023] Open
Abstract
Background High HIV viral load (VL) is associated with increased transmission risk and faster disease progression. HIV controllers achieve viral suppression without antiretroviral (ARV) treatment. We evaluated viremic control in a community-randomized trial with >48,000 participants. Methods A massively multiplexed antibody profiling system, VirScan, was used to quantify pre- and post-infection antibody reactivity to HIV peptides in 664 samples from 429 participants (13 controllers, 135 viremic non-controllers, 64 other non-controllers, 217 uninfected persons). Controllers had VLs <2,000 copies/mL with no ARV drugs detected at the first HIV-positive visit and one year later. Viremic non-controllers had VLs 2,000 copies/mL with no ARV drugs detected at the first HIV-positive visit. Other non-controllers had either ARV drugs detected at the first HIV-positive visit (n=47) or VLs <2,000 copies/mL with no ARV drugs detected at only one HIV-positive visit (n=17). Results We identified pre-infection HIV antibody reactivities that correlated with post-infection VL. Pre-infection reactivity to an epitope in the HR2 domain of gp41 was associated with controller status and lower VL. Pre-infection reactivity to an epitope in the C2 domain of gp120 was associated with non-controller status and higher VL. Different patterns of antibody reactivity were observed over time for these two epitopes. Conclusion These studies suggest that pre-infection HIV antibodies are associated with controller status and modulation of HIV VL. These findings may inform research on antibody-based interventions for HIV treatment.
Collapse
Affiliation(s)
- Wendy Grant-McAuley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - William Morgenlander
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah E. Hudelson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Manjusha Thakar
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Estelle Piwowar-Manning
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Joel Blankson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ethan Wilson
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Helen Ayles
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Peter Bock
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | | | - Barry Kosloff
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Kwame Shanaube
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
| | - Sue-Ann Meehan
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | - Anneen van Deventer
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Richard Hayes
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kai Kammers
- Quantitative Sciences Division, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Oliver Laeyendecker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Baltimore, MD, United States
| | - H. Benjamin Larman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
5
|
Fogel JM, Zewdie K, Clarke WA, Piwowar-Manning E, Breaud A, Moore A, Kosloff B, Shanaube K, van Zyl G, Scheepers M, Floyd S, Bock P, Ayles H, Fidler S, Hayes R, Donnell D, Eshleman SH, for the HPTN 071 (PopART) Study Team. Antiretroviral Drug Detection in a Community-Randomized Trial of Universal HIV Testing and Treatment: HPTN 071 (PopART). Open Forum Infect Dis 2022; 9:ofac576. [PMID: 36447611 PMCID: PMC9697607 DOI: 10.1093/ofid/ofac576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/28/2022] [Indexed: 08/19/2023] Open
Abstract
Background Antiretroviral therapy (ART) reduces human immunodeficiency virus (HIV) transmission risk. The primary aim of this study was to evaluate ART uptake in a trial in Zambia and South Africa that implemented a community-wide universal testing and treatment package to reduce HIV incidence. Methods Study communities were randomized to 3 arms: A, combination-prevention intervention with universal ART; B, combination-prevention intervention with ART according to local guidelines; and C, standard of care. Samples were collected from people with HIV (PWH) during a survey visit conducted 2 years after study implementation: these samples were tested for 22 antiretroviral (ARV) drugs. Antiretroviral therapy uptake was defined as detection of ≥1 ARV drug. Resistance was evaluated in 612 randomly selected viremic participants. A 2-stage, cluster-based approach was used to assess the impact of the study intervention on ART uptake. Results Antiretroviral drugs were detected in 4419 of 6207 (71%) samples (Arm A, 73%; Arm B, 70%; Arm C, 60%); 4140 (94%) of samples with ARV drugs had viral loads <400 copies/mL. Drug resistance was observed in 237 of 612 (39%) viremic participants (95 of 102 [93%] with ARV drugs; 142 of 510 [28%] without drugs). Antiretroviral therapy uptake was associated with older age, female sex, enrollment year, seroconverter status, and self-reported ART (all P < .001). The adjusted risk ratio for ART uptake was similar for Arm A versus C (1.21; 95% confidence interval [CI], .94-1.54; P = .12) and Arm B versus C (1.14; 95% CI, .89-1.46; P = .26). Conclusions At the 2-year survey, 71% of PWH were on ART and 94% of those participants were virally suppressed. Universal testing and treatment was not significantly associated with increased ART uptake in this cohort.
Collapse
Affiliation(s)
- Jessica M Fogel
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kidist Zewdie
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - William A Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Estelle Piwowar-Manning
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Barry Kosloff
- Zambart, University of Zambia, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Gert van Zyl
- Division of Medical Virology, Stellenbosch University, Cape Town, South Africa
| | - Michelle Scheepers
- Department of Paediatrics and Child Health, Desmond Tutu TB Center, Stellenbosch University, Western Cape, South Africa
| | - Sian Floyd
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Peter Bock
- Department of Paediatrics and Child Health, Desmond Tutu TB Center, Stellenbosch University, Western Cape, South Africa
| | - Helen Ayles
- Zambart, University of Zambia, Lusaka, Zambia
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Richard Hayes
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Deborah Donnell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Susan H Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
6
|
Gawler N, Reynolds SJ, Hsiao NY, Clarke W, Maartens G, Abrams EJ, Myer L, Redd AD, Phillips TK. Routine Antiretroviral Pharmacy Refill Information Can Predict Failure Postpartum in Previously Suppressed South African Women With HIV. Open Forum Infect Dis 2022; 9:ofac483. [PMID: 36275867 PMCID: PMC9578152 DOI: 10.1093/ofid/ofac483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/14/2022] [Indexed: 11/14/2022] Open
Abstract
Background Detection of antiretrovirals (ARVs) in biological specimens is a reliable, objective way to measure adherence. However, routine ARV testing is not feasible in many high-burden settings. This study explored if pharmacy data could accurately predict HIV viremia postpartum in previously virally suppressed women. Methods South African women with HIV who initiated antiretroviral therapy (ART) during pregnancy and achieved viral suppression (VS; viral load [VL]≤50 copies/mL) were followed postpartum; during follow-up, plasma VL was measured and ARV adherence self-reported. A portion of samples were tested for the presence of ARV using mass spectrometry. Patient-level routine pharmacy data were used to classify if women should have the drug in hand for the past 7 days before the visit date. Logistic regressions were used to calculate associations between adherence and viral nonsuppression (VNS; VL > 50) or failure (VF; VL > 1000) at the first study visit of women who had ARV measured. Data for all women were examined for associations of self-reported adherence and drug in hand with VS and VF at 2, 6, and 12 months postpartum. Results Women with no ARV detected were significantly more likely to have VNS (odds ratio [OR], 26.4). Having no drug in hand for 7 days was also predictive of VNS in these same women (OR, 7.0) and the full cohort (n = 572) at 3 (OR, 2.9), 6 (OR, 8.7), and 12 months (OR, 14.5). Similar results were seen for VF. Conclusions These data show that routine pharmacy data can act as a highly predictive mechanism for identifying patients at risk of VNS and VF due to nonadherence.
Collapse
Affiliation(s)
- Nicola Gawler
- Division of Epidemiology & Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
| | - Steven J Reynolds
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Division of Intramural Research, NIAID, NIH, Baltimore, Maryland, USA
- Rakai Health Sciences Program, Entebbe, Uganda
| | - Nei-Yuan Hsiao
- Division of Medical Virology, University of Cape Town and National Health Laboratory Service, Cape Town, South Africa
| | - William Clarke
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gary Maartens
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Elaine J Abrams
- ICAP at Columbia University, Mailman School of Public Health, and Department of Paediatrics, Vagelos College of Physicians & Surgeons, Columbia University, New York, New York, USA
| | - Landon Myer
- Division of Epidemiology & Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
| | - Andrew D Redd
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Division of Intramural Research, NIAID, NIH, Baltimore, Maryland, USA
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Tamsin K Phillips
- Division of Epidemiology & Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
7
|
Jahun I, Ehoche A, Bamidele M, Yakubu A, Bronson M, Dalhatu I, Greby S, Agbakwuru C, Baffa I, Iwara E, Alagi M, Asaolu O, Mukhtar A, Ikpeazu A, Nzelu C, Tapdiyel J, Bassey O, Abimiku A, Patel H, Parekh B, Aliyu S, Aliyu G, Charurat M, Swaminathan M. Evaluation of accuracy and performance of self-reported HIV and antiretroviral therapy status in the Nigeria AIDS Indicator and Impact Survey (2018). PLoS One 2022; 17:e0273748. [PMID: 36037201 PMCID: PMC9423665 DOI: 10.1371/journal.pone.0273748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 08/15/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Data on awareness of HIV status among people living with HIV (PLHIV) are critical to estimating progress toward epidemic control. To ascertain the accuracy of self-reported HIV status and antiretroviral drug (ARV) use in the Nigeria HIV/AIDS Indicator and Impact Survey (NAIIS), we compared self-reported HIV status with HIV rapid diagnostic test (RDT) results and self-reported ARV use with detectable blood ARV levels. METHODS On the basis of responses and test results, participants were categorized by HIV status and ARV use. Self-reported HIV status and ARV use performance characteristics were determined by estimating sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV). Proportions and other analyses were weighted to account for complex survey design. RESULTS During NAIIS, 186,405 participants consented for interview out of which 58,646 reported knowing their HIV status. Of the 959 (weighted, 1.5%) who self-reported being HIV-positive, 849 (92.1%) tested HIV positive and 64 (7.9%) tested HIV negative via RDT and polymerase chain reaction test for discordant positive results. Of the 849 who tested HIV positive, 743 (89.8%) reported using ARV and 72 (10.2%) reported not using ARV. Of 57,687 who self-reported being HIV negative, 686 (1.2%) tested HIV positive via RDT, with ARV biomarkers detected among 195 (25.1%). ARV was detected among 94.5% of those who self-reported using ARV and among 42.0% of those who self-reported not using ARV. Overall, self-reported HIV status had sensitivity of 52.7% (95% confidence interval [CI]: 49.4%-56.0%) with specificity of 99.9% (95% CI: 99.8%-99.9%). Self-reported ARV use had sensitivity of 95.2% (95% CI: 93.6%-96.7%) and specificity of 54.5% (95% CI: 48.8%-70.7%). CONCLUSIONS Self-reported HIV status and ARV use screening tests were found to be low-validity measures during NAIIS. Laboratory tests to confirm self-reported information may be necessary to determine accurate HIV and clinical status for HIV studies in Nigeria.
Collapse
Affiliation(s)
- Ibrahim Jahun
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Akipu Ehoche
- Maryland Global Initiatives, Abuja, Federal Capital Territory, Nigeria
| | - Moyosola Bamidele
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Aminu Yakubu
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Megan Bronson
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health Atlanta, GA, United States of America
| | - Ibrahim Dalhatu
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Stacie Greby
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Chinedu Agbakwuru
- Maryland Global Initiatives, Abuja, Federal Capital Territory, Nigeria
| | - Ibrahim Baffa
- Maryland Global Initiatives, Abuja, Federal Capital Territory, Nigeria
| | - Emem Iwara
- Maryland Global Initiatives, Abuja, Federal Capital Territory, Nigeria
| | - Matthias Alagi
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Olugbenga Asaolu
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Ahmed Mukhtar
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Akudo Ikpeazu
- Federal Ministry of Health, Abuja, Federal Capital Territory, Nigeria
| | - Charles Nzelu
- Federal Ministry of Health, Abuja, Federal Capital Territory, Nigeria
| | - Jelpe Tapdiyel
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Orji Bassey
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| | - Alash’le Abimiku
- Maryland Global Initiatives, Abuja, Federal Capital Territory, Nigeria
| | - Hetal Patel
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health Atlanta, GA, United States of America
| | - Bharat Parekh
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health Atlanta, GA, United States of America
| | - Sani Aliyu
- National Agency for the Control of AIDS, Abuja, Federal Capital Territory, Nigeria
| | - Gambo Aliyu
- Maryland Global Initiatives, Abuja, Federal Capital Territory, Nigeria
| | | | - Mahesh Swaminathan
- Centers for Disease Control and Prevention, Division of Global HIV and TB, Center for Global Health-Nigeria, Abuja, Federal Capital Territory, Nigeria
| |
Collapse
|
8
|
Fogel JM, Wilson EA, Piwowar-Manning E, Breaud A, Clarke W, Petropoulos C, Moore A, Fraser C, Kosloff B, Shanaube K, van Zyl G, Scheepers M, Floyd S, Bock P, Ayles H, Fidler S, Hayes R, Donnell D, Eshleman SH. HIV drug resistance in a community-randomized trial of universal testing and treatment: HPTN 071 (PopART). J Int AIDS Soc 2022; 25:e25941. [PMID: 35775502 PMCID: PMC9248006 DOI: 10.1002/jia2.25941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 05/19/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Universal HIV testing and treatment (UTT) has individual and public health benefits. HPTN 071 (PopART), a community-randomized trial in Zambia and South Africa, demonstrated that UTT decreased HIV incidence. This endpoint was assessed in a cohort of >48,000 randomly selected adults in the study communities. We evaluated the impact of UTT on HIV drug resistance in this cohort and compared other resistance-related outcomes in participants with recent versus non-recent HIV infection. METHODS Two years after the start of HPTN 071 (2016-2017), 6259 participants were HIV positive and 1902 were viremic (viral load >400 copies/ml). HIV genotyping and antiretroviral (ARV) drug testing were performed for viremic participants in three groups: seroconverters (infected <1 year), non-seroconverters (infected >1 year, random subset) and participants with unknown duration of infection (random subset). A two-stage cluster-based approach was used to assess the impact of the study intervention on drug resistance. Treatment failure was defined as being viremic with ARV drugs detected. Participants were classified as ARV naïve based on self-report and ARV drug testing. RESULTS Genotyping results were obtained for 758 participants (143 seroconverters; 534 non-seroconverters; and 81 unknown duration of infection). The estimated prevalence of resistance in the study communities was 37% for all viremic persons and 11% for all HIV-positive persons. There was no association between UTT and drug resistance. Resistance was detected in 14.0% of seroconverters and 40.8% of non-seroconverters (non-nucleoside reverse transcriptase inhibitor resistance: 14.0% and 39.9%; nucleoside/nucleotide reverse transcriptase inhibitor resistance: 0.7% and 15.5%; protease inhibitor resistance: 0% and 1.9%; multi-class resistance: 0.7% and 16.1%, respectively). ARV drugs were detected in 2/139 (1.4%) of seroconverters and 94/534 (17.6%) of non-seroconverters tested. These participants were classified as failing ART; 88 (93.6%) of the non-seroconverters failing ART had resistance. Mutations used for surveillance of transmitted drug resistance were detected in 10.5% of seroconverters and 15.1% of non-seroconverters who were ARV naive. CONCLUSIONS UTT was not associated with an increase in drug resistance in this cohort. Higher rates of drug resistance and multi-class resistance were observed in non-seroconverters compared to seroconverters.
Collapse
Affiliation(s)
- Jessica M Fogel
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ethan A Wilson
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Estelle Piwowar-Manning
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Christophe Fraser
- The Big Data Institute and Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Barry Kosloff
- Zambart, Lusaka, Zambia.,Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Gert van Zyl
- Division of Medical Virology, Stellenbosch University, Cape Town, South Africa
| | - Michelle Scheepers
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | - Sian Floyd
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Peter Bock
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | - Helen Ayles
- Zambart, Lusaka, Zambia.,Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Sarah Fidler
- Department of Infectious Disease, HIV Clinical Trials Unit, Imperial College London, London, UK
| | - Richard Hayes
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Deborah Donnell
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Susan H Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
9
|
Abstract
Nucleoside analogues are reagents that resemble the structure of natural nucleosides and are widely applied in antiviral and anticancer therapy. Molnupiravir, a recently reported nucleoside analogue drug, has shown its inhibitory effect against SARS-CoV-2. Rapid tracing of molnupiravir and its metabolites is important in the evaluation of its pharmacology effect, but direct sensing of molnupiravir as a single molecule has not been reported to date. Here, we demonstrate a nanopore-based sensor with which direct sensing of molnupiravir and its two major metabolites β-d-N4-hydroxycytidine and its triphosphate can be achieved simultaneously. In conjunction with a custom machine learning algorithm, an accuracy of 92% was achieved. This sensing strategy may be useful in the current pandemic and is in principle suitable for other nucleoside analogue drugs.
Collapse
Affiliation(s)
- Wendong Jia
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, China
| | - Chengzhen Hu
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, China
| | - Yuqin Wang
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, China
| | - Panke Zhang
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, China
| | - Hong-Yuan Chen
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, China
| | - Shuo Huang
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, China
| |
Collapse
|
10
|
Mosekiemang TT, Stander MA, de Villiers A. Ultra-high pressure liquid chromatography coupled to travelling wave ion mobility-time of flight mass spectrometry for the screening of pharmaceutical metabolites in wastewater samples: Application to antiretrovirals. J Chromatogr A 2021; 1660:462650. [PMID: 34788673 DOI: 10.1016/j.chroma.2021.462650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 10/20/2022]
Abstract
The presence of pharmaceutical compounds in the aquatic environment is a significant environmental health concern, which is exacerbated by recent evidence of the contribution of drug metabolites to the overall pharmaceutical load. In light of a recent report of the occurrence of metabolites of antiretroviral drugs (ARVDs) in wastewater, we investigate in the present work the occurrence of further ARVD metabolites in samples obtained from a domestic wastewater treatment plant in the Western Cape, South Africa. Pharmacokinetic data indicate that ARVDs are biotransformed into several positional isomeric metabolites, only two of which have been reported wastewater samples. Given the challenges associated with the separation and identification of isomeric species in complex wastewater samples, a method based on liquid chromatography hyphenated to ion mobility spectrometry-high resolution mass spectrometry (LC-IMS-HR-MS) was implemented. Gradient LC separation was achieved on a sub-2 µm reversed phase column, while the quadrupole-time-of-flight MS was operated in data independent acquisition (DIA) mode to increase spectral coverage of detected features. A mass defect filter (MDF) template was implemented to detect ARVD metabolites with known phase I and phase II mass shifts and fractional mass differences and to filter out potential interferents. IMS proved particularly useful in filtering the MS data for co-eluting species according to arrival time to provide cleaner mass spectra. This approach allowed us to confirm the presence of two known hydroxylated efavirenz and nevirapine metabolites using authentic standards, and to tentatively identify a carboxylate metabolite of abacavir previously reported in literature. Furthermore, three hydroxylated-, two sulphated and one glucuronidated metabolite of efavirenz, two hydroxylated metabolites of nevirapine and one hydroxylated metabolite of ritonavir were tentatively or putatively identified in wastewater samples for the first time. Assignment of the metabolites is discussed in terms of high resolution fragmentation data, while collisional cross section (CCS) values measured for the detected analytes are reported to facilitate further work in this area.
Collapse
Affiliation(s)
- Tlou T Mosekiemang
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland 7602, South Africa
| | - Maria A Stander
- Central Analytical Facility, Stellenbosch University, Private Bag X1, Matieland 7602, South Africa
| | - André de Villiers
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland 7602, South Africa.
| |
Collapse
|
11
|
Grant-McAuley W, Klock E, Laeyendecker O, Piwowar-Manning E, Wilson E, Clarke W, Breaud A, Moore A, Ayles H, Kosloff B, Shanaube K, Bock P, Mandla N, van Deventer A, Fidler S, Donnell D, Hayes R, Eshleman SH, for the HPTN 071 (PopART) Study Team. Evaluation of multi-assay algorithms for identifying individuals with recent HIV infection: HPTN 071 (PopART). PLoS One 2021; 16:e0258644. [PMID: 34919554 PMCID: PMC8682874 DOI: 10.1371/journal.pone.0258644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 10/01/2021] [Indexed: 11/18/2022] Open
Abstract
Background
Assays and multi-assay algorithms (MAAs) have been developed for population-level cross-sectional HIV incidence estimation. These algorithms use a combination of serologic and/or non-serologic biomarkers to assess the duration of infection. We evaluated the performance of four MAAs for individual-level recency assessments.
Methods
Samples were obtained from 220 seroconverters (infected <1 year) and 4,396 non-seroconverters (infected >1 year) enrolled in an HIV prevention trial (HPTN 071 [PopART]); 28.6% of the seroconverters and 73.4% of the non-seroconverters had HIV viral loads ≤400 copies/mL. Samples were tested with two laboratory-based assays (LAg-Avidity, JHU BioRad-Avidity) and a point-of-care assay (rapid LAg). The four MAAs included different combinations of these assays and HIV viral load. Seroconverters on antiretroviral treatment (ART) were identified using a qualitative multi-drug assay.
Results
The MAAs identified between 54 and 100 (25% to 46%) of the seroconverters as recently-infected. The false recent rate of the MAAs for infections >2 years duration ranged from 0.2%-1.3%. The MAAs classified different overlapping groups of individuals as recent vs. non-recent. Only 32 (15%) of the 220 seroconverters were classified as recent by all four MAAs. Viral suppression impacted the performance of the two LAg-based assays. LAg-Avidity assay values were also lower for seroconverters who were virally suppressed on ART compared to those with natural viral suppression.
Conclusions
The four MAAs evaluated varied in sensitivity and specificity for identifying persons infected <1 year as recently infected and classified different groups of seroconverters as recently infected. Sensitivity was low for all four MAAs. These performance issues should be considered if these methods are used for individual-level recency assessments.
Collapse
Affiliation(s)
- Wendy Grant-McAuley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ethan Klock
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Oliver Laeyendecker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Estelle Piwowar-Manning
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ethan Wilson
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ayana Moore
- FHI360, Durham, North Carolina, United States of America
| | - Helen Ayles
- Zambart, University of Zambia School of Medicine, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Barry Kosloff
- Zambart, University of Zambia School of Medicine, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Kwame Shanaube
- Zambart, University of Zambia School of Medicine, Lusaka, Zambia
| | - Peter Bock
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | - Nomtha Mandla
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | - Anneen van Deventer
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Deborah Donnell
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Richard Hayes
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| | | |
Collapse
|
12
|
Wu Y, Chu L, Yang H, Wang W, Zhang Q, Yang J, Qiao S, Li X, Shen Z, Zhou Y, Liu S, Deng H. Simultaneous Determination of 6 Antiretroviral Drugs in Human Hair Using an LC-ESI+-MS/MS Method: Application to Adherence Assessment. Ther Drug Monit 2021; 43:756-765. [PMID: 33587427 PMCID: PMC8355263 DOI: 10.1097/ftd.0000000000000878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The determination of antiretroviral drugs in hair is receiving considerable research interest to assess long-term adherence to antiretroviral therapy (ART). Currently in China, lamivudine, zidovudine, nevirapine, efavirenz, ritonavir, and lopinavir are combined as first-line and second-line free therapy regimens and are recommended for people living with HIV (PLWH). Simultaneous determination of the 6 antiretroviral drugs in human hair is important for accurately and widely assessing long-term adherence in Chinese PLWH receiving different ART regimens. METHODS Six drugs were extracted from 10-mg hair samples incubated in methanol for 16 hours at 37°C and then analyzed by liquid chromatography with tandem mass spectrometry using a mobile phase of 95% methanol, with an electrospray ionization source in multiple reaction monitoring and positive mode. RESULTS The LC-ESI+-MS/MS method exhibited a linear range (R2 > 0.99) within 6-5000, 10-5000, 6-50,000, 12-50,000, 8-5000, and 8-12,500 pg/mg for lamivudine, zidovudine, nevirapine, efavirenz, ritonavir, and lopinavir. For all 6 drugs, the limits of quantification ranged between 6 and 12 pg/mg. The intraday and interday coefficients of variation were within 15%, and the recoveries ranged from 91.1% to 113.7%. Furthermore, the other validation parameters (ie, selectivity, matrix effect, stability, and carryover) met the acceptance criteria stipulated by guidelines of the US Food and Drug Administration and European Medicines Agency. Significant intergroup differences were observed between high-adherence and low-adherence groups, with high intercorrelations in the hair content of the 6 drugs. CONCLUSIONS The developed method demonstrated good reliability, to comprehensively and accurately assess adherence in PLWH receiving different ART regimens.
Collapse
Affiliation(s)
- Yan Wu
- Department of Brain and Learning Science, School of Biological Science and Medical Engineering, Southeast University
- Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education
- Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing, China
| | - Liuxi Chu
- Department of Brain and Learning Science, School of Biological Science and Medical Engineering, Southeast University
- Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education
- Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing, China
| | - Haoran Yang
- Department of Brain and Learning Science, School of Biological Science and Medical Engineering, Southeast University
- Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education
- Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing, China
| | - Wei Wang
- Department of Brain and Learning Science, School of Biological Science and Medical Engineering, Southeast University
- Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education
- Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing, China
| | - Quan Zhang
- Department of Health Promotion, Education and Behavior, South Carolina SmartState Center for Healthcare Quality (CHQ), Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
- Institute of Applied Psychology and School of Public Administration, Hohai University
| | - Jin Yang
- Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education
- Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing, China
- Department of Preventive Medicine, School of Public Health, Southeast University, Nanjing; and
| | - Shan Qiao
- Department of Health Promotion, Education and Behavior, South Carolina SmartState Center for Healthcare Quality (CHQ), Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Xiaoming Li
- Department of Health Promotion, Education and Behavior, South Carolina SmartState Center for Healthcare Quality (CHQ), Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Zhiyong Shen
- Unit of AIDS Prevention and Control, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Yuejiao Zhou
- Unit of AIDS Prevention and Control, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Shuaifeng Liu
- Unit of AIDS Prevention and Control, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Huihua Deng
- Department of Brain and Learning Science, School of Biological Science and Medical Engineering, Southeast University
- Key Laboratory of Child Development and Learning Science (Southeast University), Ministry of Education
- Institute of Child Development and Education, Research Center for Learning Science, Southeast University, Nanjing, China
| |
Collapse
|
13
|
Determination of Antiviral Drugs and Their Metabolites Using Micro-Solid Phase Extraction and UHPLC-MS/MS in Reversed-Phase and Hydrophilic Interaction Chromatography Modes. Molecules 2021; 26:molecules26082123. [PMID: 33917128 PMCID: PMC8067820 DOI: 10.3390/molecules26082123] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 11/17/2022] Open
Abstract
Two new ultra-high performance liquid chromatography (UHPLC) methods for analyzing 21 selected antivirals and their metabolites were optimized, including sample preparation step, LC separation conditions, and tandem mass spectrometry detection. Micro-solid phase extraction in pipette tips was used to extract antivirals from the biological material of Hanks balanced salt medium of pH 7.4 and 6.5. These media were used in experiments to evaluate the membrane transport of antiviral drugs. Challenging diversity of physicochemical properties was overcome using combined sorbent composed of C18 and ion exchange moiety, which finally allowed to cover the whole range of tested antivirals. For separation, reversed-phase (RP) chromatography and hydrophilic interaction liquid chromatography (HILIC), were optimized using extensive screening of stationary and mobile phase combinations. Optimized RP-UHPLC separation was carried out using BEH Shield RP18 stationary phase and gradient elution with 25 mmol/L formic acid in acetonitrile and in water. HILIC separation was accomplished with a Cortecs HILIC column and gradient elution with 25 mmol/L ammonium formate pH 3 and acetonitrile. Tandem mass spectrometry (MS/MS) conditions were optimized in both chromatographic modes, but obtained results revealed only a little difference in parameters of capillary voltage and cone voltage. While RP-UHPLC-MS/MS exhibited superior separation selectivity, HILIC-UHPLC-MS/MS has shown substantially higher sensitivity of two orders of magnitude for many compounds. Method validation results indicated that HILIC mode was more suitable for multianalyte methods. Despite better separation selectivity achieved in RP-UHPLC-MS/MS, the matrix effects were noticed while using both chromatographic modes leading to signal enhancement in RP and signal suppression in HILIC.
Collapse
|
14
|
Myer L, Redd AD, Mukonda E, Lynch BA, Phillips TK, Eisenberg A, Hsiao NY, Capoferri A, Zerbe A, Clarke W, Lesosky M, Breaud A, McIntyre J, Bruno D, Martens C, Abrams EJ, Reynolds SJ. Antiretroviral Adherence, Elevated Viral Load, and Drug Resistance Mutations in Human Immunodeficiency Virus-infected Women Initiating Treatment in Pregnancy: A Nested Case-control Study. Clin Infect Dis 2021; 70:501-508. [PMID: 30877752 DOI: 10.1093/cid/ciz209] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/15/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Elevated viral load (VL) early after antiretroviral therapy (ART) initiation appears frequently in pregnant and postpartum women living with human immunodeficiency virus; however the relative contributions of pre-ART drug resistance mutations (DRMs) vs nonadherence in the etiology of elevated VL are unknown. METHODS Within a cohort of women initiating ART during pregnancy in Cape Town, South Africa, we compared women with elevated VL after initial suppression (cases, n = 80) incidence-density matched to women who maintained suppression over time (controls, n = 87). Groups were compared on pre-ART DRMs and detection of antiretrovirals in stored plasma. RESULTS The prevalence of pre-ART DRMs was 10% in cases and 5% in controls (adjusted odds ratio [aOR], 1.53 [95% confidence interval {CI}, .4-5.9]); all mutations were to nonnucleoside reverse transcriptase inhibitors. At the time of elevated VL, 19% of cases had antiretrovirals detected in plasma, compared with 87% of controls who were suppressed at a matched time point (aOR, 131.43 [95% CI, 32.8-527.4]). Based on these findings, we estimate that <10% of all elevated VL in the cohort may be attributable to pre-ART DRMs vs >90% attributable to ART nonadherence. CONCLUSIONS DRMs account for a small proportion of all elevated VL among women occurring in the 12 months after ART initiation during pregnancy in this setting, with nonadherence appearing to drive most episodes of elevated VL. Alongside the drive for access to more robust antiretroviral agents in resource-limited settings, there is an ongoing need for effective strategies to support ART adherence in this patient population.
Collapse
Affiliation(s)
- Landon Myer
- Division of Epidemiology and Biostatistics, University of Cape Town, South Africa.,Centre for Infectious Diseases Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, South Africa
| | - Andrew D Redd
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda.,Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Elton Mukonda
- Division of Epidemiology and Biostatistics, University of Cape Town, South Africa
| | - Briana A Lynch
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda
| | - Tamsin K Phillips
- Division of Epidemiology and Biostatistics, University of Cape Town, South Africa.,Centre for Infectious Diseases Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, South Africa
| | - Anna Eisenberg
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda
| | - Nei-Yuan Hsiao
- Division of Medical Virology, Department of Pathology, University of Cape Town, South Africa.,National Health Laboratory Services, Groote Schuur Hospital, Cape Town, South Africa
| | - Adam Capoferri
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Alison Zerbe
- ICAP at Columbia University Mailman School of Public Health, New York, New York
| | - William Clarke
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Maia Lesosky
- Division of Epidemiology and Biostatistics, University of Cape Town, South Africa
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - James McIntyre
- Division of Epidemiology and Biostatistics, University of Cape Town, South Africa.,Anova Health Institute, Johannesburg, South Africa
| | - Daniel Bruno
- Genomics Unit, Research Technologies Branch, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana
| | - Craig Martens
- Genomics Unit, Research Technologies Branch, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana
| | - Elaine J Abrams
- ICAP at Columbia University Mailman School of Public Health, New York, New York.,Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Steven J Reynolds
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda.,Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
15
|
Grant-McAuley W, Fogel JM, Galai N, Clarke W, Breaud A, Marzinke MA, Mbwambo J, Likindikoki S, Aboud S, Donastorg Y, Perez M, Barrington C, Davis W, Kerrigan D, Eshleman SH. Antiretroviral drug use and HIV drug resistance in female sex workers in Tanzania and the Dominican Republic. PLoS One 2020; 15:e0240890. [PMID: 33119663 PMCID: PMC7595323 DOI: 10.1371/journal.pone.0240890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/05/2020] [Indexed: 02/04/2023] Open
Abstract
Objective Female sex workers (FSW) have increased risk of HIV infection. Antiretroviral treatment (ART) can improve HIV outcomes and prevent HIV transmission. We analyzed antiretroviral (ARV) drug use and HIV drug resistance among HIV-positive FSW in the Dominican Republic and Tanzania. Methods Plasma samples collected at study entry with viral loads >1,000 copies/mL were tested for ARV drugs and HIV drug resistance. ARV drug testing was performed using a qualitative assay that detects 22 ARV drugs in five classes. HIV genotyping was performed using the ViroSeq HIV-1 Genotyping System. Phylogenetic analyses were performed to determine HIV subtype and assess transmission clusters. Results Among 410 FSW, 144 (35.1%) had viral loads >1,000 copies/mL (DR: n = 50; Tanzania: n = 94). ARV drugs were detected in 36 (25.0%) of 144 samples. HIV genotyping results were obtained for 138 (95.8%) cases. No transmission clusters were observed in either country. HIV drug resistance was detected in 54 (39.1%) of 138 samples (31/35 [88.6%] with drugs detected; 23/103 [22.3%] without drugs detected); 29/138 (21.0%) had multi-class resistance (MCR). None with MCR had integrase strand transfer inhibitor resistance. In eight cases, one or more ARV drug was detected without corresponding resistance mutations; those women were at risk of acquiring additional drug resistance. Using multivariate logistic regression, resistance was associated with ARV drug detection (p<0.001), self-reported ART (full adherence [p = 0.034]; partial adherence [p<0.001]), and duration of HIV infection (p = 0.013). Conclusions In this cohort, many women were on ART, but were not virally suppressed. High levels of HIV drug resistance, including MCR, were observed. Resistance was associated with detection of ARV drugs, self-report of ART with full or partial adherence, and duration of HIV infection. These findings highlight the need for better HIV care among FSW to improve their health, reduce HIV drug resistance, and decrease risk of transmission to others.
Collapse
Affiliation(s)
- Wendy Grant-McAuley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jessica M. Fogel
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Noya Galai
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, Maryland, United States of America
- Department of Statistics, University of Haifa, Mt Carmel, Israel
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Mark A. Marzinke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jessie Mbwambo
- Department of Psychiatry, Muhimibili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Samuel Likindikoki
- Department of Psychiatry, Muhimibili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Said Aboud
- Department of Microbiology and Immunology, Muhimibili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Yeycy Donastorg
- Unidad de Investigacion de Vacunas, Instituto Dermatologico y Cirugia de la Piel, Santo Domingo, Dominican Republic
| | - Martha Perez
- Unidad de Investigacion de Vacunas, Instituto Dermatologico y Cirugia de la Piel, Santo Domingo, Dominican Republic
| | - Clare Barrington
- Department of Health Behavior, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Wendy Davis
- Center on Health, Risk and Society, American University, Washington, District of Columbia, United States of America
| | - Deanna Kerrigan
- Center on Health, Risk and Society, American University, Washington, District of Columbia, United States of America
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
16
|
Niessen WMA. Tandem mass spectrometry of small-molecule antiviral drugs: 1. HIV-related antivirals. INTERNATIONAL JOURNAL OF MASS SPECTROMETRY 2020; 455:116370. [PMID: 33519296 PMCID: PMC7834215 DOI: 10.1016/j.ijms.2020.116370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 06/04/2023]
Abstract
Antiviral drugs are a class of compounds developed specifically for the treatment of viral infections. In the development and subsequent application of antiviral drugs, like for any other class of drugs, quantitative analysis in biological matrix is important, e.g., to establish bioavailability, to study pharmacokinetics, and later on possibly for therapeutic drug monitoring. Liquid chromatography-mass spectrometry (LC-MS) with tandem mass spectrometry (MS-MS) operated in selected-reaction monitoring (SRM) mode is the method of choice in quantitative bioanalysis. As information of the fragmentation of antiviral drugs in MS-MS is very much scattered in the scientific literature, it was decided to collect this information and to review it, not only to understand which product ions are actually used in SRM, but also to assist in other studies, e.g., in the identification of drug metabolites or (forced) degradation products. In this first study, attention is paid to antiviral agents used against HIV infection. The review provides fragmentation schemes of ca. 40 antiviral agents as well as several phosphorylated anabolites. The identity of the product ions used in SRM, i.e., elemental composition and exact-m/z, is tabulated, and more detailed fragmentation schemes are provided.
Collapse
Affiliation(s)
- W M A Niessen
- hyphen MassSpec, Margrietstraat 34, 2215 HJ, Voorhout, the Netherlands
| |
Collapse
|
17
|
Redd AD, Mukonda E, Hu NC, Philips TK, Zerbe A, Lesosky M, Hsiao NY, Clarke W, Reynolds SJ, Abrams EJ, Myer L. ART Adherence, Resistance, and Long-term HIV Viral Suppression in Postpartum Women. Open Forum Infect Dis 2020; 7:ofaa346. [PMID: 33072803 DOI: 10.1093/ofid/ofaa346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 08/10/2020] [Indexed: 02/05/2023] Open
Abstract
HIV+ South African women who achieved viral suppression during routine antenatal care, but later experienced a viremic episode (viral load >1000 copies/mL), were examined for presence of antiretrovirals and classified as "nonadherers" or "suboptimal adherers." Women were tested for drug resistance mutations (DRMs) at several time points and underwent viral load testing 36-60 months postpartum. Suboptimal adherers were more likely to have DRM detected during their viremic episode (P = .03) and at a subsequent viremic time point (P = .05). There was no difference in levels of viral suppression 36-60 months later in women with DRM detected vs women who had no evidence of DRM (P = .5).
Collapse
Affiliation(s)
- Andrew D Redd
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Baltimore, Maryland, USA.,Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Elton Mukonda
- Division of Epidemiology and Biostatistics, University of Cape Town, Cape Town, South Africa
| | - Nai-Chung Hu
- Division of Epidemiology and Biostatistics, University of Cape Town, Cape Town, South Africa
| | - Tamsin K Philips
- Division of Epidemiology and Biostatistics, University of Cape Town, Cape Town, South Africa
| | - Allison Zerbe
- ICAP at Columbia University Mailman School of Public Health, New York, New York, USA
| | - Maia Lesosky
- Division of Epidemiology and Biostatistics, University of Cape Town, Cape Town, South Africa
| | - Nei-Yuan Hsiao
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa.,National Health Laboratory Services, Groote Schuur Hospital, Cape Town, South Africa
| | - William Clarke
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Steven J Reynolds
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Baltimore, Maryland, USA.,Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Rakai Health Sciences Program, Kalisizo, Uganda
| | - Elaine J Abrams
- ICAP at Columbia University Mailman School of Public Health, New York, New York, USA.,Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Landon Myer
- Division of Epidemiology and Biostatistics, University of Cape Town, Cape Town, South Africa.,Centre for Infectious Diseases Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
18
|
Pollock S, Toegel F, Holtyn AF, Rodewald AM, Leoutsakos JM, Fingerhood M, Silverman K. Effects of incentives on viral suppression in people living with HIV who use cocaine or opiates. Drug Alcohol Depend 2020; 212:108000. [PMID: 32362436 PMCID: PMC7293918 DOI: 10.1016/j.drugalcdep.2020.108000] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/17/2020] [Accepted: 03/27/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Antiretroviral therapy (ART) adherence is essential to maintenance of undetectable viral loads among people living with HIV, which improves health and reduces HIV transmission. Despite these benefits, some people living with HIV do not maintain the level of adherence required to sustain an undetectable viral load. This problem is particularly common among people who use drugs. OBJECTIVE To determine effects of incentivizing viral suppression in people living with HIV who used cocaine or opiates. METHODS In this secondary analysis of data collected during a randomized controlled trial, participants (N=102) with detectable HIV viral loads (>200 copies/mL) were randomly assigned to a Usual Care or Incentive group. Usual Care participants did not earn incentives for viral suppression. Incentive participants earned incentives ($10/day maximum) for providing blood samples with reduced or undetectable (<200 copies/mL) viral loads. All participants completed assessments every three months. Results collected during the first year were compared based on group assignment and drug use. RESULTS Among participants who used cocaine or opiates, Incentive participants (n = 27) provided more (OR:4.0, CI:1.6-10.3, p = .004) blood samples with an undetectable viral load (69 %) than Usual Care participants (n = 25; 41 %). Among participants who did not use cocaine or opiates, Incentive participants (n = 25) provided more (OR:4.1, CI:1.5-10.7, p = .005) blood samples with an undetectable viral load (78 %) than Usual Care participants (n = 25; 36 %). Effects of incentives did not differ by drug use (OR:1.0, CI:0.3-4.0, p = .992). CONCLUSIONS Incentivizing viral suppression can promote undetectable viral loads in people living with HIV who use cocaine or opiates.
Collapse
Affiliation(s)
- Sarah Pollock
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue Suite 350 East, Baltimore, MD, 21224, USA.
| | - Forrest Toegel
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue Suite 350 East, Baltimore, MD, 21224, USA.
| | - August F Holtyn
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue Suite 350 East, Baltimore, MD, 21224, USA.
| | - Andrew M Rodewald
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue Suite 350 East, Baltimore, MD, 21224, USA.
| | - Jeannie-Marie Leoutsakos
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue Suite 350 East, Baltimore, MD, 21224, USA.
| | - Michael Fingerhood
- Department of Medicine, Comprehensive Care Practice, Johns Hopkins Bayview Medical Center, 5200 Eastern Ave # W2, Baltimore, MD 21224, USA.
| | - Kenneth Silverman
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue Suite 350 East, Baltimore, MD, 21224, USA.
| |
Collapse
|
19
|
Toegel F, Holtyn AF, Pollock S, Rodewald AM, Leoutsakos JM, Fingerhood M, Silverman K. Effects of incentivizing viral suppression in previously incarcerated adults living with HIV. HIV Res Clin Pract 2020; 21:1-10. [PMID: 32133931 PMCID: PMC7259460 DOI: 10.1080/25787489.2020.1735816] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/12/2020] [Indexed: 12/21/2022]
Abstract
Background: The amount of HIV in a person's blood can be suppressed to an undetectable level through antiretroviral therapy medications (ART). Adhering to an ART regimen can improve a person's health and reduce HIV transmission. Despite these benefits, many people with HIV do not maintain the level of adherence required to achieve an undetectable viral load. This problem is particularly common among people who have been incarcerated.Objective: To determine effects of incentivizing viral suppression in previously incarcerated adults with HIV.Methods: Adults with HIV (N = 102) and detectable viral load (>200 copies/mL) were randomly assigned to a Usual Care or Incentive group. Usual Care participants did not earn incentives for viral suppression. Incentive participants earned incentives ($10/day maximum) for providing blood samples with a reduced or undetectable (<200 copies/mL) viral load. Assessments were conducted every 3 months. Results collected during the first year were aggregated and compared based on group assignment and incarceration history.Results: Previously incarcerated participants in the Incentive group provided more (OR: 2.9; CI: 1.3-6.8; p <.05) blood samples with an undetectable viral load (69%) than those in the Usual Care group (41%). Never-incarcerated participants in the Incentive group provided more (OR: 6.8; CI: 2.2-21.0; p <.01) blood samples with an undetectable viral load (78%) than those in the Usual Care group (36%). Effects of incentives did not differ by incarceration history.Conclusions: Incentivizing viral suppression can increase viral suppression (undetectable viral load) in people who have been incarcerated.
Collapse
Affiliation(s)
- Forrest Toegel
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - August F. Holtyn
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Pollock
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew M. Rodewald
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeannie-Marie Leoutsakos
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Fingerhood
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kenneth Silverman
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
20
|
Simultaneous Analysis of Drugs in Forensic Cases by Liquid Chromatography–High-Resolution Orbitrap Mass Spectrometry. Chromatographia 2019. [DOI: 10.1007/s10337-019-03814-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
21
|
Palumbo PJ, Zhang Y, Fogel JM, Guo X, Clarke W, Breaud A, Richardson P, Piwowar-Manning E, Hart S, Hamilton EL, Hoa NTK, Liulchuk M, Anandari L, Ha TV, Dumchev K, Djoerban Z, Hoffman I, Hanscom B, Miller WC, Eshleman SH. HIV drug resistance in persons who inject drugs enrolled in an HIV prevention trial in Indonesia, Ukraine, and Vietnam: HPTN 074. PLoS One 2019; 14:e0223829. [PMID: 31600343 PMCID: PMC6786608 DOI: 10.1371/journal.pone.0223829] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 09/27/2019] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Persons who inject drugs (PWID) have high HIV incidence and prevalence, and may have limited access to antiretroviral therapy (ART) in some settings. We evaluated HIV drug resistance in PWID in a randomized clinical trial (HPTN 074). The study intervention included ART at any CD4 cell count with enhanced support for ART and substance use treatment. METHODS HPTN 074 enrolled HIV-infected PWID (index participants) with viral loads ≥1,000 copies/mL and their HIV-uninfected injection-network partners in Indonesia, Ukraine, and Vietnam; the study limited enrollment of people who reported being on ART. HIV drug resistance testing and antiretroviral (ARV) drug testing were performed using samples collected from index participants at study enrollment. RESULTS Fifty-four (12.0%) of 449 participants had HIV drug resistance; 29 (53.7%) of the 54 participants had multi-class resistance. Prevalence of resistance varied by study site and was associated with self-report of prior or current ART, detection of ARV drugs, and a history of incarceration. Resistance was detected in 10 (5.6%) of 177 newly diagnosed participants. Participants with resistance at enrollment were less likely to be virally suppressed after 52 weeks of follow-up, independent of study arm. CONCLUSIONS In HPTN 074, many of the enrolled index participants had HIV drug resistance and more than half of those had multi-class resistance. Some newly-diagnosed participants had resistance, suggesting that they may have been infected with drug-resistant HIV strains. Behavioral and geographic factors were associated with baseline resistance. Baseline resistance was associated with reduced viral suppression during study follow-up. These findings indicate the need for enhanced HIV care in this high-risk population to achieve sustained viral suppression on ART.
Collapse
Affiliation(s)
- Philip J. Palumbo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Yinfeng Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jessica M. Fogel
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Xu Guo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Paul Richardson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Estelle Piwowar-Manning
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Stephen Hart
- Frontier Science Foundation, Amherst, NY, United States of America
| | - Erica L. Hamilton
- Science Facilitation Department, Durham, NC, United States of America
| | - Ngo T. K. Hoa
- University of North Carolina Vietnam, Hanoi, Vietnam
| | - Mariya Liulchuk
- Gromashevsky Institute for Epidemiology and Infectious Diseases of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Latifah Anandari
- University of Indonesia/Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Tran Viet Ha
- Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | | | - Zubairi Djoerban
- Departments of Hematology, Medical Oncology, and Medicine, University of Indonesia/Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Irving Hoffman
- Department of Medicine, University of North Carolina Chapel Hill School of Medicine, Chapel Hill, NC, United States of America
| | - Brett Hanscom
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - William C. Miller
- Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, OH, United States of America
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
22
|
Selhorst P, Combrinck CE, Manning K, Botha FCJ, Labuschagne JPL, Anthony C, Matten DL, Breaud A, Clarke W, Quinn TC, Redd AD, Williamson C, Muller E. Longer-Term Outcomes of HIV-Positive-to-HIV-Positive Renal Transplantation. N Engl J Med 2019; 381:1387-1389. [PMID: 31577883 PMCID: PMC10014633 DOI: 10.1056/nejmc1903013] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Andrew D Redd
- National Institute of Allergy and Infectious Diseases, Bethesda, MD
| | | | - Elmi Muller
- University of Cape Town, Cape Town, South Africa
| |
Collapse
|
23
|
Hansoti B, Mwinnyaa G, Hahn E, Rao A, Black J, Chen V, Clark K, Clarke W, Eisenberg AL, Fernandez R, Iruedo J, Laeyendecker O, Maharaj R, Mda P, Miller J, Mvandaba N, Nyanisa Y, Reynolds SJ, Redd AD, Ryan S, Stead DF, Wallis LA, Quinn TC. Targeting the HIV Epidemic in South Africa: The Need for Testing and Linkage to Care in Emergency Departments. EClinicalMedicine 2019; 15:14-22. [PMID: 31709410 PMCID: PMC6833451 DOI: 10.1016/j.eclinm.2019.08.007] [Citation(s) in RCA: 17] [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: 05/13/2019] [Revised: 08/12/2019] [Accepted: 08/12/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The Eastern Cape province of South Africa has one of the highest burdens of HIV in the world. Emergency Departments (EDs) can serve as optimal clinical sites for the identification of new HIV infections and entry into care. We sought to determine the current burden of HIV disease among ED patients in the Eastern Cape. METHODS We conducted a prospective cross-sectional observational study in the EDs of three Hospitals in the Eastern Cape province of South Africa from June 2017 to July 2018. All adult, non-critical patients presenting to the ED were systematically approached and offered a Point-Of-Care (POC) HIV test in accordance with South African guidelines. All HIV-positive individuals had their blood tested for the presence of antiretroviral therapy (ART) and the presence of viral suppression (≤ 1000 copies/ml). HIV incidence was estimated using a multi-assay algorithm, validated for a subtype C epidemic. FINDINGS Of the 2901 patients for whom HIV status was determined (either known HIV-positive or underwent POC HIV testing), 811 (28.0%) were HIV positive, of which 234 (28.9%) were newly diagnosed. HIV prevalence was higher in Mthatha [34% (388/1134) at Mthatha Regional Hospital and 28% (142/512) at Nelson Mandela Academic Hospital], compared to Port Elizabeth [22% (281/1255) at Livingstone Hospital]. HIV incidence was estimated at 4.5/100 person-years (95% CI: 2.4, 6.50) for women and 1.5 (CI 0.5, 2.5) for men. Of all HIV positive individuals tested for ART (585), 54% (316/585) tested positive for the presence of ARTs, and for all HIV positive participants with viral load data (609), 49% (299/609) were found to be virally suppressed. INTERPRETATION Our study not only observed a high prevalence and incidence of HIV among ED patients but also highlights significant attrition along the HIV care cascade for HIV positive individuals. Furthermore, despite developing an optimal testing environment, we were only able to enrol a small sub-set of the ED population. Given the high HIV prevalence and high attrition in the ED population, HIV services in the ED should also develop strategies that can accommodate large testing volumes and ART initiation.
Collapse
Affiliation(s)
- Bhakti Hansoti
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
| | - George Mwinnyaa
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, 31 Center Dr # 7A03, Bethesda, MD 20892, USA
| | - Elizabeth Hahn
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Aditi Rao
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
| | - John Black
- Faculty of Health Sciences, Walter Sisulu University, Umtata Part 1, Mthatha, South Africa
- Department of Medicine, Livingstone Hospital, Stanford Road, Korsten, Port Elizabeth 6020, South Africa
| | - Victoria Chen
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Kathryn Clark
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
| | - William Clarke
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Anna L. Eisenberg
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, 31 Center Dr # 7A03, Bethesda, MD 20892, USA
| | | | - Joshua Iruedo
- Faculty of Health Sciences, Walter Sisulu University, Umtata Part 1, Mthatha, South Africa
| | - Oliver Laeyendecker
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, 31 Center Dr # 7A03, Bethesda, MD 20892, USA
| | - Roshen Maharaj
- Faculty of Health Sciences, Walter Sisulu University, Umtata Part 1, Mthatha, South Africa
- Department of Emergency Medicine, Livingstone Hospital, Stanford Road, Korsten, Port Elizabeth 6020, South Africa
| | - Pamela Mda
- Nelson Mandela Hospital Clinical Research Unit, Sisson St, Fort Gale, Mthatha 5100, South Africa
| | - Jernelle Miller
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Nomzamo Mvandaba
- Faculty of Health Sciences, Walter Sisulu University, Umtata Part 1, Mthatha, South Africa
| | - Yandisa Nyanisa
- Faculty of Health Sciences, Walter Sisulu University, Umtata Part 1, Mthatha, South Africa
| | - Steven J. Reynolds
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, 31 Center Dr # 7A03, Bethesda, MD 20892, USA
| | - Andrew D. Redd
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, 31 Center Dr # 7A03, Bethesda, MD 20892, USA
| | - Sofia Ryan
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
| | - David F. Stead
- Faculty of Health Sciences, Walter Sisulu University, Umtata Part 1, Mthatha, South Africa
- Department of Medicine, Frere Hospital, Amalinda Main Rd, Braelyn, East London 5201, South Africa
| | - Lee A. Wallis
- Division of Emergency Medicine, University of Cape Town, Main Rd, Observatory, Cape Town 7925, South Africa
| | - Thomas C. Quinn
- The Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, 31 Center Dr # 7A03, Bethesda, MD 20892, USA
| |
Collapse
|
24
|
Silverman K, Holtyn AF, Rodewald AM, Siliciano RF, Jarvis BP, Subramaniam S, Leoutsakos JM, Getty CA, Ruhs S, Marzinke MA, Fingerhood M. Incentives for Viral Suppression in People Living with HIV: A Randomized Clinical Trial. AIDS Behav 2019; 23:2337-2346. [PMID: 31297681 PMCID: PMC6768703 DOI: 10.1007/s10461-019-02592-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The HIV/AIDS epidemic can be eliminated if 73% of people living with HIV take antiretroviral medications and achieve undetectable viral loads. This study assessed the effects of financial incentives in suppressing viral load. People living with HIV with detectable viral loads (N = 102) were randomly assigned to Usual Care or Incentive groups. Incentive participants earned up to $10 per day for 2 years for providing blood samples that showed either reduced or undetectable viral loads. This report presents data on the 1st year after random assignment. Incentive participants provided more (adjusted OR = 15.6, CI 4.2-58.8, p < 0.001) blood samples at 3-month assessments with undetectable viral load (72.1%) than usual care control participants (39.0%). We collected most blood samples. The study showed that incentives can substantially increase undetectable viral loads in people living with HIV. Financial incentives for suppressed viral loads could contribute to the eradication of the HIV/AIDS epidemic.
Collapse
Affiliation(s)
- Kenneth Silverman
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, Suite 350 East, Baltimore, MD, 21224, USA.
| | - August F Holtyn
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, Suite 350 East, Baltimore, MD, 21224, USA
| | - Andrew M Rodewald
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, Suite 350 East, Baltimore, MD, 21224, USA
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brantley P Jarvis
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, Suite 350 East, Baltimore, MD, 21224, USA
- Knowesis, LLC, Fairfax, VA, USA
| | - Shrinidhi Subramaniam
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, Suite 350 East, Baltimore, MD, 21224, USA
- Department of Psychology and Child Development, California State University Stanislaus, Turlock, CA, USA
| | - Jeannie-Marie Leoutsakos
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, Suite 350 East, Baltimore, MD, 21224, USA
| | - Carol-Ann Getty
- Department of Psychiatry and Behavioral Sciences, Center for Learning and Health, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, Suite 350 East, Baltimore, MD, 21224, USA
- Addictions Department, Kings College, London, England, UK
| | | | - Mark A Marzinke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Fingerhood
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
25
|
Fogel JM, Zhang Y, Palumbo PJ, Guo X, Clarke W, Breaud A, Richardson P, Piwowar-Manning E, Hamilton EL, Ha TV, Dumchev K, Djoerban Z, Hoffman I, Hanscom B, Miller WC, Eshleman SH. Use of Antiretroviral Drug Testing to Assess the Accuracy of Self-reported Data from HIV-Infected People Who Inject Drugs. AIDS Behav 2019; 23:2101-2108. [PMID: 30600453 DOI: 10.1007/s10461-018-2379-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We used antiretroviral (ARV) drug testing to evaluate the accuracy of self-reported data for HIV status and antiretroviral treatment (ART) among people who inject drugs enrolled in an HIV prevention trial. ARV drugs were detected in enrollment samples from 72/482 = 14.9% HIV-infected participants (39/52 = 75.0% who reported being on ART; 33/430 = 7.7% who reported not being on ART). Overall, 213/482 = 44.2% participants indicated that they were not aware of their HIV-positive status prior to study entry; of those, 30 had ARV drugs detected at enrollment, including 15 who also had ARV drugs detected at the screening visit. These participants were likely aware of their HIV-positive status at study entry but did not report this to study staff. This study shows that self-reported data on HIV testing history and ART may not be accurate and that ARV drug testing can help identify persons who are aware of their HIV-positive status and are on ART.
Collapse
|
26
|
Moorhouse L, Schaefer R, Thomas R, Nyamukapa C, Skovdal M, Hallett TB, Gregson S. Application of the HIV prevention cascade to identify, develop and evaluate interventions to improve use of prevention methods: examples from a study in east Zimbabwe. J Int AIDS Soc 2019; 22 Suppl 4:e25309. [PMID: 31328375 PMCID: PMC6643077 DOI: 10.1002/jia2.25309] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 05/08/2019] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION The HIV prevention cascade could be used in developing interventions to strengthen implementation of efficacious HIV prevention methods, but its practical utility needs to be demonstrated. We propose a standardized approach to using the cascade to guide identification and evaluation of interventions and demonstrate its feasibility for this purpose through a project to develop interventions to improve HIV prevention methods use by adolescent girls and young women (AGYW) and potential male partners in east Zimbabwe. DISCUSSION We propose a six-step approach to using a published generic HIV prevention cascade formulation to develop interventions to increase motivation to use, access to and effective use of an HIV prevention method. These steps are as follows: (1) measure the HIV prevention cascade for the chosen population and method; (2) identify gaps in the cascade; (3) identify explanatory factors (barriers) contributing to observed gaps; (4) review literature to identify relevant theoretical frameworks and interventions; (5) tailor interventions to the local context; and (6) implement and evaluate the interventions using the cascade steps and explanatory factors as outcome indicators in the evaluation design. In the Zimbabwe example, steps 1-5 aided development of four interventions to overcome barriers to effective use of pre-exposure prophylaxis (PrEP) in AGYW (15-24 years) and voluntary medical male circumcision in male partners (15-29). For young men, prevention cascade analyses identified gaps in motivation and access as barriers to voluntary medical male circumcision uptake, so an intervention was designed including financial incentives and an education session. For AGYW, gaps in motivation (particularly lack of risk perception) and access were identified as barriers to PrEP uptake: an interactive counselling game was developed addressing these barriers. A text messaging intervention was developed to improve PrEP adherence among AGYW, addressing reasons underlying lack of effective PrEP use through improving the capacity ("skills") to take PrEP effectively. A community-led intervention (community conversations) was developed addressing community-level factors underlying gaps in motivation, access and effective use. These interventions are being evaluated currently using outcomes from the HIV prevention cascade (step 6). CONCLUSIONS The prevention cascade can guide development and evaluation of interventions to strengthen implementation of HIV prevention methods by following the proposed process.
Collapse
Affiliation(s)
- Louisa Moorhouse
- Department of Infectious Disease EpidemiologyMRC Centre for Global Infectious Disease AnalysisImperial College LondonLondonUK
| | - Robin Schaefer
- Department of Infectious Disease EpidemiologyMRC Centre for Global Infectious Disease AnalysisImperial College LondonLondonUK
| | - Ranjeeta Thomas
- Department of Health PolicyLondon School of Economics and Political ScienceLondonUK
| | - Constance Nyamukapa
- Department of Infectious Disease EpidemiologyMRC Centre for Global Infectious Disease AnalysisImperial College LondonLondonUK
- Biomedical Research and Training InstituteHarareZimbabwe
| | - Morten Skovdal
- Department of Public HealthUniversity of CopenhagenCopenhagenDenmark
| | - Timothy B Hallett
- Department of Infectious Disease EpidemiologyMRC Centre for Global Infectious Disease AnalysisImperial College LondonLondonUK
| | - Simon Gregson
- Department of Infectious Disease EpidemiologyMRC Centre for Global Infectious Disease AnalysisImperial College LondonLondonUK
- Biomedical Research and Training InstituteHarareZimbabwe
| |
Collapse
|
27
|
Charbe NB, Zacconi FC, Amnerkar N, Ramesh B, Tambuwala MM, Clementi E. Bio-analytical Assay Methods used in Therapeutic Drug Monitoring of Antiretroviral Drugs-A Review. CURRENT DRUG THERAPY 2019; 14:16-57. [DOI: 10.2174/1574885514666181217125550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 09/17/2018] [Accepted: 11/09/2018] [Indexed: 12/12/2022]
Abstract
Background: Several clinical trials, as well as observational statistics, have exhibited that the advantages of antiretroviral [ARV] treatment for humans with Human Immunodeficiency Virus / Acquired Immune Deficiency Syndrome HIV/AIDS exceed their risks. Therapeutic drug monitoring [TDM] plays a key role in optimization of ARV therapy. Determination of ARV’s in plasma, blood cells, and other biological matrices frequently requires separation techniques capable of high effectiveness, specific selectivity and high sensitivity. High-performance liquid chromatography [HPLC] coupled with ultraviolet [UV], Photodiode array detectors [PDA], Mass spectrophotometer [MS] detectors etc. are the important quantitative techniques used for the estimation of pharmaceuticals in biological samples. </P><P> Objective: This review article is aimed to give an extensive outline of different bio-analytical techniques which have been reported for direct quantitation of ARV’s. This article aimed to establish an efficient role played by the TDM in the optimum therapeutic outcome of the ARV treatment. It also focused on establishing the prominent role played by the separation techniques like HPLC and UPLC along with the detectors like UV and Mass in TDM. </P><P> Methods: TDM is based on the principle that for certain drugs, a close relationship exists between the plasma level of the drug and its clinical effect. TDM is of no value if the relationship does not exist. The analytical methodology employed in TDM should: 1) distinguish similar compounds; 2) be sensitive and precise and 3) is easy to use. </P><P> Results: This review highlights the advancement of the chromatographic techniques beginning from the HPLC-UV to the more advanced technique like UPLC-MS/MS. TDM is essential to ensure adherence, observe viral resistance and to personalize ARV dose regimens. It is observed that the analytical methods like immunoassays and liquid chromatography with detectors like UV, PDA, Florescent, MS, MS/MS and Ultra performance liquid chromatography (UPLC)-MS/MS have immensely contributed to the clinical outcome of the ARV therapy. Assay methods are not only helping physicians in limiting the side effects and drug interactions but also assisting in monitoring patient’s compliance. </P><P> Conclusion: The present review revealed that HPLC has been the most widely used system irrespective of the availability of more sensitive chromatographic technique like UPLC.
Collapse
Affiliation(s)
- Nitin B. Charbe
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna McKenna 4860, Macul, Santiago 7820436, Chile
| | - Flavia C. Zacconi
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna McKenna 4860, Macul, Santiago 7820436, Chile
| | - Nikhil Amnerkar
- Adv V. R. Manohar Institute of Diploma in Pharmacy, Wanadongri, Hingna Road, Nagpur, Maharashtra 441110, India
| | - B. Ramesh
- Sri Adichunchunagiri University, Sri Adichunchunagiri College of Pharmacy, BG Nagar, Karnataka 571418, India
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, University of Ulster, Coleraine, County Londonderry, Northern Ireland BT52 1SA, United Kingdom
| | - Emilio Clementi
- Clinical Pharmacology Unit, CNR Institute of Neuroscience, Department of Biomedical and Clinical Sciences, Luigi Sacco University Hospital, Universita di Milano, Milan, Italy
| |
Collapse
|
28
|
Hansoti B, Stead D, Eisenberg A, Mvandaba N, Mwinnyaa G, Patel EU, Parrish A, Reynolds SJ, Redd AD, Fernandez R, Rothman RE, Laeyendecker O, Quinn TC. A Window Into the HIV Epidemic from a South African Emergency Department. AIDS Res Hum Retroviruses 2019; 35:139-144. [PMID: 30215268 DOI: 10.1089/aid.2018.0127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aim of the study was to describe the HIV care continuum in emergency department (ED) patients in the Eastern Cape region of South Africa. This is a cross-sectional, identity-unlinked serosurvey, whereby discarded/excess samples from all patients who had blood drawn during the study period for routine care and sufficient serum remaining were tested for HIV, hepatitis B virus, and hepatitis C virus infection; HIV viral load (VL); and presence of antiretroviral (ARV) drugs. We also estimated cross-sectional incidence using the Limiting-Antigen Avidity assay and HIV VL. The study was conducted between September and November 2016 at the Frere Hospital Emergency Department in East London, South Africa. The overall HIV prevalence in our study population was 26.9% [95% confidence interval (CI): 25.0-28.8; n = 2,100]. The highest prevalence was observed among females in the 30-39 years age group [60.3% (95% CI: 53.2-67.1)]. HIV prevalence was significantly higher among females compared with males in both the 20-29 years age group and 30-39 years age group (p < .05), but nearly identical to older age groups. ARV drugs were detected in 53.5% (95% CI: 48.1-58.9) of HIV-infected subjects. The frequency of HIV viral suppression (< 1,000 copies/mL) was 48.5% (95% CI: 44.3-52.7), and was not statistically different between males and females (age-adjusted prevalence ratio = 1.15, 95% CI: 0.95-1.39). The HIV incidence rate was estimated to be 2.6% (95% CI: 1.2-3.9). The Frere Hospital ED has an extremely high burden of HIV infection. The detection of ARV drugs and prevalence of viral suppression fall short of the World Health Organization 90-90-90 goals in this population. Furthermore, there were a large number of patients with recent infection in the ED. The ED is a critical venue for testing and linkage to care of high-yield population who are likely missed by current testing and linkage-to-care programs.
Collapse
Affiliation(s)
- Bhakti Hansoti
- Department of Emergency Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - David Stead
- Department of Medicine, Frere and Cecilia Makiwane Hospitals, East London, South Africa
- Department of Medicine, Faculty of Health Sciences, Walter Sisulu University, East London, South Africa
| | - Anna Eisenberg
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Baltimore, Maryland
| | - Nomzamo Mvandaba
- Department of Medicine, Faculty of Health Sciences, Walter Sisulu University, East London, South Africa
| | - George Mwinnyaa
- Department of Infectious Diseases, The Johns Hopkins University, Baltimore, Maryland
| | - Eshan U. Patel
- Department of Infectious Diseases, The Johns Hopkins University, Baltimore, Maryland
| | - Andy Parrish
- Department of Medicine, Frere and Cecilia Makiwane Hospitals, East London, South Africa
- Department of Medicine, Faculty of Health Sciences, Walter Sisulu University, East London, South Africa
| | - Steven J. Reynolds
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Baltimore, Maryland
- Department of Infectious Diseases, The Johns Hopkins University, Baltimore, Maryland
| | - Andrew D. Redd
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Baltimore, Maryland
- Department of Infectious Diseases, The Johns Hopkins University, Baltimore, Maryland
| | - Reinaldo Fernandez
- Department of Infectious Diseases, The Johns Hopkins University, Baltimore, Maryland
| | - Richard E. Rothman
- Department of Emergency Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Oliver Laeyendecker
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Baltimore, Maryland
- Department of Infectious Diseases, The Johns Hopkins University, Baltimore, Maryland
| | - Thomas C. Quinn
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Baltimore, Maryland
- Department of Infectious Diseases, The Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
29
|
Fogel JM, Sandfort T, Zhang Y, Guo X, Clarke W, Breaud A, Cummings V, Hamilton EL, Ogendo A, Kayange N, Panchia R, Dominguez K, Chen YQ, Eshleman SH. Accuracy of Self-Reported HIV Status Among African Men and Transgender Women Who Have Sex with Men Who were Screened for Participation in a Research Study: HPTN 075. AIDS Behav 2019; 23:289-294. [PMID: 30051192 DOI: 10.1007/s10461-018-2231-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Some HIV-infected individuals in research studies may choose not to disclose knowledge of their HIV status to study staff. We evaluated the accuracy of self-reported HIV status among African men and transgender women who have sex with men and who were screened for a research study. Sixty-seven of 183 HIV-infected participants reported a prior HIV diagnosis. Samples from the remaining 116 participants were tested for antiretroviral (ARV) drugs. Thirty-six of the 116 participants had ARV drugs detected, indicating that they were on antiretroviral treatment; these participants were classified as previously diagnosed based on ARV drug testing. Among participants classified as previously diagnosed, disclosure of a prior HIV diagnosis varied among study sites (p = 0.006) and was more common among those who reported having sex with men only (p = 0.002). ARV drug testing in addition to self-report improves the accuracy for identifying individuals with a prior HIV diagnosis.
Collapse
Affiliation(s)
- Jessica M Fogel
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Theodorus Sandfort
- HIV Center for Clinical and Behavioral Studies, Columbia University, New York, NY, USA
| | - Yinfeng Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xu Guo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vanessa Cummings
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Arthur Ogendo
- Kenya Medical Research Institute (KEMRI) CDC, Kisumu, Kenya
| | - Noel Kayange
- College of Medicine - Johns Hopkins Project, Blantyre, Malawi
| | - Ravindre Panchia
- Perinatal HIV Research Unit, University of the Witwatersrand, Soweto HPTN CRS, Soweto, South Africa
| | - Karen Dominguez
- Desmond Tutu HIV Centre, UCT Medical School, Cape Town, South Africa
| | - Ying Q Chen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Susan H Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pathology, The Johns Hopkins Medical Institutions, Ross Building, Room 646, 720 Rutland Avenue, Baltimore, MD, 21205, USA.
| |
Collapse
|
30
|
Sivay MV, Fogel JM, Wang J, Zhang Y, Piwowar-Manning E, Clarke W, Breaud A, Blankson J, Hamilton EL, Kahn K, Selin A, Gomez-Olive FX, MacPhail C, Hughes JP, Pettifor A, Eshleman SH. Natural control of HIV infection in young women in South Africa: HPTN 068. HIV CLINICAL TRIALS 2018; 19:202-208. [PMID: 30522410 DOI: 10.1080/15284336.2018.1531534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Some individuals control HIV replication without antiretroviral (ARV) therapy. OBJECTIVE To analyze viral suppression in young women in rural South Africa enrolled in a trial evaluating a behavioral intervention for HIV prevention. METHODS Plasma samples were obtained from women ages 13-24 (81 infected at enrollment, 164 seroconverters). ARV testing was performed using an assay that detects 20 ARV drugs. Women were classified as viremic controllers if they were virally suppressed for ≥12 months with no ARV drug use. RESULTS Samples from 216/245 (88.2%) women had no ARV drugs detected at their first HIV-positive visit. Thirty-four (15.7%) of the 216 women had a viral load <2,000 copies/mL. Fifteen of the 34 women were followed for ≥12 months; 12 were virally suppressed with no ARV drugs detected during follow-up. These women were classified as viremic controllers (overall: 12/216 = 5.6%). The median CD4 cell count at the first HIV-positive visit was higher among the 12 controllers than among the 204 women who were not using ARV drugs (759 vs. 549 cells/mm3, p = 0.02). Some women had a viral load <40 copies/mL at a single study visit, but none were classified as elite controllers (viral load <40 copies/mL for ≥12 months with no ARV drug use). CONCLUSIONS In this cohort, 5.6% of women who were not using ARV drugs had sustained viral suppression. This represents a minimum estimate of the frequency of viremic controllers in this cohort, since some women were not followed long enough to meet the criteria for classification.
Collapse
Affiliation(s)
- Mariya V Sivay
- a Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - Jessica M Fogel
- a Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - Jing Wang
- b Statistical Center for HIV/ AIDS Research & Prevention (SCHARP) , Seattle , Washington , USA
| | - Yinfeng Zhang
- a Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - Estelle Piwowar-Manning
- a Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - William Clarke
- a Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - Autumn Breaud
- a Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - Joel Blankson
- c Department of Medicine , Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - Erica L Hamilton
- d Science Facilitation Department , FHI 360 , Durham , North Carolina , USA
| | - Kathleen Kahn
- e MRC/Wits Rural Public Health and Health, Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Amanda Selin
- f University of North Carolina at Chapel Hill, Carolina Population Center , Chapel Hill , North Carolina , USA
| | - F Xavier Gomez-Olive
- e MRC/Wits Rural Public Health and Health, Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Catherine MacPhail
- e MRC/Wits Rural Public Health and Health, Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa.,g School of Health and Society , University of Wollongong , Wollongong , Australia
| | - James P Hughes
- h Department of Biostatistics , University of Washington , Seattle , Washington , USA
| | - Audrey Pettifor
- e MRC/Wits Rural Public Health and Health, Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa.,i Department of Epidemiology , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina , USA
| | - Susan H Eshleman
- a Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| |
Collapse
|
31
|
Zhang Y, Sivay MV, Hudelson SE, Clarke W, Breaud A, Wang J, Piwowar-Manning E, Agyei Y, Fogel JM, Hamilton EL, Selin A, MacPhail C, Kahn K, Gomez-Olive FX, Hughes JP, Pettifor A, Eshleman SH. Antiretroviral Drug Use and HIV Drug Resistance Among Young Women in Rural South Africa: HPTN 068. J Acquir Immune Defic Syndr 2018; 79:315-322. [PMID: 29985265 PMCID: PMC6185777 DOI: 10.1097/qai.0000000000001793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Antiretroviral (ARV) drugs are used for HIV treatment and prevention. We analyzed ARV drug use and HIV drug resistance in a cohort of young women in rural South Africa enrolled in the HIV Prevention Trials Network (HPTN) 068 study, which evaluated the use of a cash transfer conditional on school attendance to reduce HIV incidence. METHODS ARV drug testing was performed using plasma samples from 2526 young women. This included 2526 enrollment samples (80 HIV-infected and 2446 HIV-uninfected) and 162 seroconversion samples (first HIV-positive study visit). Testing was performed using a qualitative assay that detects 20 ARV drugs from 5 drug classes. HIV drug resistance testing was performed with the ViroSeq HIV-1 Genotyping System for samples that had HIV viral loads ≥400 copies per milliliter. RESULTS At enrollment, ARV drugs were detected in 10 (12.5%) of 80 HIV-infected young women. None of 2446 HIV-uninfected young women had ARV drugs detected at enrollment. ARV drugs were also detected in 16 (9.9%) of 162 seroconverters. At enrollment, 9 (13.4%) of 67 young women with genotyping results had HIV drug resistance; resistance was also detected in 9 (6.9%) of 131 seroconverters with genotyping results. CONCLUSIONS Most of the HIV-infected young women in this cohort from rural South Africa were not taking ARV drugs, suggesting they were unaware of their HIV status or were not in care. HIV drug resistance was detected in young women with both prevalent and new HIV infection.
Collapse
Affiliation(s)
- Yinfeng Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mariya V. Sivay
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah E. Hudelson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jing Wang
- Statistical Center for HIV/AIDS Research & Prevention (SCHARP), Seattle, WA, USA
| | | | - Yaw Agyei
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jessica M. Fogel
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Amanda Selin
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Catherine MacPhail
- School of Health and Society, University of Wollongong, Australia; MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kathleen Kahn
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - F. Xavier Gomez-Olive
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - James P. Hughes
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Audrey Pettifor
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC; MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
32
|
Moyo S, Gaseitsiwe S, Mohammed T, Pretorius Holme M, Wang R, Kotokwe KP, Boleo C, Mupfumi L, Yankinda EK, Chakalisa U, van Widenfelt E, Gaolathe T, Mmalane MO, Dryden-Peterson S, Mine M, Lebelonyane R, Bennett K, Leidner J, Wirth KE, Tchetgen Tchetgen E, Powis K, Moore J, Clarke WA, Lockman S, Makhema JM, Essex M, Novitsky V. Cross-sectional estimates revealed high HIV incidence in Botswana rural communities in the era of successful ART scale-up in 2013-2015. PLoS One 2018; 13:e0204840. [PMID: 30356287 PMCID: PMC6200198 DOI: 10.1371/journal.pone.0204840] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 09/12/2018] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Botswana is close to reaching the UNAIDS "90-90-90" HIV testing, antiretroviral treatment (ART), and viral suppression goals. We sought to determine HIV incidence in this setting with both high HIV prevalence and high ART coverage. METHODS We used a cross-sectional approach to assessing HIV incidence. A random, population-based sample of adults age 16-64 years was enrolled in 30 rural and peri-urban communities as part of the Botswana Combination Prevention Project (BCPP), from October 2013 -November 2015. Data and samples from the baseline household survey were used to estimate cross-sectional HIV incidence, following an algorithm that combined Limiting-Antigen Avidity Assay (LAg-Avidity EIA), ART status (documented or by testing ARV drugs in plasma) and HIV-1 RNA load. The LAg-Avidity EIA cut-off normalized optical density (ODn) was set at 1.5. The HIV-1 RNA cut-off was set at 400 copies/mL. For estimation purposes, the Mean Duration of Recent Infection was 130 days and the False Recent Rate (FRR) was evaluated at values of either 0 or 0.39%. RESULTS Among 12,610 individuals participating in the baseline household survey, HIV status was available for 12,570 participants and 3,596 of them were HIV positive. LAg-Avidity EIA data was generated for 3,581 (99.6%) of HIV-positive participants. Of 326 participants with ODn ≤1.5, 278 individuals were receiving ART verified through documentation and were considered to represent longstanding HIV infections. Among the remaining 48 participants who reported no use of ART, 14 had an HIV-1 RNA load ≤400 copies/mL (including 3 participants with ARVs in plasma) and were excluded, as potential elite/viremic controllers or undisclosed ART. Thus, 34 LAg-Avidity-EIA-recent, ARV-naïve individuals with detectable HIV-1 RNA (>400 copies/mL) were classified as individuals with recent HIV infections. The annualized HIV incidence among 16-64 year old adults was estimated at 1.06% (95% CI 0.68-1.45%) with zero FRR, and at 0.64% (95% CI 0.24-1.04%) using a previously defined FRR of 0.39%. Within a subset of younger individuals 16-49 years old, the annualized HIV incidence was estimated at 1.29% (95% CI 0.82-1.77%) with zero FRR, and at 0.90% (95% CI 0.42-1.38%) with FRR set to 0.39%. CONCLUSIONS Using a cross-sectional estimate of HIV incidence from 2013-2015, we found that at the time of near achievement of the UNAIDS 90-90-90 targets, ~1% of adults (age 16-64 years) in Botswana's rural and peri-urban communities became HIV infected annually.
Collapse
Affiliation(s)
- Sikhulile Moyo
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Simani Gaseitsiwe
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | | | - Molly Pretorius Holme
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Rui Wang
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Corretah Boleo
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Lucy Mupfumi
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | | | - Unoda Chakalisa
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | | | | | | | - Scott Dryden-Peterson
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Madisa Mine
- Botswana Ministry of Health and Wellness, Gaborone, Botswana
| | | | - Kara Bennett
- Bennett Statistical Consulting, Inc., Ballston Lake, New York, United States of America
| | - Jean Leidner
- Goodtables Data Consulting, Norman, OK, United States of America
| | - Kathleen E. Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Eric Tchetgen Tchetgen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Kathleen Powis
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Departments of Internal Medicine and Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Janet Moore
- U.S. Centers for Disease Control, Atlanta, Georgia, United States of America
| | | | - Shahin Lockman
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Joseph M. Makhema
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Max Essex
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Vlad Novitsky
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| |
Collapse
|
33
|
Abstract
: Among 3596 HIV-positive participants enrolled in the Botswana Combination Prevention Project who self-reported no prior antiretroviral (ARV) therapy use and were tested for viral load (n = 951; 27% of all participants), 136 (14%) had HIV-1 RNA less than 400 copies/ml. ARV drugs were detected in 52 (39%) of 134 participants tested. Adjusting for undisclosed ARV use increased the overall estimate of virally suppressed individuals on ARV therapy by 1.4% from 70.2 to 71.6%.
Collapse
|
34
|
Archibald TL, Murrell DE, Brown SD. Chromatographic methods in HIV medicine: Application to therapeutic drug monitoring. Biomed Chromatogr 2018; 32. [PMID: 29240228 DOI: 10.1002/bmc.4170] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/01/2017] [Accepted: 12/05/2017] [Indexed: 01/11/2023]
Abstract
HIV antiretroviral therapy spans several different drug classes, meant to combat various aspects of viral infection and replication. Many authors have argued the benefits of therapeutic drug monitoring (TDM) for the HIV patient including compliance assurance and assessment of appropriate drug concentrations; however, the array of drug chemistries and combinations makes TDM an arduous task. HPLC-UV and LC-MS/MS are both frequent instruments for the quantification of HIV drugs in biological matrices with investigators striving to balance sensitivity and affordability. Plasma, the dominant matrix for these analyses, is prepared using protein precipitation, liquid-liquid extraction or solid-phase extraction depending on the specific complement of analytes. Despite the range of polarities found in drug classes relevant to HIV therapeutics, most chromatographic separations utilize a hydrophobic column (C18 ). Additionally, as the clinically relevant samples for these assays are infected with HIV, along with possible co-infections, another important aspect of sample preparation concerns viral inactivation. Although not routine in clinical practice, many published analytical methods from the previous two decades have demonstrated the ability to conduct TDM in HIV patients receiving various medicinal combinations. This review summarizes the analytical methods relevant to TDM of HIV drugs, while highlighting respective challenges.
Collapse
Affiliation(s)
- Timothy L Archibald
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA
| | - Derek E Murrell
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA
| | - Stacy D Brown
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA
| |
Collapse
|
35
|
The validity of self-reported antiretroviral use in persons living with HIV: a population-based study. AIDS 2018; 32:363-369. [PMID: 29194115 DOI: 10.1097/qad.0000000000001706] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To assess the validity of self-reported antiretroviral therapy use (ART) using population-based cohort data. METHODS Self-reported ART use and nonuse was compared with a validated laboratory assay in 557 HIV-positive participants in the Rakai Community Cohort Study surveyed between September and December 2011 in Rakai, Uganda. The study population included participants from seven communities, including one fishing community with high HIV prevalence (∼41%). ART use was assayed using liquid chromatography-tandem mass spectrometry, which detects 20 antiretroviral drugs. HIV viral load measurements were also obtained. Individuals with at least two antiretroviral drugs detected were considered to be using ART. RESULTS One hundred and fifty-three (27%) participants self-reported ART use of whom 148 (97%) had at least two antiretroviral drugs detected. There were at least two antiretroviral drugs detected in 11% (n = 44/404) of individuals with no self-reported ART use. Overall, the specificity of self-reported ART use was 99% (95% CI 97-100%) and the sensitivity was 77% (70-83%). Positive and negative predictive values were 97% (95% CI 93-99%) and 89% (95% CI 86-92%), respectively. Nondisclosure of ART use was significantly more common in younger persons (<30 years) and among those in trading occupations. CONCLUSION Self-reported ART use has high specificity and moderate sensitivity providing reasonable, but conservative estimates of population-based ART use. There is more under-reporting of ART use among younger persons and traders suggesting a need for more research on barriers to self-reporting of ART use in these sub-groups.
Collapse
|
36
|
Nováková L, Pavlík J, Chrenková L, Martinec O, Červený L. Current antiviral drugs and their analysis in biological materials - Part II: Antivirals against hepatitis and HIV viruses. J Pharm Biomed Anal 2017; 147:378-399. [PMID: 29031512 DOI: 10.1016/j.jpba.2017.07.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/01/2017] [Indexed: 12/18/2022]
Abstract
This review is a Part II of the series aiming to provide comprehensive overview of currently used antiviral drugs and to show modern approaches to their analysis. While in the Part I antivirals against herpes viruses and antivirals against respiratory viruses were addressed, this part concerns antivirals against hepatitis viruses (B and C) and human immunodeficiency virus (HIV). Many novel antivirals against hepatitis C virus (HCV) and HIV have been introduced into the clinical practice over the last decade. The recent broadening portfolio of these groups of antivirals is reflected in increasing number of developed analytical methods required to meet the needs of clinical terrain. Part II summarizes the mechanisms of action of antivirals against hepatitis B virus (HBV), HCV, and HIV, their use in clinical practice, and analytical methods for individual classes. It also provides expert opinion on state of art in the field of bioanalysis of these drugs. Analytical methods reflect novelty of these chemical structures and use by far the most current approaches, such as simple and high-throughput sample preparation and fast separation, often by means of UHPLC-MS/MS. Proper method validation based on requirements of bioanalytical guidelines is an inherent part of the developed methods.
Collapse
Affiliation(s)
- Lucie Nováková
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic.
| | - Jakub Pavlík
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Lucia Chrenková
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Ondřej Martinec
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Lukáš Červený
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| |
Collapse
|
37
|
Evaluation of a Multidrug Assay for Monitoring Adherence to a Regimen for HIV Preexposure Prophylaxis in a Clinical Study, HIV Prevention Trials Network 073. Antimicrob Agents Chemother 2017; 61:AAC.02743-16. [PMID: 28438932 DOI: 10.1128/aac.02743-16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/17/2017] [Indexed: 02/06/2023] Open
Abstract
Daily oral tenofovir disoproxil fumarate (TDF)-emtricitabine (FTC) is a safe and effective intervention for HIV preexposure prophylaxis (PrEP). We evaluated the performance of a qualitative assay that detects 20 antiretroviral (ARV) drugs (multidrug assay) in assessing recent PrEP exposure (detection limit, 2 to 20 ng/ml). Samples were obtained from 216 Black men who have sex with men (208 HIV-uninfected men and 8 seroconverters) who were enrolled in a study in the United States evaluating the acceptability of TDF-FTC PrEP (165 of the uninfected men and 5 of the seroconverters accepted PrEP). Samples from 163 of the 165 HIV-uninfected men who accepted PrEP and samples from all 8 seroconverters were also tested for tenofovir (TFV) and FTC using a quantitative assay (detection limit for both drugs, 0.31 ng/ml). HIV drug resistance was assessed in seroconverter samples. The multidrug assay detected TFV and/or FTC in 3 (1.4%) of the 208 uninfected men at enrollment, 84 (40.4%) of the 208 uninfected men at the last study visit, and 1 (12.5%) of the 8 seroconverters. No other ARV drugs were detected. The quantitative assay confirmed all positive results from the multidrug assay and detected TFV and/or FTC in 9 additional samples (TFV range, 0.65 to 16.5 ng/ml; FTC range, 0.33 to 14.6 ng/ml). Resistance mutations were detected in 4 of the 8 seroconverter samples. The multidrug assay had 100% sensitivity and specificity for detecting TFV and FTC at drug concentrations consistent with daily PrEP use. The quantitative assay detected TFV and FTC at lower levels, which also might have provided protection against HIV infection.
Collapse
|
38
|
Helfer AG, Michely JA, Weber AA, Meyer MR, Maurer HH. Liquid chromatography-high resolution-tandem mass spectrometry using Orbitrap technology for comprehensive screening to detect drugs and their metabolites in blood plasma. Anal Chim Acta 2017; 965:83-95. [DOI: 10.1016/j.aca.2017.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 02/06/2017] [Accepted: 03/02/2017] [Indexed: 01/30/2023]
|
39
|
Development and validation of a liquid chromatography-MS/MS method for simultaneous quantification of tenofovir and efavirenz in biological tissues and fluids. J Pharm Biomed Anal 2017; 136:120-125. [DOI: 10.1016/j.jpba.2016.12.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/01/2016] [Accepted: 12/07/2016] [Indexed: 11/20/2022]
|
40
|
Prathipati PK, Mandal S, Destache CJ. Simultaneous quantification of tenofovir, emtricitabine, rilpivirine, elvitegravir and dolutegravir in mouse biological matrices by LC-MS/MS and its application to a pharmacokinetic study. J Pharm Biomed Anal 2016; 129:473-481. [PMID: 27497648 PMCID: PMC5003708 DOI: 10.1016/j.jpba.2016.07.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/20/2016] [Accepted: 07/26/2016] [Indexed: 11/17/2022]
Abstract
Combination antiretroviral (cARV) treatment is more common in human immunodeficiency virus (HIV) infection. In many instances, treatment regimen includes two or more combination of drugs from six different classes. Some of the antiretroviral combination medications are under study at preclinical and clinical stages. A precise method is required to quantify the drug concentration in biological matrices to study pharmacokinetic behavior and tissue distribution profile in animals and/or humans. We have developed and validated a sensitive and precise liquid chromatography-tandem mass spectrometry method for simultaneous quantification of selected antiretroviral drugs, tenofovir (TNF), emtricitabine (FTC), rilpivirine (RPV), dolutegravir (DTG) and elvitegravir (EVG) in mouse biological matrices. This method involves a solid phase extraction, simple isocratic chromatographic separation using Restek Pinnacle DB BiPh column (50mm×2.1mm, 5μm) and mass spectrometric detection by an API 3200 Q Trap instrument. The total run time for each sample was 6min. The method was validated in the concentration range of 5-2000ng/mL for FTC, RPV, DTG, EVG and 10-4000ng/mL for TNF respectively with correlation coefficients (r(2)) higher than 0.9976. The results of intra and inter-run assay precision and accuracy were within acceptance limits for all the five analytes. This method was used to support the study of pharmacokinetics and tissue distribution profile of nanoformulated antiretroviral drugs in mice.
Collapse
Affiliation(s)
- Pavan Kumar Prathipati
- Pharmacy Practice Department, Creighton University School of Pharmacy & Health Professions, Omaha, NE, United States.
| | - Subhra Mandal
- Pharmacy Practice Department, Creighton University School of Pharmacy & Health Professions, Omaha, NE, United States
| | - Christopher J Destache
- Pharmacy Practice Department, Creighton University School of Pharmacy & Health Professions, Omaha, NE, United States
| |
Collapse
|
41
|
Asturias-Arribas L, Delfino MR, Alonso-Lomillo MA, Domínguez-Renedo O, Arcos-Martínez MJ. Electrochemical Oxidation of the Antiretroviral Drug Nelfinavir on Modified Screen-printed Electrodes. ELECTROANAL 2016. [DOI: 10.1002/elan.201600073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Laura Asturias-Arribas
- Analytical Chemistry Department, Faculty of Sciences; University of Burgos.; Plaza Misael Bañuelos s/n 09001 Burgos Spain
| | - Mario Raúl Delfino
- Instrumental Analysis Laboratory, Faculty of Natural and Exact Sciences; National University of Northeast.; Av. Libertad 5460 3400 Corrientes Argentina
- Sensors and Biosensors Laboratory, Faculty of Biochemistry and Biological Sciences; National University of Litoral.; Pje. El Pozo s/n. 3000 Santa Fe Argentina
| | - M. Asunción Alonso-Lomillo
- Analytical Chemistry Department, Faculty of Sciences; University of Burgos.; Plaza Misael Bañuelos s/n 09001 Burgos Spain
| | - Olga Domínguez-Renedo
- Analytical Chemistry Department, Faculty of Sciences; University of Burgos.; Plaza Misael Bañuelos s/n 09001 Burgos Spain
| | - M. Julia Arcos-Martínez
- Analytical Chemistry Department, Faculty of Sciences; University of Burgos.; Plaza Misael Bañuelos s/n 09001 Burgos Spain
| |
Collapse
|
42
|
Maurer HH, Meyer MR. High-resolution mass spectrometry in toxicology: current status and future perspectives. Arch Toxicol 2016; 90:2161-2172. [PMID: 27369376 DOI: 10.1007/s00204-016-1764-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
Abstract
This paper reviews high-resolution mass spectrometry (HRMS) approaches using time-of-flight or Orbitrap techniques for research and application in various toxicology fields, particularly in clinical toxicology and forensic toxicology published since 2013 and referenced in PubMed. In the introduction, an overview on applications of HRMS in various toxicology fields is given with reference to current review articles. Papers concerning HRMS in metabolism, screening, and quantification of pharmaceuticals, drugs of abuse, and toxins in human body samples are critically reviewed. Finally, a discussion on advantages as well as limitations and future perspectives of these methods is included.
Collapse
Affiliation(s)
- H H Maurer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, 66421, Homburg, Saar, Germany.
| | - Markus R Meyer
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
43
|
Pleil JD, Isaacs KK. High-resolution mass spectrometry: basic principles for using exact mass and mass defect for discovery analysis of organic molecules in blood, breath, urine and environmental media. J Breath Res 2016; 10:012001. [DOI: 10.1088/1752-7155/10/1/012001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
44
|
Abstract
OBJECTIVE The Johns Hopkins Hospital Emergency Department has served as a window on the HIV epidemic for 25 years, and as a pioneer in emergency department-based screening/linkage-to-care (LTC) programs. We document changes in the burden of HIV and HIV care metrics to the evolving HIV epidemic in inner-city Baltimore. DESIGN/METHODS We analyzed seven serosurveys conducted on 18 ,144 adult Johns Hopkins Hospital Emergency Department patients between 1987 and 2013 as well as our HIV-screening/LTC program (2007, 2013) for trends in HIV prevalence, cross-sectional annual incidence estimates, undiagnosed HIV, LTC, antiretrovirals treatment, and viral suppression. RESULTS HIV prevalence in 1987 was 5.2%, peaked at more than 11% from 1992 to 2003 and declined to 5.6% in 2013. Seroprevalence was highest for black men (initial 8.0%, peak 20.0%, last 9.9%) and lowest for white women. Among HIV-positive individuals, proportion of undiagnosed infection was 77% in 1987, 28% in 1992, and 12% by 2013 (P < 0.001). Cross-sectional annual HIV incidence estimates declined from 2.28% in 2001 to 0.16% in 2013. Thirty-day LTC improved from 32% (2007) to 72% (2013). In 2013, 80% of HIV-positive individuals had antiretrovirals ARVs detected in sera, markedly increased from 2007 (27%) (P < 0.001). Proportion of HIV-positive individuals with viral suppression (<400 copies/ml) increased from 23% (2001) to 59% (2013) (P < 0.001). CONCLUSION Emergency department-based HIV testing has evolved from describing the local epidemic to a strategic interventional role, serving as a model for early HIV detection and LTC. Our contribution to community-based HIV-screening and LTC program parallels declines in undiagnosed HIV infection and incidence, and increases in antiretroviral use with associated viral suppression in the community.
Collapse
|
45
|
Determination of HIV Status in African Adults With Discordant HIV Rapid Tests. J Acquir Immune Defic Syndr 2015; 69:430-8. [PMID: 25835607 DOI: 10.1097/qai.0000000000000610] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In resource-limited settings, HIV infection is often diagnosed using 2 rapid tests. If the results are discordant, a third tie-breaker test is often used to determine HIV status. This study characterized samples with discordant rapid tests and compared different testing strategies for determining HIV status in these cases. METHODS Samples were previously collected from 173 African adults in a population-based survey who had discordant rapid test results. Samples were classified as HIV positive or HIV negative using a rigorous testing algorithm that included two fourth-generation tests, a discriminatory test, and 2 HIV RNA tests. Tie-breaker tests were evaluated, including rapid tests (1 performed in-country), a third-generation enzyme immunoassay, and two fourth-generation tests. Selected samples were further characterized using additional assays. RESULTS Twenty-nine samples (16.8%) were classified as HIV positive and 24 of those samples (82.8%) had undetectable HIV RNA. Antiretroviral drugs were detected in 1 sample. Sensitivity was 8.3%-43% for the rapid tests; 24.1% for the third-generation enzyme immunoassay; 95.8% and 96.6% for the fourth-generation tests. Specificity was lower for the fourth-generation tests than the other tests. Accuracy ranged from 79.5% to 91.3%. CONCLUSIONS In this population-based survey, most HIV-infected adults with discordant rapid tests were virally suppressed without antiretroviral drugs. Use of individual assays as tie-breaker tests was not a reliable method for determining HIV status in these individuals. More extensive testing algorithms that use a fourth-generation screening test with a discriminatory test and HIV RNA test are preferable for determining HIV status in these cases.
Collapse
|
46
|
Abstract
HIV RNA levels are usually high early in HIV infection. In the HPTN 061 study, men were tested for HIV infection every 6 months; 6 (21.4%) of 28 men who acquired HIV infection during the study had low or undetectable HIV RNA at the time of HIV diagnosis. Antiretroviral drugs were not detected at the time of HIV diagnosis. False-negative HIV test results were obtained for 2 men using multiple assays. Antiretroviral drug resistance mutations were detected in HIV from 1 man. Additional studies are needed to identify factors associated with low HIV RNA levels during early HIV infection.
Collapse
|
47
|
Qu L, Wang W, Zeng D, Lu Y, Yin Z. Quantitative performance of online SPE-LC coupled to Q-Exactive for the analysis of sofosbuvir in human plasma. RSC Adv 2015. [DOI: 10.1039/c5ra20233g] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A quantitative strategy towards the detection of sofosbuvir in human plasma was developed with online-SPE-LC-HRMS using t-MS2, t-SIM and F-SIM modes.
Collapse
Affiliation(s)
- Lihua Qu
- Center of Basic Molecular Science (CBMS)
- Department of Chemistry
- Tsinghua University
- Beijing
- P. R. China
| | - Wenjun Wang
- Center of Basic Molecular Science (CBMS)
- Department of Chemistry
- Tsinghua University
- Beijing
- P. R. China
| | - Debin Zeng
- College of Pharmacy
- State Key Laboratory of Elemento-Organic Chemistry
- Nankai University
- Tianjin
- P. R. China
| | - Yaxin Lu
- College of Pharmacy
- State Key Laboratory of Elemento-Organic Chemistry
- Nankai University
- Tianjin
- P. R. China
| | - Zheng Yin
- Center of Basic Molecular Science (CBMS)
- Department of Chemistry
- Tsinghua University
- Beijing
- P. R. China
| |
Collapse
|
48
|
Piwowar-Manning E, Fogel JM, Laeyendecker O, Wolf S, Cummings V, Marzinke MA, Clarke W, Breaud A, Wendel S, Wang L, Swanson P, Hackett J, Mannheimer S, Del Rio C, Kuo I, Harawa NT, Koblin BA, Moore R, Blankson JN, Eshleman SH. Failure to identify HIV-infected individuals in a clinical trial using a single HIV rapid test for screening. HIV CLINICAL TRIALS 2014; 15:62-8. [PMID: 24710920 PMCID: PMC4167641 DOI: 10.1310/hct1502-62] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND In the HIV Prevention Trials Network (HPTN) 061 study, 8 (2.3%) of 348 HIV-infected participants identified as HIV uninfected at study enrollment using a single HIV rapid test for screening were found to be HIV infected after additional testing. OBJECTIVES To evaluate the performance of different HIV assays for detection of HIV infection in HPTN 061 participants with missed infection and individuals with viral suppression. METHODS Plasma samples from 8 HPTN 061 participants, 17 elite controllers, and 101 individuals on antiretroviral treatment (ART) were tested for HIV with 3 rapid tests, 2 laboratory-based immunoassays, and a Western blot assay. The HPTN 061 samples were also tested with 2 HIV RNA assays and an antiretroviral drug assay. RESULTS Of the 8 HPTN 061 participants with missed infection, 1 was an elite controller, 1 was taking ART, 2 were missed because of testing or clerical errors, 1 had recent HIV infection (identified using a multi-assay algorithm), and 3 had acute HIV infection. Two (1.7%) of 118 individuals with viral suppression (both taking ART) had at least 1 false-negative test. CONCLUSIONS In clinical trials, HIV infections can be missed for a variety of reasons. Using more than one assay to screen for HIV infection may reduce the number of missed infections.
Collapse
Affiliation(s)
| | - Jessica M Fogel
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Oliver Laeyendecker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Shauna Wolf
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vanessa Cummings
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark A Marzinke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sarah Wendel
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Lei Wang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Priscilla Swanson
- Infectious Disease Research, Abbott Diagnostics, Abbott Park, Illinois
| | - John Hackett
- Infectious Disease Research, Abbott Diagnostics, Abbott Park, Illinois
| | - Sharon Mannheimer
- Department of Medicine, Harlem Hospital, Columbia University, Mailman School of Public Health, New York, New York
| | - Carlos Del Rio
- Department of Global Health, Emory University Rollins School of Public Health, Atlanta, Georgia
| | - Irene Kuo
- Department of Epidemiology and Biostatistics, The George Washington University, Washington, DC
| | - Nina T Harawa
- Department of Research, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Beryl A Koblin
- Laboratory of Infectious Disease Prevention, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York
| | - Richard Moore
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joel N Blankson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susan H Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|