1
|
Zhao S, Wang W, Li S, He J, Duan W, Fang Z, Ma X, Li Z, Guo C, Wang W, Wu H, Zhang T, Huang X. The prevalence of low-level viraemia and its association with virological failure in people living with HIV: a systematic review and meta-analysis. Emerg Microbes Infect 2025; 14:2447613. [PMID: 39727007 PMCID: PMC11722027 DOI: 10.1080/22221751.2024.2447613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 12/18/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Low-level viraemia (LLV) following antiretroviral therapy (ART) in people living with HIV (PLWH) has not received sufficient attention. To the determine the prevalence of LLV and its association with virological failure (VF), we systematically reviewed evidence-based interventions for PLWH. We searched PubMed, the Cochrane Library, Embase, and Web of Science from inception to 22 May 2024. Cohorts with samples sizes smaller than 1000 in size were excluded. Data from 16 cohort studies, encompassing 13,49,306 PLWH, revealed a pooled prevalence of LLV of 13.81%. Relative risk (RR) and 95% confidence intervals (CI) identified the following risk factors for LLV: viral load (VL) ≥ 105 copies/mL at baseline (1.79, 1.11-2.88), AIDS-defined illness at baseline (1.24, 1.10-1.40), and protease inhibitor-based regimen at ART initiation (1.53, 1.45-1.62) are the risk factors for LLV. Conversely, CD4 count ≥200 cells/μL at baseline (0.90, 0.82-0.98), non-nucleoside reverse transcriptase inhibitor-based regimen (0.81, 0.68-0.96) and the integrase strand transfer inhibitor (INSTI)-based regimen (0.60, 0.42-0.85) were associated with a reduced risk of LLV. Pooling the adjusted hazard ratio (aHR) and the 95% CI, we found that LLV increased the risk of VF with rising VL among 96,711 PLWH (aHR 2.77, 95% CI 2.03-3.76) and increased the risk of all-cause mortality at high VL levels among 14,229 PLWH (aHR 1.66, 95% CI 1.16-2.37). Therefore, the prevalence of LLV in PLWH should not be overlooked. This study aims to guide better management strategies to improve clinical outcomes in patients with LLV.
Collapse
Affiliation(s)
- Shengnan Zhao
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Wenjing Wang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Sibo Li
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jiaze He
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Wenshan Duan
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zhen Fang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoran Ma
- School of Life Sciences, Tianjin University, Tianjin, People’s Republic of China
| | - Zhen Li
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Caiping Guo
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Wen Wang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Hao Wu
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Tong Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaojie Huang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
2
|
Abdulahi IJ, Björkman P, Abdissa A, Medstrand P, Reepalu A, Elvstam O. Low-level viremia in people with HIV in Ethiopia is associated with subsequent lack of viral suppression and attrition from care. Glob Health Action 2025; 18:2464342. [PMID: 39945786 PMCID: PMC11827032 DOI: 10.1080/16549716.2025.2464342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Low-level viremia during antiretroviral therapy (ART) has been associated with inferior outcomes, but knowledge on the impact of low-level viremia in the current era of dolutegravir-based ART in low-income countries is limited. OBJECTIVE To investigate whether low-level viremia predicts virologic non-suppression and attrition from care in people with HIV receiving ART in Ethiopia. METHODS We included people receiving ART at public health facilities in an urban area in central Ethiopia and categorized persons with ≥1 available viral load 2019-2020 as having either suppression (<150 copies/mL) or low-level viremia (151-1,000 copies/mL); people with >1,000 copies/mL were excluded. We used multivariable logistic regression adjusted for age, sex, ART regimen, type of health facility, and duration of ART to analyze the associations between viremia category and incidence of unsuppressed viral load (>1,000 copies/mL) and attrition from care (death or loss to follow-up) during 3 years of follow-up. RESULTS Among 12,165 participants, the median age was 44 years, 64.2% were female, and 89.1% received tenofovir/lamivudine/dolutegravir. Of the study population, 11,959 (98.3%) had suppression and 206 (1.7%) had low-level viremia. Over 3 years of follow-up, 2.2% of participants with suppression and 11.3% with low-level viremia had unsuppressed viral load. Low-level viremia was associated with both unsuppressed viremia (adjusted odds ratio [aOR], 3.7; 95% confidence interval [CI], 2.2-6.2) and attrition (aOR, 3.4; 95% CI, 1.7-6.6). CONCLUSION Among Ethiopian people with HIV receiving ART, low-level viremia predicted subsequent virologic non-suppression and attrition from care, supporting current recommendations for heightened attention to low-level viremia in ART recipients.
Collapse
Affiliation(s)
- Ilili Jemal Abdulahi
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Agenda 2030 Graduate School, Lund University, Lund, Malmö, Sweden
- Viral Disease Research Division, Armauer Hansen Research Institute, Addis Abeba, Ethiopia
| | - Per Björkman
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Department of Infectious Diseases, Skåne University Hospital, Sweden
| | - Alemseged Abdissa
- Viral Disease Research Division, Armauer Hansen Research Institute, Addis Abeba, Ethiopia
| | - Patrik Medstrand
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Anton Reepalu
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Department of Infectious Diseases, Skåne University Hospital, Sweden
| | - Olof Elvstam
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Department of Infectious Diseases, Växjö Central Hospital, Växjö, Sweden
| |
Collapse
|
3
|
Liu J, Fu C, Yang X, Zhang X, Wei S, Ma J, Zhao Q, Huo Y. HIV-1 subtype distribution and drug resistance profiles among PLWHA with detectable viremia in Henan Province, China, 2023. Sci Rep 2025; 15:17825. [PMID: 40404795 PMCID: PMC12098818 DOI: 10.1038/s41598-025-02949-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 05/16/2025] [Indexed: 05/24/2025] Open
Abstract
In China, Henan Province is a region with a high HIV burden. Furthermore, drug resistance (DR) among people living with HIV/AIDS (PLWHA) on antiretroviral therapy (ART) is becoming a critical barrier to achieving viral suppression. Monitoring DR in PLWHA with detectable viremia after ART is crucial, especially given the widespread use of NNRTI and NRTI, which have historically been the backbone of China's national ART program due to affordability and availability. In this study, plasma samples from PLWHA with viral load (VL) ≥ 40 copies/mL after ≥ 6 months of ART from January 2023 to December 2023 were collected for genotypic DR testing. Of 804 individuals with detectable viremia, a total of 14 HIV-1 subtypes were detected. The top five subtypes were B (55.97%, 450/804), CRF01_AE (22.14%, 178/804), CRF07_BC (15.42%, 124/804), CFR55_01B (3.36%, 27/804), and CRF08_BC (1.00%, 8/804). DR analysis revealed a prevalence of 67.79% (545/804). The resistance rates for non-nucleoside reverse transcriptase inhibitors (NNRTIs), nucleoside reverse transcriptase inhibitors (NRTIs), protease inhibitors (PIs), and integrase strand transfer inhibitors (INSTIs) were 62.94% (506/804), 53.23% (428/804), 7.34% (59/804), and 3.98% (32/804), respectively. The prevalence of DR among PLWHA below and ≥ 200 copies/mL were 40.63% (13/32) and 68.91% (532/772), respectively. Multivariate regression analysis showed that age < 29 years and 40-49 years, baseline CD4 count < 500 cells/µL, current VL of 103-105 copies/mL, and initial ART regimen of NRTIs + NNRTIs were closely associated with the occurrence of DR. These findings highlight the urgent need for expanded access to drug resistance testing and the adoption of WHO-recommended regimens, such as dolutegravir (DTG), to mitigate the growing threat of DR. Our results suggest that drug resistance testing should be applied to all PLWHA with detectable viremia, and treatment strategies should be optimized to align with global guidelines.
Collapse
Affiliation(s)
- Jinjin Liu
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The SixthPeople's Hospital of Zhengzhou), Erqi District, No.29, Jingguang South Road, Zhengzhou, 450000, China
| | - Chaohong Fu
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The SixthPeople's Hospital of Zhengzhou), Erqi District, No.29, Jingguang South Road, Zhengzhou, 450000, China
| | - Xuan Yang
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The SixthPeople's Hospital of Zhengzhou), Erqi District, No.29, Jingguang South Road, Zhengzhou, 450000, China
| | - Xiaohua Zhang
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The SixthPeople's Hospital of Zhengzhou), Erqi District, No.29, Jingguang South Road, Zhengzhou, 450000, China
| | - Shuguang Wei
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The SixthPeople's Hospital of Zhengzhou), Erqi District, No.29, Jingguang South Road, Zhengzhou, 450000, China
| | - Jie Ma
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The SixthPeople's Hospital of Zhengzhou), Erqi District, No.29, Jingguang South Road, Zhengzhou, 450000, China
| | - Qingxia Zhao
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The SixthPeople's Hospital of Zhengzhou), Erqi District, No.29, Jingguang South Road, Zhengzhou, 450000, China.
| | - Yuqi Huo
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The SixthPeople's Hospital of Zhengzhou), Erqi District, No.29, Jingguang South Road, Zhengzhou, 450000, China.
| |
Collapse
|
4
|
McKenzie KP, Nguyen DT, Komba LB, Ketang'enyi EW, Kipiki NE, Mgeyi EN, Mwita LF. Low-level viraemia as a risk factor for virologic failure in children and adolescents living with HIV on antiretroviral therapy in Tanzania: a multicentre, retrospective cohort study. J Int AIDS Soc 2025; 28:e26474. [PMID: 40356263 PMCID: PMC12069799 DOI: 10.1002/jia2.26474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
INTRODUCTION Viral load (VL) of 1000 copies/ml or greater is commonly used to define virologic failure (VF) in children and adolescents living with HIV (CALHIV) in low- and middle-income countries (LMICs). However, evidence in adults suggests that low-level viraemia (LLV) (VL 50-999 copies/ml) increases the risk of subsequent VF. There is limited research on LLV in CALHIV. METHODS This study retrospectively reviewed VL data from Baylor College of Medicine Children's Foundation-Tanzania (sites in Mbeya and Mwanza) collected between January 2015 and December 2022. CALHIV (0-19 years) on antiretroviral therapy for ≥6 months with at least one VL <50 copies/ml plus ≥2 subsequent VLs were included. VF was defined as both VL ≥1000 and ≥200 copies/ml. Multivariable Cox regression models were used to assess the association between LLV and VF, reporting adjusted hazard ratios (aHR) with 95% confidence intervals (CI). RESULTS Among 2618 CALHIV included in the outcome analysis (median age 13.2 years, 52.5% female), 81.9% were on first-line dolutegravir-based regimens and LLV was found in 40.5%. CALHIV with LLV had an increased risk of VF with aHRs of 1.63 (CI 1.38-1.91) (VL ≥1000 copies/ml) and 3.85 (3.33, 4.46) (VL ≥200 copies/ml). When stratifying by LLV (50-199, 200-399 and 400-999 copies/ml), all levels were associated with increased risk for VF (VL ≥1000 copies/ml) with aHRs of 1.39 (1.13, 1.69), 1.69 (1.33, 2.16) and 2.03 (1.63, 2.53). When VF was defined as VL ≥200 copies/ml, the corresponding aHRs were 1.41 (1.15, 1.72), 7.99 (6.68, 9.57) and 9.37 (7.85, 11.18). CONCLUSIONS LLV is associated with a greater risk of VF in CALHIV. The risk of VF increases with higher levels of LLV. This study provides further evidence for revising guidelines in LMICs that define VF as VL ≥1000 copies/ml.
Collapse
Affiliation(s)
- Kevin P. McKenzie
- Department of PediatricsBaylor College of MedicineHoustonTexasUSA
- Texas Children's Global Health NetworkHoustonTexasUSA
- Baylor College of Medicine Children's Foundation – TanzaniaMbeyaTanzania
| | - Duc T. Nguyen
- Department of PediatricsBaylor College of MedicineHoustonTexasUSA
| | - Lilian B. Komba
- Baylor College of Medicine Children's Foundation – TanzaniaMbeyaTanzania
| | | | - Neema E. Kipiki
- Baylor College of Medicine Children's Foundation – TanzaniaMwanzaTanzania
| | - Evance N. Mgeyi
- Baylor College of Medicine Children's Foundation – TanzaniaMbeyaTanzania
| | - Lumumba F. Mwita
- Baylor College of Medicine Children's Foundation – TanzaniaMbeyaTanzania
- Baylor College of Medicine Children's Foundation – TanzaniaMwanzaTanzania
| |
Collapse
|
5
|
Yu H, Yang Y, Cao D, Cao Y, Shi Y, Xiao G, Li P, Feng Y, Wei H, Sun J, Duan S, Ye R, Jin C. Association of HIV low-level viremia with CD4 count recovery among persons living with HIV during antiretroviral therapy in Dehong Prefecture, Southwest China in 2008-2021: a longitudinal study. BMC Infect Dis 2025; 25:586. [PMID: 40269740 PMCID: PMC12020194 DOI: 10.1186/s12879-025-10940-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND The impact of human immunodeficiency virus (HIV) low-level viremia (LLV) on CD4 + T lymphocyte (CD4) cell count recovery during antiretroviral therapy (ART) remains unknown in China. This study aimed to investigate the association between LLV and CD4 count recovery among adults on ART in Southwest China. METHODS A longitudinal cohort study of persons living with HIV (PLHIV) were conducted in Dehong Prefecture, Southwest China. Incidence of CD4 count recovery (CD4 cell count ≥ 500 cells/µl) was calculated for each follow-up year and characteristics of LLV (VL between 50 and 999 copies/ml) were described. Group-based trajectory model (GBTM) was used to identify and characterize the trajectories of CD4 cell count and VL during follow up. Longitudinal associations between LLV and CD4 count recovery were examined using a generalized estimating equation (GEE) with LLV as a time-updated variable. RESULTS The study included a total of 7,485 PLHIV who received ART between 2008 and 2021 in Dehong. The median follow-up duration was 8.5 years. At baseline, the participants had a median age of 36 years, with males accounting for 60.5%. The median CD4 cell count at baseline was 268 cells/µl. Results of GBTM demonstrated that 730 patients (9.8%) experience LLV trajectories and 2,125 patients (28.4%) reached CD4 count recovery during follow-up. Compared to participants with the trajectory of VL < 50 copies/ml, the probability of CD4 count recovery were lower among participants with the trajectories of LLV 50-199 copies/ml (adjusted odds ratio [aOR] 0.69, 95% confidence interval [CI] 0.63-0.76) and LLV 200-999 copies/ml (aOR 0.51, 95% CI 0.45-0.59), CONCLUSION: Sustained LLV is associated with poorer CD4 recovery among HIV patients who are receiving ART. Interventions to ensure that PLHIV maintain durably undetectable VL during treatment should be prioritized to achieve immune recovery.
Collapse
Affiliation(s)
- Hailiang Yu
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Yuecheng Yang
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, Dehong, 678400, China
| | - Dongdong Cao
- Dehong Dai and Jingpo Autonomous Prefecture People's Hospital, Dehong, 678499, China
| | - Yanfen Cao
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, Dehong, 678400, China
| | - Yun Shi
- Mangshi People's Hospital, Dehong Dai and Jingpo Autonomous Prefecture, Dehong, 678499, China
| | - Guifang Xiao
- Traditional Chinese Medicine Hospital of Dehong Dai and Jingpo Autonomous Prefecture, Dehong, 678400, China
| | - Pinyin Li
- Ruili City People's Hospital of Dehong Dai and Jingpo Autonomous Prefecture, Dehong, 678600, China
| | - Yanling Feng
- Longchuan County People's Hospital of Dehong Dai and Jingpo Autonomous Prefecture, Dehong, 678700, China
| | - Hua Wei
- Yingjiang County People's Hospital of Dehong Dai and Jingpo Autonomous Prefecture, Dehong, 678300, China
| | - Jinting Sun
- Lianghe County People's Hospital of Dehong Dai and Jingpo Autonomous Prefecture, Dehong, 679200, China
| | - Song Duan
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, Dehong, 678400, China
| | - Runhua Ye
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, Dehong, 678400, China.
| | - Cong Jin
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
| |
Collapse
|
6
|
Shu Y, Liu J, Yang C, Li J, Zhang M, Li Y, Deng X, Dong X. Prevalence of drug resistance mutations in low-level viremia patients under antiretroviral therapy in Southwestern China: a cross-sectional study. J Antimicrob Chemother 2025; 80:947-954. [PMID: 39835338 PMCID: PMC11962373 DOI: 10.1093/jac/dkaf017] [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: 07/26/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVES This study aimed to evaluate the prevalence and characteristics of drug resistance mutations (DRMs) in patients with low-level viremia (LLV) in Southwestern China, as it has become a growing challenge in AIDS clinical practice. METHODS This cross-sectional study was performed in Yunnan Province, Southwestern China. LLV was defined as 50-999 copies/mL of plasma viral load with antiretroviral therapy (ART) for at least 6 months. HIV-1 DRM detection used validated in-house protocol. RESULTS A total of 470 sequences were obtained, and 13 HIV-1 genotypes were identified, among which CRF08_BC (47.5%), CRF07_BC (22.3%) and CRF01_AE (10.0%) subtypes were the most prevalent. The overall prevalence of DRMs was 45.7% (215/470), and the prevalence of DRMs to non-nucleoside reverse transcriptase inhibitors (NNRTIs), nucleoside reverse transcriptase inhibitors (NRTIs) and protease inhibitors (PIs) was 39.4% (185/470), 20.6% (97/470) and 5.3% (25/470), respectively. The most common NNRTI-associated mutations were K103N (16.0%), E138A (6.6%), V179D (6.6%) and P225H (4.9%), and those in NRTIs were M184V (17.0%), D67N (3.4%) and K65R (3.0%). PI-associated mutations were infrequent, occurring in less than 1.8% of cases. The prevalence of NNRTI-associated mutations (K101E and Y188C) was found to be statistically significant among various LLV groups. Additionally, significant variations were observed in the prevalence of NNRTI-associated mutations (V106I, V106M, E138A and P225H), NRTI-associated mutation (K65R) and PI-associated mutations (L33F and Q58E) across different subtypes. CONCLUSIONS The prevalence of DRMs in ART-experienced patients with LLV was high, and HIV-1 genotypes exhibited diversity in Yunnan Province. These findings indicate that regular DRM monitoring during LLV episodes was essential for effective clinical treatment and management in this region.
Collapse
Affiliation(s)
- Yuanlu Shu
- School of Public Health, Kunming Medical University, Kunming 650500, China
- Department of Laboratory Medicine, Yunnan Provincial Infectious Disease Hospital, Kunming 650301, China
| | - Jiafa Liu
- Department of Public Health, Yunnan Provincial Infectious Disease Hospital, Kunming 650301, China
| | - Cuixian Yang
- Department of Laboratory Medicine, Yunnan Provincial Infectious Disease Hospital, Kunming 650301, China
| | - Jianjian Li
- Department of Laboratory Medicine, Yunnan Provincial Infectious Disease Hospital, Kunming 650301, China
| | - Mi Zhang
- Department of Science and Education, Yunnan Provincial Infectious Disease Hospital, Kunming 650301, China
| | - Yuan Li
- School of Public Health, Kunming Medical University, Kunming 650500, China
- Department of Laboratory Medicine, Yunnan Provincial Infectious Disease Hospital, Kunming 650301, China
| | - Xuemei Deng
- Department of Laboratory Medicine, Yunnan Provincial Infectious Disease Hospital, Kunming 650301, China
| | - Xingqi Dong
- Department of Laboratory Medicine, Yunnan Provincial Infectious Disease Hospital, Kunming 650301, China
| |
Collapse
|
7
|
Cao B, Liu M, Song S, Guo M, Tang L, Ding P, Yuan T, Wang T, Zhong L. HIV-1 DNA Genotypic Drug Resistance Testing Guides Antiretroviral Therapy in Patients with Low-Level Viremia. AIDS Res Hum Retroviruses 2025; 41:197-202. [PMID: 39807872 DOI: 10.1089/aid.2024.0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
In 2023, we published a case study involving a 10-year-old child infected with HIV-1 with low-level viremia (LLV). We showed that this child patient achieved successful viral suppression by modifying the antiretroviral therapy (ART) regimen according to the HIV-1 DNA genotypic drug resistance testing. In this study, we aimed to address whether HIV-1 DNA genotypic drug resistance testing could direct successfully virological suppression in patients infected with HIV-1 experiencing persistent LLV based on evidence from a cohort study. The subjects of this study were all people living with HIV-1 who received ART and followed in the Yuexi County (Liangshan, China) from December 2010 to February 2024. From June 2021 to February 2024, a total of 10 mL of peripheral blood was collected from each subject at each follow-up and separated. HIV-1 RNA and HIV-1 DNA were quantified, followed by HIV-1 genotypic drug resistance testing. ART regimens were accordingly adjusted, while follow-up tests were performed in terms of HIV-1 RNA and DNA measurements. The prevalent HIV-1 DNA drug resistance mutations (DRMs) included M184V, K103N, K101E/P, and V108I. The primary resistance mutations observed for nucleoside reverse transcriptase inhibitor (NRTI) were against abacavir, lamivudine, and emtricitabine. For non-NRTI, the primary DRMs were associated with efavirenz and nevirapine. Five out of the six patients were subjected to regimen adjustments according to HIV-1 DNA DRMs, while one patient was continuously treated with unchanged regimen. Viral suppression was achieved in all five ART-changed cases, with observation of remarkable of HIV-1 DNA decline. The ART-unchanged case showed progressive treatment failure with drastic increase of plasma HIV-1 RNA and whole blood HIV-1 DNA. For patients with LLV, HIV-1 DNA genotypic drug resistance testing directed ART regimen considerations are highly recommended for achieving viral suppression.
Collapse
Affiliation(s)
- Bianchuan Cao
- Department of Infectious Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Tuberculosis, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Infection and Immune Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mei Liu
- Antiviral Therapy Center, The First People's Hospital of Yuexi County, Liangshan, China
| | - Shaofang Song
- Antiviral Therapy Center, The First People's Hospital of Yuexi County, Liangshan, China
| | - Mingxian Guo
- Antiviral Therapy Center, The First People's Hospital of Yuexi County, Liangshan, China
| | - Lingyu Tang
- Antiviral Therapy Center, The First People's Hospital of Yuexi County, Liangshan, China
| | - Ping Ding
- Antiviral Therapy Center, The First People's Hospital of Yuexi County, Liangshan, China
| | - Tianru Yuan
- Antiviral Therapy Center, The First People's Hospital of Yuexi County, Liangshan, China
| | - Tong Wang
- Center of Clinical Laboratory, The First Affiliated Hospital, MOE Key Laboratory of Tumor Molecular Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Li Zhong
- Department of Infectious Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Tuberculosis, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Infection and Immune Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
8
|
Yang X, Cai R, Ma Y, Zhang HH, Sun X, Olatosi B, Weissman S, Li X, Zhang J. Using Machine Learning Techniques to Predict Viral Suppression Among People With HIV. J Acquir Immune Defic Syndr 2025; 98:209-216. [PMID: 39561000 PMCID: PMC11798697 DOI: 10.1097/qai.0000000000003561] [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: 05/28/2023] [Accepted: 08/16/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND This study aims to develop and examine the performance of machine learning (ML) algorithms in predicting viral suppression among statewide people living with HIV (PWH) in South Carolina. METHODS Extracted through the electronic reporting system in South Carolina, the study population was adult PWH who were diagnosed between 2005 and 2021. Viral suppression was defined as viral load <200 copies/mL. The predictors, including sociodemographics, a historical information of viral load indicators (eg, viral rebound), comorbidities, health care utilization, and annual county-level factors (eg, social vulnerability), were measured in each 4-month windows. Using historic information in different lag time windows (1-, 3- or 5-lagged time windows with each 4-month window as a unit), both traditional and ML approaches (eg, Long Short-Term Memory Network) were applied to predict viral suppression. Comparisons of prediction performance between different models were assessed by area under curve (AUC), recall, precision, F1 score, and Youden index. RESULTS ML approaches outperformed the generalized linear mixed model. In all the 3 lagged analysis of a total of 15,580 PWH, the Long Short-Term Memory Network (Lag 1: AUC = 0.858; Lag 3: AUC = 0.877; Lag 5: AUC = 0.881) algorithm outperformed all the other methods in terms of AUC performance for predicting viral suppression. The top-ranking predictors that were common in different models included historical information of viral suppression, viral rebound, and viral blips in the Lag-1 time window. Inclusion of county-level variables did not improve the model prediction accuracy. CONCLUSIONS Supervised ML algorithms may offer better performance for risk prediction of viral suppression than traditional statistical methods.
Collapse
Affiliation(s)
- Xueying Yang
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
- Department of Health Promotion, Education and Behavior, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
| | - Ruilie Cai
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
| | - Yunqing Ma
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
| | - Hao H. Zhang
- Department of Mathematics, University of Arizona, Tucson, AZ, USA, 85721
| | - XiaoWen Sun
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
| | - Bankole Olatosi
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
- Department of Health Services Policy and Management, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
| | - Sharon Weissman
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
- Department of Internal Medicine, School of Medicine, University of South Carolina, Columbia, SC, USA, 29208
| | - Xiaoming Li
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
- Department of Health Promotion, Education and Behavior, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
| | - Jiajia Zhang
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA, 29208
| |
Collapse
|
9
|
Lu X, Li Y, Liu M, Wang Y, An N, Sun D, Li Q. Drug resistance mutations to integrase inhibitors, proteinase, and reverse transcriptase inhibitors in newly diagnosed HIV-1 infections in Hebei province, China, 2018-2022. Front Cell Infect Microbiol 2025; 15:1510916. [PMID: 40066069 PMCID: PMC11891186 DOI: 10.3389/fcimb.2025.1510916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/21/2025] [Indexed: 05/13/2025] Open
Abstract
Background HIV-1 protease (PR)-reverse transcriptase (RT) inhibitors as national free antiretroviral drugs have been used for 20 years. Integrase strand transfer inhibitors (INSTIs) have been conditionally used as a component of HIV/AIDS treatment regimens in recent years. However, the systematic investigation on the changes in primary drug resistance (PDR) in Hebei province, China was limited. Methods A continuous cross-sectional investigation on HIV-1 PDR was conducted, integrating detection of drug resistance genotype, molecular network, and statistical analysis. Results The overall prevalence of PDR was 8.3%, with 77 of 925 samples showing different levels of resistance to INSTIs (1.9%), protease inhibitors (PIs, 0.2%), nucleoside reverse transcriptase inhibitors (NRTIs, 1.2%), and non-NRTIs (NNRTIs, 5.2%). In the PR-RT gene coding region, E138EK/G was the most common (1.6%), followed by K103N (1.4%), G190GE/A/S (0.6%), K101E (0.5%), A98G (0.4%), and T215I/TS (0.3%), associated with the low- to high-level resistance to doravirine (DOR), efavirenz (EFV), etravirine (ETR), nevirapine (NVP), rilpivirine (RPV), and zidovudine (AZT). In the INSTI gene coding region, six mutations were identified, namely, four major mutations (P145PS, Q148QH, Y143S, and T66A) and two accessory mutations (S153SF and G163GRS/EK). Of these mutations, the most frequent INSTI mutations were S153SF (0.6%) and G163GRS/EK (0.6%), followed by P145PS (0.2%), Y143S (0.2%), Q148QH (0.1%), and T66A (0.1%). G163GRS/EK, P145PS, Y143S, and T66A were associated with the resistance to elvitegravir (EVG) and raltegravir (RAL). S153SF and Q148QH were mainly related to the resistance to dolutegravir (DTG), bictegravir (BIC), and caboteravir (CAB). Furthermore, 30 resistant sequences were circulating in 16 transmission networks with HIV-1 DR mutations (DRMs), accounting for 62.5% of 77 total participants with DRMs. Multivariable analysis showed that those who had CRF07_BC had 1.79 times greater odds of PDR compared with participants with CRF01_AE. Compared to participants with volunteer blood donor, those with voluntary consultation and testing had 0.27 times greater odds of PDR. Conclusions The overall prevalence of HIV-1 PDR in Hebei is high, belonging to a moderate resistant level (5.0%-15.0%). It is necessary for us to strengthen the effective surveillance of PDR among treatment-naive patients, and we should adjust the treatment plan according to the results of PDR surveillance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qi Li
- Department of AIDS Research, Hebei Key Laboratory of Pathogen and Epidemiology of Infectious Disease, Hebei Provincial Center for Disease Control and Prevention, Shijiazhuang, Hebei, China
| |
Collapse
|
10
|
Yu H, Ye R, Cao D, Cao Y, Shi Y, Xiao G, Li P, Feng Y, Wei H, Sun J, Duan S, Yang Y, Jin C. Associations of First-Year Low-Level Viremia with Subsequent Viral Non-Suppression in People Living with HIV on Antiretroviral Therapy - Dehong Dai and Jingpo Autonomous Prefecture, Yunnan Province, China, 2008-2021. China CDC Wkly 2025; 7:45-52. [PMID: 39866288 PMCID: PMC11754852 DOI: 10.46234/ccdcw2025.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
What is already known about this topic? Human immunodeficiency virus (HIV) low-level viremia (LLV) during antiretroviral therapy (ART) occurs frequently in Dehong Dai and Jingpo Autonomous Prefecture, Yunnan Province. What is added by this report? Among people living with HIV who achieved virological success [viral load (VL) <1,000 copies/mL] after initiating ART in Dehong Prefecture, Southwest China, 17.6% experienced first-year LLV of 50-999 copies/mL First-year LLV emerged as an independent risk factor for subsequent viral non-suppression compared with participants maintaining first-year VL <50 copies/mL. What are the implications for public health practice? Enhanced monitoring and interventions for early LLV occurrence during the first year of ART are essential, including adherence education and timely VL testing.
Collapse
Affiliation(s)
- Hailiang Yu
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Runhua Ye
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, Dehong Prefecture, Yunnan Province, China
| | - Dongdong Cao
- Dehong Dai and Jingpo Autonomous Prefecture People’s Hospital, Dehong Prefecture, Yunnan Province, China
| | - Yanfen Cao
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, Dehong Prefecture, Yunnan Province, China
| | - Yun Shi
- Mangshi People’s Hospital, Dehong Dai and Jingpo Autonomous Prefecture, Dehong Prefecture, Yunnan Province, China
| | - Guifang Xiao
- Traditional Chinese Medicine Hospital of Dehong Dai and Jingpo Autonomous Prefecture, Dehong Prefecture, Yunnan Province, China
| | - Pinyin Li
- Ruili City People’s Hospital of Dehong Dai and Jingpo Autonomous Prefecture, Dehong Prefecture, Yunnan Province, China
| | - Yanling Feng
- Longchuan County People’s Hospital of Dehong Dai and Jingpo Autonomous Prefecture, Dehong Prefecture, Yunnan Province, China
| | - Hua Wei
- Yingjiang County People’s Hospital of Dehong Dai and Jingpo Autonomous Prefecture, Dehong Prefecture, Yunnan Province, China
| | - Jinting Sun
- Lianghe County People’s Hospital of Dehong Dai and Jingpo Autonomous Prefecture, Dehong Prefecture, Yunnan Province, China
| | - Song Duan
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, Dehong Prefecture, Yunnan Province, China
| | - Yuecheng Yang
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, Dehong Prefecture, Yunnan Province, China
| | - Cong Jin
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
11
|
Ruffieux Y, Mwansa-Kambafwile J, Metekoua C, Tombe-Nyahuma T, Bohlius J, Muchengeti M, Egger M, Rohner E. HIV-1 Viremia and Cancer Risk in 2.8 Million People: the South African HIV Cancer Match Study. Clin Infect Dis 2024:ciae652. [PMID: 39736138 DOI: 10.1093/cid/ciae652] [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: 10/10/2024] [Revised: 12/12/2024] [Accepted: 12/27/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Most research on HIV-1 viremia and cancer risk is from high-income countries. We evaluated the association between HIV-1 viremia and the risk of various cancer types among people with HIV (PWH) in South Africa. METHODS We analysed data from the South African HIV Cancer Match study, based on laboratory measurements from the National Health Laboratory Services and cancer records from the National Cancer Registry from 2004-2014. Using Cox proportional hazards models, we estimated hazard ratios (HR) for cancer incidence per unit increase in time-updated Log10 HIV-1 RNA viral load copies/mL. We created partially adjusted (sex, age, calendar year) and fully adjusted models (additionally including time-updated CD4 count). RESULTS We included 2,770,200 PWH with 10,175 incident cancers; most common were cervical cancer (N=2,481), Kaposi sarcoma (N=1,902), breast cancer (N=1,063), and non-Hodgkin lymphoma (N=863). Hazard ratios for the association of HIV-1 viremia and cancer risk changed after partial and full adjustment and were generally attenuated for infection-related cancers but tended to increase for infection-unrelated cancers. In the fully adjusted model, HIV-1 viremia was associated with an increased risk of Kaposi sarcoma (HR per unit increase in Log10 HIV-1 RNA viral load: 1.38, 95% CI 1.35-1.42), leukemia (HR: 1.28, 95% CI 1.13-1.45), non-Hodgkin lymphoma (HR: 1.24, 95% CI 1.19-1.29), conjunctival cancer (HR: 1.19, 95% CI 1.11-1.25), and colorectal cancer (HR: 1.11, 95% CI 1.02-1.21). Associations with other cancer types were weaker or absent. CONCLUSIONS Our findings underline the importance of sustained viral suppression for cancer prevention among PWH in South Africa.
Collapse
Affiliation(s)
- Yann Ruffieux
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Judith Mwansa-Kambafwile
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
- School of Public Health, University of Witwatersrand, Johannesburg, South Africa
| | - Carole Metekoua
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
- Graduate School for Health Sciences, University of Bern, Switzerland
| | - Tinashe Tombe-Nyahuma
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
| | - Julia Bohlius
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Mazvita Muchengeti
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
- School of Public Health, University of Witwatersrand, Johannesburg, South Africa
- South African DSI-NRF Centre of Excellence in Epidemiological Modelling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa
| | - Matthias Egger
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Centre for Infectious Disease Epidemiology and Research (CIDER), School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Eliane Rohner
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
12
|
Lee SY, Lin YC, Chen CP, Cheng SH, Chang SY, Ku SY, Cheng CY. Assessment of risk factors for virological nonsuppression following switch to dolutegravir and lamivudine, or bictegravir, emtricitabine, and tenofovir alafenamide fumarate in a real-world cohort of treatment-experienced adults living with HIV. PLoS One 2024; 19:e0314003. [PMID: 39565750 PMCID: PMC11578452 DOI: 10.1371/journal.pone.0314003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024] Open
Abstract
Conflicting data exists regarding the baseline determinants of virological nonsuppression outcomes in treatment-experienced people living with human immunodeficiency virus (PWH) switching to antiretroviral treatment (ART) with bictegravir/emtricitabine/tenofovir alafenamide fumarate (BIC/FTC/TAF) or dolutegravir/lamivudine (DTG/3TC) in Asia. This retrospective observational study, conducted at a designated HIV-care hospital from October 2019 to January 2023, aimed to address this gap. We assessed the odds of virological nonsuppression (VNS) at weeks 48 using logistic regression. A total of 988 patients were included, 35 patients (3.5%) with VNS at week 48. Pre-existing primary resistance-associated mutations (RAM) to nucleoside reverse transcriptase inhibitor (NRTI) and non-nucleoside reverse transcriptase inhibitor (NNRTI) were identified in 11.0% (51/465) and 14.4% (67/465), respectively. The identified risk factor was a record of virological failure ≥2 times (AOR 5.32, 95% CI 2.04-13.85), while an HIV viral load <50 copies/mL within the past three months before switch (AOR: 0.27, 95% CI 0.11-0.72) was identified as a protective factor. No cases of acquired drug resistance-associated mutations were detected at week 48. Additionally, DTG/3TC was noninferior to BIC/FTC/TAF in achieving or maintaining HIV RNA levels of <50 copies/mL, within a -10% noninferiority margin in the per-protocol analysis (responder proportion: 98.2% vs. 95.0%, respectively; adjusted treatment difference [95% CI], 3.2% [0.7% to 5.3%]). In conclusion, DTG/3TC and BIC/FTC/TAF demonstrated good effectiveness in a real-world cohort, but frequent virological failure before the switch might impact the benefits of these regimens in the short term of follow-up.
Collapse
Affiliation(s)
- Shu-Yuan Lee
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan City, Taiwan
| | - Yi-Chun Lin
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan City, Taiwan
| | - Cheng-Pin Chen
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan City, Taiwan
- School of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei City, Taiwan
| | - Shu-Hsing Cheng
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan City, Taiwan
- School of Public Health, Taipei Medical University, Taipei City, Taiwan
| | - Shu-Ying Chang
- Department of Nursing, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan City, Taiwan
| | - Shin-Yen Ku
- Department of Nursing, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan City, Taiwan
| | - Chien-Yu Cheng
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan City, Taiwan
- Institute of Public Health, School of Medicine, National Yang-Ming Chiao Tung University, Taipei City, Taiwan
| |
Collapse
|
13
|
Zhang W, Shi J, Wang Y, Li E, Yan D, Zhang Z, Zhu M, Yu J, Wang Y. Risk factors and clinical prediction models for low-level viremia in people living with HIV receiving antiretroviral therapy: an 11-year retrospective study. Front Microbiol 2024; 15:1451201. [PMID: 39552647 PMCID: PMC11563986 DOI: 10.3389/fmicb.2024.1451201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Objective This study explores the risk factors for low-level viremia (LLV) occurrence after ART and develops a risk prediction model. Method Clinical data and laboratory indicators of people living with HIV (PLWH) at Hangzhou Xixi Hospital from 5 April 2011 to 29 December 2022 were collected. LASSO Cox regression and multivariate Cox regression analysis were performed to identify laboratory indicators and establish a nomogram for predicting LLV occurrence. The nomogram's discrimination and calibration were assessed via ROC curve and calibration plots. The concordance index (C-index) and decision curve analysis (DCA) were used to evaluate its effectiveness. Result Predictive factors, namely, age, ART delay time, white blood cell (WBC) count, baseline CD4+ T-cell count (baseline CD4), baseline viral load (baseline VL), and total bilirubin (TBIL), were incorporated into the nomogram to develop a risk prediction model. The optimal model (which includes 6 variables) had an AUC for LLV after 1-year, 3-year, and 5-year of listing of 0.68 (95% CI, 0.61-0.69), 0.69 (95% CI, 0.65-0.70), and 0.70 (95% CI, 0.66-0.71), respectively. The calibration curve showed high consistency between predicted and actual observations. The C-index and DCA indicated superior prediction performance of the nomogram. There was a significant difference in CD4 levels between LLV and non-LLV groups during the follow-up time. The dynamic SCR, ALT, TG and BG levels and occurrence of complications differed significantly between the high- and low-risk groups. Conclusion A simple-to-use nomogram containing 6 routinely detected variables was developed for predicting LLV occurrence in PLWH after ART.
Collapse
Affiliation(s)
- Wenhui Zhang
- Department of Infection, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Nursing, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinchuan Shi
- Department of Infection, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Wang
- Medical Laboratory, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Er Li
- Department of Nursing, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dingyan Yan
- Department of Infection, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Nursing, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhongdong Zhang
- Department of Infection, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mingli Zhu
- Medical Laboratory, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianhua Yu
- Department of Infection, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Wang
- Department of Infection, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
- Clinical Research Laboratory, Hangzhou Xixi Hospital, Zhejiang University of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
14
|
Sebeza J, Mbwana MS, Ramadhani HO, Ally ZM, Lascko T, Memiah P, Tuyishime S, Rwibasira G. Association between differentiated HIV care delivery model and low-level viremia among people living with HIV in Rwanda. AIDS Res Ther 2024; 21:79. [PMID: 39487508 PMCID: PMC11529002 DOI: 10.1186/s12981-024-00650-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 09/02/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Low-level viremia (LLV) (HIV-RNA 51-999 copies/mL) is associated with increased risk of non viral load suppression (HIV-RNA ≥ 1000 copies/mL). We assessed the association between differentiated service delivery model (DSDM) and LLV among people living with HIV (PLHIV) in Rwanda. METHODS We conducted a retrospective cohort analysis using routinely collected data of adults living with HIV from 28-healthcare facilities in Rwanda before and after the introduction of DSDM. Under DSDM, PLHIV initiated treatment within seven days of HIV diagnosis and medication pick-up up to six months for those with sustained viral load suppression suppression. Proportions of LLV at 6,12 and 18 months were quantified. Multivariable log binomial regression models were used to assess the effect of DSDM on LLV. To handle missing data, multiple imputations was performed. RESULTS Of 976 people living with HIV, 645(66.0%) were female and 463(47.4%) initiated treatment during DSDM. The median age was 37 (interquartile range: 32-43) years. LLV was 7.4%, 6.6% and 5.4%, at 6,12 and 18 months, respectively. Compared to those who initiated treatment before DSDM, starting treatment during DSDM increased six-month LLV [adjusted risk ratio (aRR) = 2.8: 95%CI (1.15-6.91)] but not at 12 [aRR = 2.3: 95%CI (0.93-5.75)] and 18 months [aRR = 0.3: 95%CI (0.09-1.20)]. Using imputed datasets, the association between DSDM and LLV persisted. CONCLUSIONS DSDM was associated with increased risk of LLV at 6-months. possibly due to the minimal amount of time PLHIV had in pondering and accepting the HIV diagnosis. Continued support is needed among people receiving early antiretroviral therapy initiation to prevent development of LLV.
Collapse
Affiliation(s)
- Jackson Sebeza
- College of Medicine and Health Sciences, School of Public Health, University of Rwanda, Street Name: KG 11 Ave Gasabo Kigali, P.O. Box 3286, Kigali, Rwanda.
- Primary Health Care Institute, Iringa, Tanzania.
| | | | - Habib O Ramadhani
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, USA
| | | | - Taylor Lascko
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, USA
| | - Peter Memiah
- Graduate school, University of Maryland, Baltimore, USA
| | | | | |
Collapse
|
15
|
Aldredge A, Mehta CC, Lahiri CD, Schneider MF, Alcaide ML, Anastos K, Plankey M, French AL, Floris-Moore M, Tien PC, Dionne J, Dehovitz J, Collins LF, Sheth AN. Consequences of low-level viremia among women with HIV in the United States. AIDS 2024; 38:1829-1838. [PMID: 39110550 PMCID: PMC11424065 DOI: 10.1097/qad.0000000000003990] [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: 04/11/2024] [Revised: 06/30/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024]
Abstract
OBJECTIVE Investigate the outcomes of women with HIV (WWH) with low-level viremia (LLV). DESIGN The prevalence of LLV and potential clinical sequelae, such as virologic failure and non-AIDS comorbidity (NACM) development, are poorly characterized among WWH. METHODS We analyzed data from the Women's Interagency HIV Study among WWH enrolled from 2003 to 2020 who reported antiretroviral therapy use at least 1 year followed by an HIV-1 viral load less than 200 copies/ml. Consecutive viral load measurements from four semi-annual visits were used to categorize women at baseline as having: virologic suppression (all viral load undetectable), intermittent LLV (iLLV; nonconsecutive detectable viral load up to 199 copies/ml), persistent LLV (pLLV; at least two consecutive detectable viral load up to 199 copies/ml), or virologic failure (any viral load ≥200 copies/ml). Adjusted hazard ratios quantified the association of virologic category with time to incident virologic failure and multimorbidity (≥2 of 5 NACM) over 5-year follow-up. RESULTS Of 1598 WWH, baseline median age was 47 years, 64% were Black, 21% Hispanic, and median CD4 + cell count was 621 cells/μl. After excluding 275 women (17%) who had virologic failure at baseline, 58, 19, and 6% were categorized as having virologic suppression, iLLV, and pLLV, respectively. Compared with WWH with virologic suppression, the adjusted hazard ratio [aHR; 95% confidence interval (CI)] for incident virologic failure was 1.88 (1.44-2.46) and 2.51 (1.66-3.79) for iLLV and pLLV, respectively; and the aHR for incident multimorbidity was 0.81 (0.54-1.21) and 1.54 (0.88-2.71) for iLLV and pLLV, respectively. CONCLUSION Women with iLLV and pLLV had an increased risk of virologic failure. Women with pLLV had a trend towards increased multimorbidity risk.
Collapse
Affiliation(s)
- Amalia Aldredge
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Healthcare System, Ponce de Leon Center
| | - C Christina Mehta
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Cecile D Lahiri
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Healthcare System, Ponce de Leon Center
| | - Michael F Schneider
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Maria L Alcaide
- Division of Infectious Diseases, Department of Medicine, University of Miami, Miami, FL
| | - Kathryn Anastos
- Division of General Internal Medicine, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Michael Plankey
- Division of General Internal Medicine, Department of Medicine, Georgetown University, DC
| | - Audrey L French
- Division of Infectious Diseases, Department of Medicine, Stroger Hospital of Cook County, Chicago, IL
| | - Michelle Floris-Moore
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Phyllis C Tien
- Division of Infectious Diseases, Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Jodie Dionne
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Jack Dehovitz
- Division of Infectious Diseases, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Lauren F Collins
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Healthcare System, Ponce de Leon Center
| | - Anandi N Sheth
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Healthcare System, Ponce de Leon Center
| |
Collapse
|
16
|
Campagna R, Nonne C, Antonelli G, Turriziani O. Archived HIV-1 Drug Resistance Mutations: Role of Proviral HIV-1 DNA Genotype for the Management of Virological Responder People Living with HIV. Viruses 2024; 16:1697. [PMID: 39599811 PMCID: PMC11599110 DOI: 10.3390/v16111697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Despite its effectiveness in controlling plasma viremia, antiretroviral therapy (ART) cannot target proviral DNA, which remains an obstacle to HIV-1 eradication. When treatment is interrupted, the reservoirs can act as a source of viral rebound, highlighting the value of proviral DNA as an additional source of information on an individual's overall resistance burden. In cases where the viral load is too low for successful HIV-1 RNA genotyping, HIV-1 DNA can help identify resistance mutations in treated individuals. The absence of treatment history, the need to adjust ART despite undetectable viremia, or the presence of LLV further support the use of genotypic resistance tests (GRTs) on HIV-1 DNA. Conventionally, GRTs have been achieved through Sanger sequencing, but the advances in NGS are leading to an increase in its use, allowing the detection of minority variants present in less than 20% of the viral population. The clinical significance of these mutations remains under debate, with interpretations varying based on context. Additionally, proviral DNA is subject to APOBEC3-induced hypermutation, which can lead to defective, nonviable viral genomes, a factor that must be considered when performing GRTs on HIV-1 DNA.
Collapse
Affiliation(s)
- Roberta Campagna
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (C.N.); (G.A.); (O.T.)
| | | | | | | |
Collapse
|
17
|
Tian RR, Li T, Zhang MX, Song TZ, Zheng HY, Zheng YT. Nonnegligible Contribution of Nonlymphoid Tissue to Viral Reservoir During the Short-Term Early cART in SIVmac239-Infected Chinese Rhesus Macaques. AIDS Res Hum Retroviruses 2024; 40:521-530. [PMID: 38535626 DOI: 10.1089/aid.2023.0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
HIV/AIDS cannot be cured because of the persistence of the viral reservoir. Because of the complexity of the cellular composition and structure of the human organs, HIV reservoirs of anatomical site are also complex. Recently, although a variety of molecules have been reported to be involved in the establishment and maintenance of the viral reservoirs, or as marker of latent cells, the research mainly focuses on blood and lymph nodes. Now, the characteristics of the viral reservoir in tissue are not yet fully understood. In this study, various tissues were collected from SIVmac239-infected monkeys, and the level of total SIV DNA, SIV 2-LTR DNA, and cell-associated virus RNA in them were compared with character of the anatomical viral reservoir under early treatment. The results showed that short-term combination antiretroviral therapy (cART) starting from 3 days after infection could significantly inhibit viremia and reduce the size of the anatomical viral reservoir, but it could not eradicate de novo infections and ongoing replication of virus. Moreover, the effects of early cART on the level of total SIV DNA, SIV 2-LTR DNA, and cell-associated virus RNA in different tissues were different, which changed the size distribution of viral reservoir in anatomical site. Finally, the contribution of nonlymphoid tissues, especially liver and lung, to the viral reservoir increased after treatment, while the contribution of intestinal lymphoid to the viral reservoir significantly reduced. These results suggested that early treatment effectively decreased the size of viral reservoir, and that the effects of cART on the tissue viral reservoir varied greatly by tissue type. The results implied that persistent existence of virus in nonlymphoid tissues after short-term treatment suggested that the role of nonlymphoid tissues cannot be ignored in development strategies for AIDS therapy.
Collapse
Affiliation(s)
- Ren-Rong Tian
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ting Li
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ming-Xu Zhang
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Tian-Zhang Song
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Hong-Yi Zheng
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yong-Tang Zheng
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming National High-Level Biosafety Research Center for Nonhuman Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
18
|
Lara-Aguilar V, Llamas-Adán M, Brochado-Kith Ó, Crespo-Bermejo C, Grande-García S, Arca-Lafuente S, de Los Santos I, Prado C, Alía M, Sainz-Pinós C, Fernández-Rodríguez A, Martín-Carbonero L, Madrid R, Briz V. Low-level HIV-1 viremia affects T-cell activation and senescence in long-term treated adults in the INSTI era. J Biomed Sci 2024; 31:80. [PMID: 39160510 PMCID: PMC11334306 DOI: 10.1186/s12929-024-01064-z] [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: 03/27/2024] [Accepted: 07/12/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Around 10% of people with HIV (PWH) exhibit a low-level viremia (LLV) under antiretroviral therapy (ART). However, its origin and clinical significance are largely unknown, particularly at viremias between 50 and 200 copies/mL and under modern ART based on integrase strand transfer inhibitors (INSTIs). Our aim was to characterize their poor immune response against HIV in comparison to individuals with suppressed viremia (SV) and non-HIV controls (NHC). METHODS Transversal observational study in 81 matched participants: 27 PWH with LLV, 27 PWH with SV, and 27 NHC. Activation (CD25, HLA-DR, and CD38) and senescence [CD57, PD1, and HAVCR2 (TIM3)] were characterized in peripheral T-cell subsets by spectral flow cytometry. 45 soluble biomarkers of systemic inflammation were evaluated by immunoassays. Differences in cell frequencies and plasma biomarkers among groups were evaluated by a generalized additive model for location, scale, and shape (GAMLSS) and generalized linear model (GLM) respectively, adjusted by age, sex at birth, and ART regimen. RESULTS The median age was 53 years and 77.8% were male. Compared to NHC, PWH showed a lower CD4+/CD8+ ratio and increased activation, senescence, and inflammation, highlighting IL-13 in LLV. In addition, LLV showed a downtrend in the frequency of CD8+ naive and effector memory (EM) type 1 compared to SV, along with higher activation and senescence in CD4+ and CD8+ EM and terminally differentiated effector memory RA+ (TEMRA) subpopulations. No significant differences in systemic inflammation were observed between PWH groups. CONCLUSION LLV between 50 and 200 copies/mL leads to reduced cytotoxic activity and T-cell dysfunction that could affect cytokine production, being unable to control and eliminate infected cells. The increase in senescence markers suggests a progressive loss of immunological memory and a reduction in the proliferative capacity of immune cells. This accelerated immune aging could lead to an increased risk of developing future comorbidities. These findings strongly advocate for heightened surveillance of these PWH to promptly identify potential future complications.
Collapse
Affiliation(s)
| | - Manuel Llamas-Adán
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - Óscar Brochado-Kith
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Institute of Health Carlos III, Madrid, Spain
| | | | | | - Sonia Arca-Lafuente
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - Ignacio de Los Santos
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Institute of Health Carlos III, Madrid, Spain
- La Princesa University Hospital, Madrid, Spain
| | - Carmen Prado
- Flow Cytometry Unit, Institute of Health Carlos III, Madrid, Spain
| | - Mario Alía
- Flow Cytometry Unit, Institute of Health Carlos III, Madrid, Spain
| | - Coral Sainz-Pinós
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - Amanda Fernández-Rodríguez
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Institute of Health Carlos III, Madrid, Spain
| | - Luz Martín-Carbonero
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Institute of Health Carlos III, Madrid, Spain
- La Paz University Hospital (IdiPAZ), Madrid, Spain
| | | | - Verónica Briz
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
19
|
Lu X, Wang Y, Ma L, Liu M, Li Y, An N, Zhang X, Tang X, Li Q. Epidemic trend, genetic characteristics, and transmission networks of HIV-1 among treatment-naive men who have sex with men in Hebei province, China. Front Microbiol 2024; 15:1405565. [PMID: 39176274 PMCID: PMC11339959 DOI: 10.3389/fmicb.2024.1405565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction Homosexual transmission has contributed greatly to the current HIV-1 epidemic in Hebei province, China. Dolutegravir (DTG) will be conditionally used as a component of free antiretroviral therapy (ART) according to manual for national free anti-AIDS treatment drugs (2023 edition) issued by China in June 2023. However, current genetic characteristics and pretreatment drug resistance (PDR) to proteinase inhibitors (PIs), reverse transcriptase inhibitors (RTs) and integrase strand transfer inhibitors (INSTIs) of HIV-1 in this population have remained unclear. Methods Serial consecutive cross-sectional analyses for HIV- 1 infection trend, genetic characteristics, PDR and molecular transmission networks were conducted from 2018 to 2022. All of participants were HIV-1- infected MSM newly diagnosed at the HIV surveillance points (HSPs) in Hebei, China. Evidence of PDR was confirmed using the world health organization (WHO) list for surveillance of drug resistance mutations. Results In this study, a total of 14 HIV-1 subtypes were circulating in the HSPs of Hebei province, China. CRF01_ AE (51.9%, 350/675), CRF07_BC (30.4%, 205/675), B (6.2%, 42/675) and URFs (5.8%, 39/675) were the four most predominant subtypes among MSM. And, CRF07_BC (r > 0) and URFs (r > 0) indicated an increasing trend, respectively; however, CRF01_AE (r < 0) showed a decline trend. The overall prevalence of HIV-1 PDR showed a substantial increase from 6.3% in 2018 to 7.9% in 2022. The prevalence of NNRTI-PDR was the highest (5.8%, 39/675), followed by INSTIs (2.4%, 16/675), NRTIs (0.6%, 4/675) and PIs (0.3%, 2/675). Furthermore, extensive HIV-1 strains bearing PDR were circulating in the MSM population via molecular transmission networks for major HIV-1 subtypes, especially CRF01_AE and CRF07_BC. Discussion Our findings reflect that HIV-1 epidemic in the MSM population is complex and severe in Hebei, China. Therefore, it is urgent for us to implement more effective intervention measures to limit the further dissemination of HIV-1, especially the spread of HIV-1 INSTI-PDR strains.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qi Li
- Department of AIDS Research, Hebei Key Laboratory of Pathogen and Epidemiology of Infectious Disease, Hebei Provincial Center for Disease Control and Prevention, Shijiazhuang, Hebei, China
| |
Collapse
|
20
|
Dinesha TR, Boobalan J, Kumar CV, Manikandan P, Muhila M, Solomon SS, Srikrishnan AK, Murugavel KG. HIV-1 low-level viraemia predicts virological failure in first-line and second-line ART-experienced individuals in India: A retrospective longitudinal study. HIV Med 2024; 25:852-861. [PMID: 38663865 DOI: 10.1111/hiv.13641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/16/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE To study the prevalence of low-level viraemia (LLV) and its association with virological failure (VF). METHODS We conducted a retrospective analysis of 3498 participants at YRG CARE, Chennai, India (2013-2018) on antiretroviral therapy (ART) for ≥6 months with two or more plasma viral load (pVL) measurements. Results were stratified for those with pVL <1000 copies/mL: fully suppressed (FS) (pVL <40), low-LLV (pVL 40-199), mid-LLV (pVL 200-399), and high-LLV (pVL 400-999). The study assessed the association with VF (pVL >1000 copies/mL) using Cox proportional hazard model. RESULTS Among 3498 participants, 2965 (84.8%) were FS and 533 (15.2%) were LLV. During the follow-up, 348 (10%) experienced VF, with 222 (6.3%) experienced after LLV (42% of LLV) and 126 (3.6%) experienced after FS (4.3% of FS). When compared with FS, those with LLV had a greater risk of VF [adjusted hazard ratio (aHR) = 12.7; 95% confidence interval (CI): 10.2-15.9]. First-line participants had a higher VF incidence (aHR = 15.8, 95% CI: 11.4-21.9) than second-line participants (aHR = 5.6, 95% CI: 4.1-7.7). Those with high-LLV had the highest VF risk (aHR = 22.856, 95% CI: 15.204-34.359 vs. aHR = 8.186, 95% CI: 5.564-12.043, for first-line vs. second-line participants, respectively), followed by those with mid-LLV (aHR = 13.375, 95% CI: 8.327-21.483 vs. aHR = 6.261, 95% CI: 4.044-9.695) and low-LLV (aHR = 12.976, 95% CI: 7.974-21.118 vs. aHR = 4.158, 95% CI: 2.826-6.119). CONCLUSIONS The prevalence of LLV was intermediate in our study population. There was a higher risk of VF among individuals with LLV, and this risk increased with the increasing levels of LLV. Close monitoring of individuals experiencing LLV could help in the early identification of VF.
Collapse
Affiliation(s)
| | - Jayaseelan Boobalan
- Y.R. Gaitonde Centre for AIDS Research and Education (YRG CARE), Chennai, India
| | | | | | - Mohanarangan Muhila
- Y.R. Gaitonde Centre for AIDS Research and Education (YRG CARE), Chennai, India
| | | | | | | |
Collapse
|
21
|
Gounder L, Tomita A, Lessells R, Moodley S, Francois KL, Khan A, Pillay M, Manyana SC, Govender S, Govender K, Moodley P, Parboosing R, Msomi N, Tanser F, Naidoo K, Chimukangara B. Geospatial and temporal mapping of detectable HIV-1 viral loads amid dolutegravir rollout in KwaZulu-Natal, South Africa. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003224. [PMID: 38805477 PMCID: PMC11132473 DOI: 10.1371/journal.pgph.0003224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/20/2024] [Indexed: 05/30/2024]
Abstract
South Africa rolled out dolutegravir (DTG) as first-line antiretroviral therapy (ART) in December 2019 to overcome high rates of pretreatment non-nucleoside reverse transcriptase inhibitor drug resistance. In the context of transition to DTG-based ART, this study spatiotemporally analysed detectable HIV viral loads (VLs) prior to- and following DTG rollout in public-sector healthcare facilities in KwaZulu-Natal (KZN) province, the epicentre of the HIV epidemic in South Africa. We retrospectively curated a HIV VL database using de-identified routine VL data obtained from the National Health Laboratory Service for the period January 2018 to June 2022. We analysed trends in HIV viraemia and mapped median log10 HIV VLs per facility on inverse distance weighted interpolation maps. We used Getis-Ord Gi* hotspot analysis to identify geospatial HIV hotspots. We obtained 7,639,978 HIV VL records from 736 healthcare facilities across KZN, of which 1,031,171 (13.5%) had detectable VLs (i.e., VLs ≥400 copies/millilitre (mL)). Of those with detectable VLs, we observed an overall decrease in HIV VLs between 2018 and 2022 (median 4.093 log10 copies/mL; 95% confidence interval (CI) 4.087-4.100 to median 3.563 log10 copies/mL; CI 3.553-3.572), p<0.01 (median test). The downward trend in proportion of HIV VLs ≥1000 copies/mL over time was accompanied by an inverse upward trend in the proportion of HIV VLs between 400 and 999 copies/mL. Moreover, specific coastal and northern districts of KZN had persistently higher VLs, with emergent hotspots demonstrating spatial clustering of high median log10 HIV VLs. The overall decrease in HIV VLs over time shows good progress towards achieving UNAIDS 95-95-95 targets in KZN, South Africa. The DTG-transition has been associated with a reduction in VLs, however, there is a need for pre-emptive monitoring of low-level viraemia. Furthermore, our findings highlight that specific districts will need intensified HIV care despite DTG rollout.
Collapse
Affiliation(s)
- Lilishia Gounder
- Department of Virology, Inkosi Albert Luthuli Academic Complex, National Health Laboratory Service, Durban, South Africa
- Department of Virology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andrew Tomita
- Centre for Rural Health, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Richard Lessells
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Sandrini Moodley
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- South African Medical Research Council (SAMRC), CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Durban, South Africa
| | - Kerri-Lee Francois
- Department of Virology, Inkosi Albert Luthuli Academic Complex, National Health Laboratory Service, Durban, South Africa
- Department of Virology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Aabida Khan
- Department of Virology, Inkosi Albert Luthuli Academic Complex, National Health Laboratory Service, Durban, South Africa
- Department of Virology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Melendhran Pillay
- Department of Virology, Inkosi Albert Luthuli Academic Complex, National Health Laboratory Service, Durban, South Africa
- Department of Virology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sontaga C. Manyana
- Department of Virology, Inkosi Albert Luthuli Academic Complex, National Health Laboratory Service, Durban, South Africa
| | - Subitha Govender
- Department of Virology, Inkosi Albert Luthuli Academic Complex, National Health Laboratory Service, Durban, South Africa
| | - Kerusha Govender
- Department of Virology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Pravi Moodley
- Department of Virology, Inkosi Albert Luthuli Academic Complex, National Health Laboratory Service, Durban, South Africa
- Department of Virology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Raveen Parboosing
- Department of Virology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- School of Pathology, University of Witwatersrand & National Health Laboratory Service, Johannesburg, South Africa
| | - Nokukhanya Msomi
- Department of Virology, Inkosi Albert Luthuli Academic Complex, National Health Laboratory Service, Durban, South Africa
- Department of Virology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Frank Tanser
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Africa Health Research Institute, KwaZulu-Natal, Durban, South Africa
- School of Nursing and Public Health, University of KwaZulu-Natal, Durban, South Africa
- Centre for Epidemic Response and Innovation, School for Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
- DSI-NRF Centre of Excellence in Epidemiological Modelling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- South African Medical Research Council (SAMRC), CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Durban, South Africa
| | - Benjamin Chimukangara
- Department of Virology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- South African Medical Research Council (SAMRC), CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Durban, South Africa
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, Maryland, United States of America
| |
Collapse
|
22
|
Liu J, Li C, Sun Y, Fu C, Wei S, Zhang X, Ma J, Zhao Q, Huo Y. Characteristics of drug resistance mutations in ART-experienced HIV-1 patients with low-level viremia in Zhengzhou City, China. Sci Rep 2024; 14:10620. [PMID: 38724547 PMCID: PMC11082154 DOI: 10.1038/s41598-024-60965-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Although most people living with HIV (PLWH) receiving antiretroviral therapy (ART) achieve continuous viral suppression, some show detectable HIV RNA as low-level viremia (LLV) (50-999 copies/mL). Drug resistance mutations (DRMs) in PLWH with LLV is of particular concern as which may lead to treatment failure. In this study, we investigated the prevalence of LLV and LLV-associated DRMs in PLWH in Zhengzhou City, China. Of 3616 ART-experienced PLWH in a long-term follow-up cohort from Jan 2022 to Aug 2023, 120 were identified as having LLV. Of these PLWH with LLV, we obtained partial pol and integrase sequences from 104 (70 from HIV-1 RNA and 34 from proviral DNA) individuals. DRMs were identified in 44 individuals. Subtyping analysis indicated that the top three subtypes were B (48.08%, 50/104), CRF07_BC (31.73%, 33/104), and CRF01_AE (15.38%, 16/104). The proportions of nucleoside reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs), and integrase strand transfer inhibitors (INSTIs) associated DRMs were 23.83% (24/104), 35.58% (37/104), 5.77% (6/104), and 3.85% (4/104), respectively, which contributed to an overall prevalence of 42.31% (44/104). When analyzed by individual DRMs, the most common mutation(s) were V184 (18.27%, 19/104), followed by V179 (11.54%, 12/104), K103 (9.62%, 10/104), Y181 (9.62%, 10/104), M41 (7.69%, 8/104), and K65R (7.69%, 8/104). The prevalence of DRMs in ART-experienced PLWH with LLV is high in Zhengzhou City and continuous surveillance can facilitate early intervention and provision of effective treatment.
Collapse
Affiliation(s)
- Jinjin Liu
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou), No.29, Jingguang South Road, Erqi District, Zhengzhou, 450000, China
| | - Chaofeng Li
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou), No.29, Jingguang South Road, Erqi District, Zhengzhou, 450000, China
| | - Yan Sun
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou), No.29, Jingguang South Road, Erqi District, Zhengzhou, 450000, China
| | - Chaohong Fu
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou), No.29, Jingguang South Road, Erqi District, Zhengzhou, 450000, China
| | - Shuguang Wei
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou), No.29, Jingguang South Road, Erqi District, Zhengzhou, 450000, China
| | - Xiaohua Zhang
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou), No.29, Jingguang South Road, Erqi District, Zhengzhou, 450000, China
| | - Jie Ma
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou), No.29, Jingguang South Road, Erqi District, Zhengzhou, 450000, China
| | - Qingxia Zhao
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou), No.29, Jingguang South Road, Erqi District, Zhengzhou, 450000, China.
| | - Yuqi Huo
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou), No.29, Jingguang South Road, Erqi District, Zhengzhou, 450000, China.
| |
Collapse
|
23
|
Bareng OT, Moyo S, Mudanga M, Sebina K, Koofhethile CK, Choga WT, Moraka NO, Maruapula D, Gobe I, Motswaledi MS, Musonda R, Nkomo B, Ramaabya D, Chebani T, Makuruetsa P, Makhema J, Shapiro R, Lockman S, Gaseitsiwe S. Low-Level Viremia among Adults Living with HIV on Dolutegravir-Based First-Line Antiretroviral Therapy Is a Predictor of Virological Failure in Botswana. Viruses 2024; 16:720. [PMID: 38793602 PMCID: PMC11125697 DOI: 10.3390/v16050720] [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: 03/18/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
We evaluated subsequent virologic outcomes in individuals experiencing low-level virem ia (LLV) on dolutegravir (DTG)-based first-line antiretroviral therapy (ART) in Botswana. We used a national dataset from 50,742 adults who initiated on DTG-based first-line ART from June 2016-December 2022. Individuals with at least two viral load (VL) measurements post three months on DTG-based first-line ART were evaluated for first and subsequent episodes of LLV (VL:51-999 copies/mL). LLV was sub-categorized as low-LLV (51-200 copies/mL), medium-LLV (201-400 copies/mL) and high-LLV (401-999 copies/mL). The study outcome was virologic failure (VF) (VL ≥ 1000 copies/mL): virologic non-suppression defined as single-VF and confirmed-VF defined as two-consecutive VF measurements after an initial VL < 1000 copies/mL. Cox regression analysis identified predictive factors of subsequent VF. The prevalence of LLV was only statistically different at timepoints >6-12 (2.8%) and >12-24 (3.9%) (p-value < 0.01). LLV was strongly associated with both virologic non-suppression (adjusted hazards ratio [aHR] = 2.6; 95% CI: 2.2-3.3, p-value ≤ 0.001) and confirmed VF (aHR = 2.5; 95% CI: 2.4-2.7, p-value ≤ 0.001) compared to initially virally suppressed PLWH. High-LLV (HR = 3.3; 95% CI: 2.9-3.6) and persistent-LLV (HR = 6.6; 95% CI: 4.9-8.9) were associated with an increased hazard for virologic non-suppression than low-LLV and a single-LLV episode, respectively. In a national cohort of PLWH on DTG-based first-line ART, LLV > 400 copies/mL and persistent-LLV had a stronger association with VF. Frequent VL testing and adherence support are warranted for individuals with VL > 50 copies/mL.
Collapse
Affiliation(s)
- Ontlametse T. Bareng
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
- Department of Medical Sciences, Faculty of Allied Health Professions, University of Botswana, Gaborone 0022, Botswana (M.S.M.)
| | - Sikhulile Moyo
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Pathology, Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7935, South Africa
- School of Health Systems and Public Health, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Mbatshi Mudanga
- Department of Strategic Information, Botswana-University of Maryland School of Medicine Health Initiative, Gaborone 0022, Botswana
| | - Kagiso Sebina
- Department of Strategic Information, Botswana-University of Maryland School of Medicine Health Initiative, Gaborone 0022, Botswana
| | - Catherine K. Koofhethile
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Wonderful T. Choga
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
- Department of Medical Sciences, Faculty of Allied Health Professions, University of Botswana, Gaborone 0022, Botswana (M.S.M.)
| | - Natasha O. Moraka
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
- Department of Medical Sciences, Faculty of Allied Health Professions, University of Botswana, Gaborone 0022, Botswana (M.S.M.)
| | - Dorcas Maruapula
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
| | - Irene Gobe
- Department of Medical Sciences, Faculty of Allied Health Professions, University of Botswana, Gaborone 0022, Botswana (M.S.M.)
| | - Modisa S. Motswaledi
- Department of Medical Sciences, Faculty of Allied Health Professions, University of Botswana, Gaborone 0022, Botswana (M.S.M.)
| | - Rosemary Musonda
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
| | | | - Dinah Ramaabya
- Botswana Ministry of Health, Gaborone 0038, Botswana (T.C.)
| | - Tony Chebani
- Botswana Ministry of Health, Gaborone 0038, Botswana (T.C.)
| | | | - Joseph Makhema
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Roger Shapiro
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Shahin Lockman
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA 02115, USA
| | - Simani Gaseitsiwe
- Botswana Harvard Health Partnership, Gaborone 0000, Botswana (C.K.K.); (N.O.M.); (D.M.)
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
24
|
Shi J, Ying G, Zheng R, Zhang Z. Clinical significance and management of low-level HIV viremia in the era of integrase strand transfer inhibitors. HIV Med 2024; 25:361-369. [PMID: 37990782 DOI: 10.1111/hiv.13585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/05/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND People living with HIV (PLWH) and receiving antiretroviral therapy (ART) have a goal of achieving and maintaining viral suppression; however, the existence of PLWH that show events of low-level viremia (LLV) between 50 and 1000 copies/mL and with different virological consequences have been observed. Moreover, some reports indicate that LLV status can lead to residual immune activation and inflammation, leading to a higher occurrence of non-AIDS-defining events (nADEs) and other adverse clinical outcomes. Until now, however, published data have shown controversial results that hinder understanding of this phenomenon's actual cause(s) and origin(s). Integrase strand transfer inhibitors (INSTIs)-based therapies could lead to lower LLV over time and, therefore, more effective virological control. OBJECTIVES This review aims to assess recent findings to provide a view of the clinical significance and management of low-level HIV viremia in the era of INSTIs.
Collapse
Affiliation(s)
- Jinchuan Shi
- Affiliated Hangzhou Xixi Hospital Zhejiang University School of Medicine, Hangzhou, PR China
| | - Gaoxiang Ying
- Affiliated Hangzhou Xixi Hospital Zhejiang University School of Medicine, Hangzhou, PR China
| | - Rongrong Zheng
- Affiliated Hangzhou Xixi Hospital Zhejiang University School of Medicine, Hangzhou, PR China
| | - Zhongdong Zhang
- Affiliated Hangzhou Xixi Hospital Zhejiang University School of Medicine, Hangzhou, PR China
| |
Collapse
|
25
|
Lombardi F, Bruzzesi E, Bouba YR, Di Carlo D, Costabile V, Ranzenigo M, Maggiolo F, Castagna A, Callegaro AP, Zoncada A, Paolucci S, Micheli V, Renica S, Bezencheck A, Rossetti B, Santoro MM. Factors Associated with Low-Level Viremia in People Living with HIV in the Italian Antiviral Response Cohort Analysis Cohort: A Case-Control Study. AIDS Res Hum Retroviruses 2024; 40:80-89. [PMID: 37345697 DOI: 10.1089/aid.2023.0015] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2023] Open
Abstract
Despite effective antiretroviral therapies (ARTs), a subset of people living with HIV (PLWH) still experience low-level viremia (LLV, i.e., 50-1,000 copies/mL). The present study compared PLWH experiencing LLV with those maintaining virological suppression (VS) and explored the potential impact of preexisting drug resistance and other factors on LLV. We conducted a retrospective, 1:1 matched case-control study within a cohort of drug-experienced VS subjects from the Italian Antiviral Response Cohort Analysis database, followed in the period 2009-2019. Cases were individuals experiencing LLV, while controls were those who maintained VS. Matching was for calendar year of first ART regimen. Preexisting drug resistance was calculated as cumulative genotypic susceptibility score (GSS) according to regimen administered at the observational period start. To explore the effect of cumulative GSS, treated as a binary variable (≥2 and <2) and other factors on LLV, we performed a logistic regression analysis. Within a main population of 3,455 PLWH, 337 cases were selected. Cases were comparable to the controls for both gender and age. However, cases showed that they had experienced a longer time since HIV diagnosis, a higher number of drugs previously administered, lower baseline CD4+ T cell count and a higher zenith viral load (VL). By multivariate analysis, we found that higher zenith VL [adjusted odds ratio (aOR) (95% confidence interval [CI]) 1.30 (1.14-1.48)], a cumulative usage of both PI [aOR (95% CI): 2.03 (1.19-3.48)] and InSTI [aOR (95% CI): 2.23 (1.47-3.38)] and a cumulative GSS <2 [aOR (95% CI) 0.67 (0.46-0.98)], were associated with a higher risk in developing LLV. In current high-efficacy ART era, in drug-experienced PLWH, the predictors of increased risk of LLV were the presence of preexisting drug resistance, higher zenith VL, and previous PI, and InSTI exposure.
Collapse
Affiliation(s)
- Francesca Lombardi
- UOC Malattie Infettive, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elena Bruzzesi
- Infectious Diseases Unit, Università Vita-Salute San Raffaele, Milan, Italy
| | - Yagai Romeo Bouba
- Chantal BIYA International Reference Center for Research on HIV/AIDS Prevention and Management (CIRCB), Yaoundé, Cameroon
| | - Domenico Di Carlo
- Department of Biomedical and Clinical Sciences "L. Sacco", CRC Pediatric "Romeo and Enrica Invernizzi", University of Milan, Milan, Italy
| | - Valentino Costabile
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Martina Ranzenigo
- Infectious Diseases Unit, Università Vita-Salute San Raffaele, Milan, Italy
| | - Franco Maggiolo
- Department of Infectious Diseases, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Antonella Castagna
- Infectious Diseases Unit, Università Vita-Salute San Raffaele, Milan, Italy
- Infectious Diseases Unit, San Raffaele Scientific Institute, Milan, Italy
| | | | | | - Stefania Paolucci
- Molecular Virology Unit, Division of Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Valeria Micheli
- Laboratory of Clinical Microbiology, Virology, and Bioemergencies, ASST Fatebenefratelli Sacco, L. Sacco University Hospital, Milan, Italy
| | - Silvia Renica
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | | | - Barbara Rossetti
- Infectious Diseases Unit, AUSL Toscana Sud-Est, Ospedale Misericordia Grosseto, Siena, Italy
| | | |
Collapse
|
26
|
Nanyeenya N, Nakanjako D, Makumbi F, Nakigozi G, Nalugoda F, Kigozi G, Nasuuna E, Kibira SPS, Nabadda S, Kiyaga C, Huzaifah M, Kiwanuka N. Effectiveness of intensive adherence counselling in achieving an undetectable viral load among people on antiretroviral therapy with low-level viraemia in Uganda. HIV Med 2024; 25:245-253. [PMID: 37853605 PMCID: PMC11047222 DOI: 10.1111/hiv.13568] [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: 05/14/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023]
Abstract
INTRODUCTION Uganda was using a threshold of 1000 copies/mL to determine viral non-suppression for antiretroviral therapy monitoring among people living with HIV, prior to this study. It was not clear whether people living with HIV with low-level viraemia (LLV, ≥50 to <1000 copies/mL) would benefit from intensive adherence counselling (IAC). The purpose of this study was to determine the effectiveness of IAC among people living with HIV, receiving antiretroviral therapy, and with LLV in Uganda, to guide key policy decisions in HIV care, including the review of the viral load (VL) testing algorithm. METHODS This cluster-randomized clinical trial comprised adults from eight HIV clinics who were living with HIV, receiving ART, and had recent VL results indicating LLV (tested from July 2022 to October 2022). Participants in the intervention arm clinics received three once-monthly sessions of IAC, and those in the comparison non-intervention arm clinics received the standard of care. At the end of the study, all participants were re-tested for VL to determine the proportions of those who then had an undetectable VL (<50 copies/mL). We assessed the statistical association between cross-tabulated variables using Fisher's exact test and then modified Poisson regression. RESULTS A total of 136 participants were enrolled into the study at eight HIV clinics. All 68 participants in the intervention arm completed all IAC sessions. Only one participant in the non-intervention arm was lost to follow-up. The average follow-up time was 3.7 months (standard deviation [SD] 0.2) and 3.5 months (SD 0.1) in the intervention and non-intervention arms, respectively. In total, 59 (43.7%) of 135 people living with HIV achieved an undetectable VL during the study follow-up period. The effect of IAC on attaining an undetectable VL among people with LLV was nearly twice as high in the intervention arm (57.4%) than in the non-intervention arm (29.9%): adjusted risk ratio 1.9 (95% confidence interval 1.0-3.5), p = 0.037. CONCLUSION IAC doubled the likelihood of an undetectable VL among people living with HIV with LLV. Therefore, IAC has been instituted as an intervention to manage people living with HIV with LLV in Uganda, and this should also be adopted in other Sub-Saharan African countries with similar settings. CLINICALTRIALS GOV IDENTIFIER NCT05514418.
Collapse
Affiliation(s)
- Nicholus Nanyeenya
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
- Ministry of Health Central Public Health Laboratories, Kampala, Uganda
| | - Damalie Nakanjako
- Department of Medicine, School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Fredrick Makumbi
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | | | | | | | - Esther Nasuuna
- Infectious Diseases Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Simon P S Kibira
- Department of Community Health and Behavioral Sciences, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Susan Nabadda
- Ministry of Health Central Public Health Laboratories, Kampala, Uganda
| | - Charles Kiyaga
- Ministry of Health Central Public Health Laboratories, Kampala, Uganda
| | - Mutyaba Huzaifah
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Noah Kiwanuka
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| |
Collapse
|
27
|
Kohler M, Brown JA, Tschumi N, Lerotholi M, Motaboli L, Mokete M, Chammartin F, Labhardt ND. Clinical Relevance of Human Immunodeficiency Virus Low-level Viremia in the Dolutegravir era: Data From the Viral Load Cohort North-East Lesotho (VICONEL). Open Forum Infect Dis 2024; 11:ofae013. [PMID: 38390465 PMCID: PMC10883284 DOI: 10.1093/ofid/ofae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/05/2024] [Indexed: 02/24/2024] Open
Abstract
Background Human immunodeficiency virus low-level viremia (LLV) is associated with subsequent treatment failure at least with non nucleoside reverse transcriptase inhibitor (NNRTI)-containing antiretroviral therapy. Data on implications of LLV occurring under dolutegravir, which has largely replaced NNRTIs in Africa, are scarce, however. Methods We included adults with human immunodeficiency virus in Lesotho who had ≥2 viral loads (VLs) taken after ≥6 months of NNRTI- or dolutegravir-based antiretroviral therapy. Within VL pairs, we assessed the association of viral suppression (<50 copies/mL) and low- and high-range LLV (50-199 and 200-999 copies/mL, respectively) with virological failure (≥1000 copies/mL) using a mixed-effects regression model. Participants could contribute VLs to the NNRTI and the dolutegravir group. Results Among 18 550 participants, 12 216 (65.9%) were female and median age at first VL included was 41.2 years (interquartile range, 33.4-51.5). In both groups, compared with a suppressed VL, odds of subsequent virological failure were higher for low-range LLV (NNRTI: adjusted odds ratio; 95% confidence interval: 1.9; 1.4-2.4 and dolutegravir: 2.1; 1.3-3.6) and high-range LLV (adjusted odds ratio; 95% confidence interval, 4.2; 3.1-5.7 and 4.4; 2.4-7.9). Conclusions In the dolutegravir era, LLV remains associated with virological failure, endorsing the need for close clinical and laboratory monitoring of those with a VL ≥50 copies/mL.
Collapse
Affiliation(s)
- Maurus Kohler
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Jennifer A Brown
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Nadine Tschumi
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Malebanye Lerotholi
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
- Ministry of Health Lesotho, Maseru, Lesotho
| | | | | | - Frédérique Chammartin
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Niklaus D Labhardt
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
28
|
Stam AJ, Buchholtz NVEJ, Bierman WFW, van Crevel R, Hoepelman AIM, Claassen MAA, Ammerlaan HSM, van Welzen BJ, van Kasteren MEE, van Lelyveld SFL, de Jong D, Tesselaar K, van Luin M, Nijhuis M, Wensing AMJ, Team LOWERITS. Dynamics of Low-Level Viremia and Immune Activation after Switching to a Darunavir-Based Regimen. Viruses 2024; 16:182. [PMID: 38399959 PMCID: PMC10893305 DOI: 10.3390/v16020182] [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: 12/10/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/25/2024] Open
Abstract
There is an ongoing debate regarding whether low-level viremia (LLV), in particular persistent LLV, during HIV treatment with optimal adherence originates from low-level viral replication, viral production, or both. We performed an observational study in 30 individuals with LLV who switched to a boosted darunavir (DRV)-based therapy. In-depth virological analyses were used to characterize the viral population and the (activity) of the viral reservoir. Immune activation was examined using cell-bound and soluble markers. The primary outcome was defined as the effect on HIV-RNA and was categorized by responders (<50 cp/mL) or non-responders (>50 cp/mL). At week 24, 53% of the individuals were considered responders, 40% non-responders, and 7% could not be assigned. Sequencing showed no evolution or selection of drug resistance in the non-responders. Production of defective virus with mutations in either the protease (D25N) or RT active site contributed to persistent LLV in two individuals. We show that in about half of the study participants, the switch to a DRV-based regimen resulted in a viral response indicative of ongoing low-level viral replication as the cause of LLV before the switch. Our data confirm that in clinical management, high genetic barrier drugs like DRV are a safe choice, irrespective of the source of LLV.
Collapse
Affiliation(s)
- Arjen J. Stam
- Translational Virology Research Group, Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Infectious Diseases, Public Health Service Amsterdam, 1018 WT Amsterdam, The Netherlands
| | - Ninée V. E. J. Buchholtz
- Translational Virology Research Group, Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Wouter F. W. Bierman
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Andy I. M. Hoepelman
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Mark A. A. Claassen
- Department of Internal Medicine, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| | - Heidi S. M. Ammerlaan
- Department of Internal Medicine, Catharina Hospital, 5623 EJ Eindhoven, The Netherlands
| | - Berend J. van Welzen
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | | | | | - Dorien de Jong
- Translational Virology Research Group, Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Kiki Tesselaar
- Department of Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Matthijs van Luin
- Department of Clinical Pharmacy, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Monique Nijhuis
- Translational Virology Research Group, Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Annemarie M. J. Wensing
- Translational Virology Research Group, Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | | |
Collapse
|
29
|
Onwah O, Nwanja E, Akpan U, Toyo O, Nwangeneh C, Oyawola B, Idemudia A, Olatunbosun K, Igboelina O, Ogundehin D, James E, Onyedinachi O, Adegboye A, Eyo A. Prevalence and predictors of persistent low-level HIV viraemia: a retrospective cohort study among people receiving dolutegravir-based antiretroviral therapy in Southern Nigeria. Ther Adv Infect Dis 2024; 11:20499361241242240. [PMID: 38572299 PMCID: PMC10989043 DOI: 10.1177/20499361241242240] [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: 09/14/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
Background Persistent low-level viraemia (PLLV) is a risk factor for virologic failure among people receiving antiretroviral therapy (ART). Objectives We assessed the prevalence and predictors of PLLV among individuals receiving Dolutegravir-based ART in southern Nigeria. Design This retrospective cohort study used routine program data from electronic medical records of persons receiving Dolutegravir-based first-line ART in 154 PEPFAR/USAID-supported health facilities in Akwa Ibom and Cross Rivers states, Nigeria. Methods Clients on first-line Dolutegravir-based ART ⩾6 months, who had a viral load result in the 12 months preceding October 2021 (baseline), and a second viral load result by September 2022 were included. Persons with low-level viraemia (LLV) (viral load 51-999 copies/ml) received additional adherence support. The outcome analysed was PLLV (two consecutive LLV results). Indices were summarized using descriptive statistics, and predictors of PLLV were determined using multivariate logistic regression. Results In total, 141,208 persons on ART were included, of which 63.3% (n = 89,944) were females. The median age was 36 [29-44] years, median ART duration was 19 [11-42] months. At the end of the study, 10.5% (14,759/141,208) had initial LLV, 90.1% (13,304/14,759) of which attained undetectable viral load (⩽50 copies/ml), and 1.1% (163/14,759) transitioned to virologic failure (⩾1000 copies/ml) by the end of the study. PLLV prevalence was 0.9% (1292/141,208). Increasing ART duration [adjusted odds ratio (aOR) = 1.0; 95% confidence interval (CI): 1.005-1.008; p < 0.001] and viral suppression (<1000 copies/ml) before initial LLV (aOR = 1.7; 95% CI: 1.50-2.00; p < 0.001) were positively associated with PLLV, while receipt of tuberculosis preventive therapy reduced the likelihood of PLLV (aOR = 0.3; 95% CI: 0.10-0.94; p = 0.039). Conclusion PLLV was uncommon among individuals receiving dolutegravir-based ART and was associated with longer ART duration, prior viral suppression, and non-receipt of tuberculosis preventive therapy. This strengthens recommendations for continuous adherence support and comprehensive health services with ART, to prevent treatment failure.
Collapse
Affiliation(s)
- Ogheneuzuazo Onwah
- Excellence Community Education Welfare Scheme, 14 Ubium Street, Ewet Housing Estate, Uyo, Akwa Ibom, Nigeria
| | - Esther Nwanja
- Excellence Community Education Welfare Scheme, Uyo, Nigeria
| | - Uduak Akpan
- Excellence Community Education Welfare Scheme, Uyo, Nigeria
| | - Otoyo Toyo
- Excellence Community Education Welfare Scheme, Uyo, Nigeria
| | | | | | | | | | | | | | | | | | | | - Andy Eyo
- Excellence Community Education Welfare Scheme, Uyo, Nigeria
| |
Collapse
|
30
|
Han WM, Broom J, Bopage R, Templeton DJ, Edmiston N, Petoumenos K. Investigating rates and predictors of viral blips, low-level viraemia and virological failure in the Australian HIV observational database. Trop Med Int Health 2024; 29:42-56. [PMID: 38009461 PMCID: PMC11108647 DOI: 10.1111/tmi.13951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
OBJECTIVES Australia has made significant progress towards achieving the UNAIDS's 95-95-95 cascade targets including HIV viral suppression. To investigate the burden of HIV viraemia, we assessed viral blips, low-level viraemia (LLV) and virologic failure (VF) in an Australian cohort. METHODS We studied the proportion of people with viral suppression, viral blips, LLV and VF in the Australian HIV observational database (AHOD) between 2010 and 2021. The association between blips or LLV, and VF was investigated using Cox regression, and predictors of viral blips and LLV were assessed using repeated-measured logistic regression. RESULTS Among 2544 AHOD participants who were in follow-up and on antiretroviral therapy (ART) from 1 January 2010 (88.7% male), 444 had experienced VF (incidence rate: 2.45 [95% CI: 2.23-2.69] per 100 person-years [PY]) during 18,125 PY of follow-up (a median of 7.6 years). The proportion of people with VF decreased over time, whereas rates of blips and LLV remained stable. Participants with blips (hazard ratio, 2.89; 95% CI: 2.31-3.61) and LLV (4.46; 95% CI: 3.38-5.89) were at increased risk of VF. Hepatitis B co-infection, longer documented treatment interruption duration, younger age and lower CD4 at ART initiation, and protease inhibitors-based initial regimen were associated with an increased risk of VF. Common predictors of blips and LLV such as higher HIV-1 RNA and lower CD4 at ART initiation, longer treatment interruption, more VL testing and types of care settings (hospitals vs. sexual health services) were identified. CONCLUSIONS Blips and LLV predict subsequent VF development. We identified important predictors of HIV viraemia including VF among individuals on INSTI-based regimens to help direct HIV management plans.
Collapse
Affiliation(s)
- Win Min Han
- Kirby Institute, UNSW Sydney, Sydney, Australia
| | - Jennifer Broom
- Infectious Diseases Research Network, Sunshine Coast University Hospital, Australia
- Faculty of Medicine, The University of Queensland, Herston, Australia
| | - Rohan Bopage
- Western Sydney Sexual Health Centre and Westmead Clinical School, University of Sydney, Australia
| | - David J Templeton
- Kirby Institute, UNSW Sydney, Sydney, Australia
- Department of Sexual Health Medicine and Sexual Assault Medical Service, Sydney Local Health District, Camperdown, Australia
- Discipline of Medicine, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Natalie Edmiston
- School of Medicine, Western Sydney University, Sydney, Australia
| | | |
Collapse
|
31
|
Chenwi CA, Nayang Mundo RA, Nka AD, Semengue ENJ, Beloumou GA, Ka'e AC, Togna Pabo WL, Takou D, Abba A, Djupsa SC, Molimbou E, Etame NK, Kengni Ngueko AM, Same DK, Bouba Pamen JN, Abah Abah AS, Billong SC, Ajeh Awoh R, Halle-Ekane GE, Cappelli G, Njom-Nlend AE, Zk Bissek AC, Temfack E, Santoro MM, Ceccherini-Silberstein F, Colizzi V, Kaseya J, Ndembi N, Ndjolo A, Perno CF, Fokam J. Plasma Viral Load of 200 Copies/mL is a Suitable Threshold to Define Viral Suppression and HIV Drug Resistance Testing in Low- and Middle-Income Countries: Evidence From a Facility-Based Study in Cameroon. J Int Assoc Provid AIDS Care 2024; 23:23259582241306484. [PMID: 39711049 DOI: 10.1177/23259582241306484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
Abstract
INTRODUCTION In low-and-middle-income-countries (LMIC), viral suppression is defined as plasma viral load (PVL) below 1000 copies/mL (low-level viremia [LLV]) and threshold for HIV drug resistance (HIVDR) testing. However, there is evidence that drug resistance mutations (DRMs) may emerge at LLV, thus compromising antiretroviral treatment (ART) response. We evaluated sequencing success rates (SSR) at LLV, described HIVDR profiles and adequacy with potential efficacy of tenofovir-lamivudine-dolutegravir (TLD). METHODS A cross-sectional study was conducted among individuals with LLV at the Chantal BIYA International Reference Centre, Yaoundé, Cameroon from January 2020 through August 2021. HIV-1 sequencing was performed on protease/reverse-transcriptase, and sequences analysed using Stanford HIVdbv9.5. SSR and HIVDR rates were assessed according to viral-load ranges, with P < .05 considered statistically significant. RESULTS In total, 131 individuals were enrolled (median [IQR] age = 41 [30-49] years; 67.9% female; 54.7% at WHO clinical-stage I/II; median ART-duration 7 [4-11] years; median CD4-count 221 [103-402] cells/mm3 and median PVL 222 [96-436] copies/mL). Overall, SSR at LLV was 34.4% (45/131) and increased significantly with decreasing-age (P = .002) and increasing-PVL (P = .017). SSR were doubled at PVL≥150 copies/mL (21.8% at [40-150] vs. 43.3% at [150-1000]; OR = 2.8, P = .01). Of the 45 sequences obtained, 75.6% were recombinant strains (CRF02_AG, CRF09_cpx, CRF11_cpx) and 24.4% pure-subtypes (A1, D, F2, G). Overall, HIVDR prevalence at LLV was 82.2% (37/45), with 74.6% and 15.6% resistance to reverse-transcriptase inhibitors (RTIs) and ritonavir-boosted protease inhibitors (PI/r) respectively. Interestingly, HIVDR rates were similar at PVLs [50-200] versus [200-1000] copies/mL (P = .69). The most frequent DRMs were M184 V (73.3%) and K103N (40.0%) for RTIs and M46I (6.7%) for PIs/r. Overall 55.6% (25/45) of individuals were on suboptimal ART (needing ART-optimisation), with 48.9% (22/45) having suboptimal TLD predictive efficacy. Optimisation need was higher in first-line (81.8%, P = .03), but similar across viral clades and PVL-ranges (P = .6). CONCLUSION In this LMIC context, sequencing for HIVDR is feasible at LLV even with broad HIV-1 diversity, with significantly higher SSR above 150 copies/mL and/or in paediatrics. About 80% of individuals with LLV harbour HIVDR strains, with half of them needing ART optimisations to limit HIVDR emergence and prevent treatment failure. Our findings underscore the clinical benefits of HIVDR during persisting LLV and the need to reconsider the threshold for viral suppression around 200copies/mL in LMICs.
Collapse
Affiliation(s)
- Collins Ambe Chenwi
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- University of Rome Tor Vergata, Rome, Italy
| | - Rachel Audrey Nayang Mundo
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Protestant University of Central Africa, Yaounde, Cameroon
| | - Alex Durand Nka
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- University of Rome Tor Vergata, Rome, Italy
- Evangelic University of Cameroon, Bandjoun, Cameroon
| | - Ezechiel Ngoufack Jagni Semengue
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- University of Rome Tor Vergata, Rome, Italy
- Evangelic University of Cameroon, Bandjoun, Cameroon
| | - Grâce Angong Beloumou
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Aude Christelle Ka'e
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- University of Rome Tor Vergata, Rome, Italy
| | - Willy Leroi Togna Pabo
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Health Sciences, University of Buea, Buea, Cameroon
| | - Désiré Takou
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Aissatou Abba
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Sandrine Claire Djupsa
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Evariste Molimbou
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- University of Rome Tor Vergata, Rome, Italy
- Evangelic University of Cameroon, Bandjoun, Cameroon
| | - Naomi-Karell Etame
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Aurelie Minelle Kengni Ngueko
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- University of Rome Tor Vergata, Rome, Italy
| | - David Kob Same
- National HIV Drug Resistance Group, Ministry of Public Health, Yaoundé, Cameroon
- UNAIDS Country Office, Yaounde, Cameroon
| | - Jolle Nounouce Bouba Pamen
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- National HIV Drug Resistance Group, Ministry of Public Health, Yaoundé, Cameroon
- Department of Disease, Epidemic and Pandemic Control, Ministry of Public Health, Yaounde, Cameroon
| | | | - Serge Clotaire Billong
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- National HIV Drug Resistance Group, Ministry of Public Health, Yaoundé, Cameroon
| | - Rogers Ajeh Awoh
- Central Technical Group, National AIDS Control Committee, Ministry of Public Health, Yaounde, Cameroon
| | | | | | - Anne-Esther Njom-Nlend
- Higher Institute of Medical Technology, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Yaounde, Cameroon
| | - Anne-Cecile Zk Bissek
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- Division of Health Operational Research, Ministry of Public Health, Yaounde, Cameroon
| | - Elvis Temfack
- Africa Centre for Disease Control and Prevention (ACDC), Headquarter, Addis Ababa, Ethiopia
| | | | | | - Vittorio Colizzi
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Protestant University of Central Africa, Yaounde, Cameroon
- Evangelic University of Cameroon, Bandjoun, Cameroon
| | - Jean Kaseya
- Africa Centre for Disease Control and Prevention (ACDC), Headquarter, Addis Ababa, Ethiopia
| | - Nicaise Ndembi
- Africa Centre for Disease Control and Prevention (ACDC), Headquarter, Addis Ababa, Ethiopia
- Institute of Human Virology, Baltimore, MD, USA
| | - Alexis Ndjolo
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Carlo Federico Perno
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- University of Rome Tor Vergata, Rome, Italy
- Bambino Gesu Pediatric Hospital Rome Italy, Rome, Italy
| | - Joseph Fokam
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Health Sciences, University of Buea, Buea, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- National HIV Drug Resistance Group, Ministry of Public Health, Yaoundé, Cameroon
- Central Technical Group, National AIDS Control Committee, Ministry of Public Health, Yaounde, Cameroon
| |
Collapse
|
32
|
Zhou J, Wang FD, Li LQ, Li JY, Chen EQ. Decreased Efficacy of Sofosbuvir/Velpatasvir in HIV Patients Coinfected with HCV Genotype 3b. Future Virol 2024; 19:33-45. [DOI: 10.2217/fvl-2023-0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 01/29/2024] [Indexed: 01/13/2025]
Affiliation(s)
- Jing Zhou
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fa-Da Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lan-Qing Li
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing-Yu Li
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - En-Qiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
33
|
Djiyou ABD, Penda CI, Madec Y, Ngondi GD, Moukoko A, Eboumbou CE, Aghokeng AF. Prevalence of HIV drug resistance among adolescents receiving ART in Cameroon with low- or high-level viraemia. J Antimicrob Chemother 2023; 78:2938-2942. [PMID: 37921335 DOI: 10.1093/jac/dkad334] [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: 08/03/2023] [Accepted: 10/11/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVES To characterize HIV drug resistance (HIVDR) below and above the WHO threshold of 1000 copies/mL, considered for the definition of HIV ART failure in resource-limited settings. METHODS From a cohort of 280 adolescents (aged 10-19 years) receiving ART for at least 6 months, genotypic resistance testing (GRT) was attempted for two groups of participants: participants with low-level viraemia [LLV; viral load (VL) 200-999 copies/mL] and those in virological failure (VF; confirmed VL ≥1000 copies/mL) using an in-house method. The Stanford HIValg Program was used to identify relevant HIVDR mutations and predict the efficacy of the newly introduced tenofovir-lamivudine-dolutegravir combination. RESULTS GRT was successfully performed in 54/58 (93.1%) eligible participants, of which 28/31 (90.3%) were in VF and 26/27 (96.3%) had LLV. A high level of resistance was found both in adolescents with LLV and those in VF, with respectively 84.6% (22/26) and 75.0% (21/28) of participants harbouring at least one HIVDR mutation. NRTIs and NNRTIs were the most affected drug classes in both population groups. In contrast, PIs were not significantly affected and dolutegravir was expected to be active for all participants tested. However, for the newly introduced dolutegravir-based combination, functional monotherapy (dolutegravir only) was potentially possible for 22.7% (5/22) of the participants with LLV. CONCLUSIONS Our findings show that the 1000 copies/mL threshold is not an indicator of virological success and we call for a revision of the current WHO definition of VF in resource-limited countries.
Collapse
Affiliation(s)
- Armando B D Djiyou
- Department of Biological Sciences, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
- MIVEGEC, Université de Montpellier, CNRS, IRD, Montpellier, France
| | - Calixte I Penda
- Department of Clinical Sciences, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
- Department of Pediatrics and Child Health, General Hospital of Douala, Douala, Cameroon
| | - Yoann Madec
- Institut Pasteur, Université de Paris, Epidemiology of Emerging Diseases, F-75015 Paris, France
| | - Grace D Ngondi
- Department of Virology, Hôpital Laquintinie, Douala, Cameroon
| | - Astrid Moukoko
- Department of Virology, Hôpital Laquintinie, Douala, Cameroon
| | - Carole E Eboumbou
- Department of Biological Sciences, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
- Department of Parasitology, Centre Pasteur du Cameroun, Yaoundé, Cameroon
| | | |
Collapse
|
34
|
Asare K, Lewis L, van der Molen J, Sookrajh Y, Khubone T, Moodley P, Lessells RJ, Naidoo K, Sosibo P, Garrett N, Dorward J. Clinical Outcomes After Viremia Among People Receiving Dolutegravir vs Efavirenz-Based First-line Antiretroviral Therapy in South Africa. Open Forum Infect Dis 2023; 10:ofad583. [PMID: 38045558 PMCID: PMC10691652 DOI: 10.1093/ofid/ofad583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/15/2023] [Indexed: 12/05/2023] Open
Abstract
Background We aimed to compare clinical outcomes after viremia between dolutegravir vs efavirenz-based first-line antiretroviral therapy (ART) as evidence is lacking outside clinical trials in resource-limited settings. Methods We conducted a retrospective cohort analysis with routine data from 59 South African clinics. We included people with HIV aged ≥15 years receiving first-line tenofovir disoproxil fumarate, lamivudine, dolutegravir (TLD) or tenofovir disoproxil fumarate, emtricitabine, efavirenz (TEE) and with first viremia (≥50 copies/mL) between June and November 2020. We used multivariable modified Poisson regression models to compare retention in care and viral suppression (<50 copies/mL) after 12 months between participants on TLD vs TEE. Results At first viremia, among 9657 participants, 6457 (66.9%) were female, and the median age (interquartile range [IQR]) was 37 (31-44) years; 7598 (78.7%) were receiving TEE and 2059 (21.3%) TLD. Retention in care was slightly higher in the TLD group (84.9%) than TEE (80.8%; adjusted risk ratio [aRR], 1.03; 95% CI, 1.00-1.06). Of 6569 participants retained in care with a 12-month viral load, viral suppression was similar between the TLD (78.9%) and TEE (78.8%) groups (aRR, 1.02; 95% CI, 0.98-1.05). However, 3368 participants changed ART during follow-up: the majority from TEE to first-line TLD (89.1%) or second-line (TLD 3.4%, zidovudine/emtricitabine/lopinavir-ritonavir 2.1%). In a sensitivity analysis among the remaining 3980 participants who did not change ART during follow-up and had a 12-month viral load, viral suppression was higher in the TLD (78.9%) than TEE (74.9%) group (aRR, 1.07; 95% CI, 1.03-1.12). Conclusions Among people with viremia on first-line ART, dolutegravir was associated with slightly better retention in care and similar or better viral suppression than efavirenz.
Collapse
Affiliation(s)
- Kwabena Asare
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, KwaZulu-Natal, South Africa
- Discipline of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Lara Lewis
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, KwaZulu-Natal, South Africa
| | - Johan van der Molen
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, KwaZulu-Natal, South Africa
| | - Yukteshwar Sookrajh
- eThekwini Municipality Health Unit, eThekwini Municipality, Durban, KwaZulu-Natal, South Africa
| | - Thokozani Khubone
- eThekwini Municipality Health Unit, eThekwini Municipality, Durban, KwaZulu-Natal, South Africa
| | - Pravikrishnen Moodley
- Department of Virology, University of KwaZulu-Natal and National Health Laboratory Service, Inkosi Albert Luthuli Central Hospital, Durban, KwaZulu-Natal, South Africa
| | - Richard J Lessells
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, KwaZulu-Natal, South Africa
- KwaZulu-Natal Research and Innovation Sequencing Platform (KRISP), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, KwaZulu-Natal, South Africa
- South African Medical Research Council (SAMRC)-CAPRISA-TB-HIV Pathogenesis and Treatment Research Unit, University of KwaZulu-Natal Nelson R Mandela School of Medicine, Durban, KwaZulu-Natal, South Africa
| | - Phelelani Sosibo
- eThekwini Municipality Health Unit, eThekwini Municipality, Durban, KwaZulu-Natal, South Africa
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, KwaZulu-Natal, South Africa
- Discipline of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Jienchi Dorward
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, KwaZulu-Natal, South Africa
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxfordshire, UK
| |
Collapse
|
35
|
Gan L, Xie X, Fu Y, Yang X, Ma S, Kong L, Song C, Song Y, Ren T, Long H. Bictegravir/Emtricitabine/Tenofovir Alafenamide Versus Dolutegravir Plus Lamivudine for Switch Therapy in Patients with HIV-1 Infection: A Real-World Cohort Study. Infect Dis Ther 2023; 12:2581-2593. [PMID: 37845566 PMCID: PMC10651567 DOI: 10.1007/s40121-023-00879-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/22/2023] [Indexed: 10/18/2023] Open
Abstract
INTRODUCTION Bictegravir/emtricitabine/tenofovir alafenamide (BIC/FTC/TAF) and dolutegravir plus lamivudine (DTG + 3TC) are well tolerated and effective in clinical trials. This study aimed to evaluate the safety and efficacy of these two schemes in a real-world setting and to obtain the first dataset for switching to BIC/FTC/TAF and DTG + 3TC in a Chinese population. METHODS This retrospective single-center cohort study in China included participants who switched to DTG + 3TC or BIC/FTC/TAF between January 2020 and February 2023. The main endpoint was the proportion of participants with HIV-1 RNA levels of ≥ 50 copies/mL. Safety, tolerance, and the incidence of low-level viremia (LLV) were evaluated. RESULTS A total of 525 participants were included, 454 of whom were included in the PP analysis. At week 48, the proportions of participants with HIV-1 RNA ≥ 50 copies/mL were 4.4% (10/225) for DTG + 3TC and 6.1% (14/229) for BIC/FTC/TAF; virological efficacy did not differ significantly between the two groups. Consistent results were obtained in an intent-to-treat (ITT) analysis. The incidences of LLV were 3.6% (7/193) and 4.9% (10/206), respectively. During the study, none of the participants stopped taking drugs because of a lack of efficacy or adverse reactions. CONCLUSIONS Both regimens are well tolerated and effective for switching HIV-1 infection therapy. However, the detection of genotypic drug resistance should be considered when baseline virological non-suppression is observed.
Collapse
Affiliation(s)
- Lin Gan
- Guiyang Public Health Clinical Center, 6 Daying Road, Yunyan District, Guiyang, 550001, China
| | - Xiaoxin Xie
- Guiyang Public Health Clinical Center, 6 Daying Road, Yunyan District, Guiyang, 550001, China
| | - Yanhua Fu
- Guiyang Public Health Clinical Center, 6 Daying Road, Yunyan District, Guiyang, 550001, China
| | - Xiaoyan Yang
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Shujing Ma
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Linghong Kong
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Chunli Song
- Guiyang Public Health Clinical Center, 6 Daying Road, Yunyan District, Guiyang, 550001, China
| | - Yebing Song
- Guiyang Public Health Clinical Center, 6 Daying Road, Yunyan District, Guiyang, 550001, China
| | - Tingting Ren
- Guiyang Public Health Clinical Center, 6 Daying Road, Yunyan District, Guiyang, 550001, China
| | - Hai Long
- Guiyang Public Health Clinical Center, 6 Daying Road, Yunyan District, Guiyang, 550001, China.
| |
Collapse
|
36
|
SeyedAlinaghi S, Afsahi AM, Moradi A, Parmoon Z, Habibi P, Mirzapour P, Dashti M, Ghasemzadeh A, Karimi E, Sanaati F, Hamedi Z, Molla A, Mehraeen E, Dadras O. Current ART, determinants for virologic failure and implications for HIV drug resistance: an umbrella review. AIDS Res Ther 2023; 20:74. [PMID: 37884997 PMCID: PMC10604802 DOI: 10.1186/s12981-023-00572-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
OBJECTIVE The purpose of this study is to investigate the incidence of determinants for virologic failure and to identify predisposing factors to enhance treatment efficacy. Tackling this global public health issue is the key to reducing the rate of virological failure and increasing the success of treatment for those living with HIV. METHODS This umbrella review delves into various aspects of current anti-retroviral therapy (ART) which is the primary treatment for human immunodeficiency virus (HIV) infection. Comprehensive searches were conducted in online databases including PubMed, Embase, Scopus, and Web of Science, up to May 26, 2023. Following the screening and selection of relevant articles, eligible articles were included in the data extraction. This study adhered to the PRISMA guideline to report the results and employed the NIH quality and bias risk assessment tool to ensure the quality of included studies. RESULTS In total, 40 review studies published from 2015 to 2023 were included. The bulk of these studies concurred on several major factors contributing to HIV drug resistance and virological failure. Key among these were medication adherence, baseline and therapeutic CD4 levels, the presence of co-infections, and the advanced clinical stage of the infection. CONCLUSION The resistance to HIV drugs and instances of determinants for virologic failure have a profound impact on the life quality of those infected with HIV. Primary contributors to this scenario include insufficient adherence to treatment, decreased CD4 T-cell count, elevated viral levels, and certain treatment regimens. Implementing appropriate interventions could address these issues. Sub-Saharan Africa exhibits elevated rates of determinants for virologic failure, attributed to the delay in HIV testing and diagnosis, and late initiation of antiretroviral therapy (ART). It is essential to undertake further research aimed at enhancing the detection of resistance in HIV patients and mitigating viral failure by addressing these underlying causes.
Collapse
Affiliation(s)
- SeyedAhmad SeyedAlinaghi
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Masoud Afsahi
- Department of Radiology, School of Medicine, University of California, San Diego (UCSD), San Diego, CA, USA
| | - Ali Moradi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zohal Parmoon
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Pedram Habibi
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Pegah Mirzapour
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Dashti
- Department of Radiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Ghasemzadeh
- Department of Radiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Karimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Foziye Sanaati
- School of Nursing and Allied Medical Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Zahra Hamedi
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ayoob Molla
- School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Esmaeil Mehraeen
- Department of Health Information Technology, Khalkhal University of Medical Sciences, Khalkhal, 5681761351, Iran.
| | - Omid Dadras
- Bergen Addiction Research, Department of Addiction Medicine, Haukland University Hospital, Bergen, Norway
| |
Collapse
|
37
|
Aoko A, Pals S, Ngugi T, Katiku E, Joseph R, Basiye F, Kimanga D, Kimani M, Masamaro K, Ngugi E, Musingila P, Nganga L, Ondondo R, Makory V, Ayugi R, Momanyi L, Mambo B, Bowen N, Okutoyi S, Chun HM. Retrospective longitudinal analysis of low-level viremia among HIV-1 infected adults on antiretroviral therapy in Kenya. EClinicalMedicine 2023; 63:102166. [PMID: 37649807 PMCID: PMC10462863 DOI: 10.1016/j.eclinm.2023.102166] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 09/01/2023] Open
Abstract
Background HIV low-level viremia (LLV) (51-999 copies/mL) can progress to treatment failure and increase potential for drug resistance. We analyzed retrospective longitudinal data from people living with HIV (PLHIV) on antiretroviral therapy (ART) in Kenya to understand LLV prevalence and virologic outcomes. Methods We calculated rates of virologic suppression (≤50 copies/mL), LLV (51-999 copies/mL), virologic non-suppression (≥1000 copies/mL), and virologic failure (≥2 consecutive virologic non-suppression results) among PLHIV aged 15 years and older who received at least 24 weeks of ART during 2015-2021. We analyzed risk for virologic non-suppression and virologic failure using time-dependent models (each viral load (VL) <1000 copies/mL used to predict the next VL). Findings Of 793,902 patients with at least one VL, 18.5% had LLV (51-199 cp/mL 11.1%; 200-399 cp/mL 4.0%; and 400-999 cp/mL 3.4%) and 9.2% had virologic non-suppression at initial result. Among all VLs performed, 26.4% were LLV. Among patients with initial LLV, 13.3% and 2.4% progressed to virologic non-suppression and virologic failure, respectively. Compared to virologic suppression (≤50 copies/mL), LLV was associated with increased risk of virologic non-suppression (adjusted relative risk [aRR] 2.43) and virologic failure (aRR 3.86). Risk of virologic failure increased with LLV range (aRR 2.17 with 51-199 copies/mL, aRR 3.98 with 200-399 copies/mL and aRR 7.99 with 400-999 copies/mL). Compared to patients who never received dolutegravir (DTG), patients who initiated DTG had lower risk of virologic non-suppression (aRR 0.60) and virologic failure (aRR 0.51); similarly, patients who transitioned to DTG had lower risk of virologic non-suppression (aRR 0.58) and virologic failure (aRR 0.35) for the same LLV range. Interpretation Approximately a quarter of patients experienced LLV and had increased risk of virologic non-suppression and failure. Lowering the threshold to define virologic suppression from <1000 to <50 copies/mL to allow for earlier interventions along with universal uptake of DTG may improve individual and program outcomes and progress towards achieving HIV epidemic control. Funding No specific funding was received for the analysis. HIV program support was provided by the President's Emergency Plan for AIDS Relief (PEPFAR) through the United States Centers for Disease Control and Prevention (CDC).
Collapse
Affiliation(s)
- Appolonia Aoko
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV&TB, Center for Global Health, Nairobi, Kenya
| | - Sherri Pals
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV/TB, Center for Global Health, Atlanta, Georgia, USA
| | | | - Elizabeth Katiku
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV&TB, Center for Global Health, Nairobi, Kenya
| | - Rachael Joseph
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV&TB, Center for Global Health, Nairobi, Kenya
| | - Frank Basiye
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV&TB, Center for Global Health, Nairobi, Kenya
| | - Davies Kimanga
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV&TB, Center for Global Health, Nairobi, Kenya
| | - Maureen Kimani
- Ministry of Health Kenya, Division of Community Health, Nairobi, Kenya
| | - Kenneth Masamaro
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV&TB, Center for Global Health, Nairobi, Kenya
| | - Evelyn Ngugi
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV&TB, Center for Global Health, Nairobi, Kenya
| | - Paul Musingila
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV&TB, Center for Global Health, Nairobi, Kenya
| | - Lucy Nganga
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV&TB, Center for Global Health, Nairobi, Kenya
| | - Raphael Ondondo
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV&TB, Center for Global Health, Nairobi, Kenya
| | - Valeria Makory
- Ministry of Health Kenya, National AIDS & STI Control Program, Nairobi, Kenya
| | - Rose Ayugi
- Ministry of Health Kenya, National AIDS & STI Control Program, Nairobi, Kenya
| | - Lazarus Momanyi
- Ministry of Health Kenya, National AIDS & STI Control Program, Nairobi, Kenya
| | - Barbara Mambo
- Ministry of Health Kenya, National AIDS & STI Control Program, Nairobi, Kenya
| | - Nancy Bowen
- Ministry of Health Kenya, National Public Health Laboratory, Nairobi, Kenya
| | | | - Helen M. Chun
- U.S. Centers for Disease Control and Prevention (CDC), Division of Global HIV/TB, Center for Global Health, Atlanta, Georgia, USA
| |
Collapse
|
38
|
Álvarez H, Mocroft A, Ryom L, Neesgaard B, Edwards S, Svedhem V, Günthard HF, Zangerle R, Smith C, Castagna A, d’Arminio Monforte A, Wit F, Stecher M, Lehman C, Mussini C, Fontas E, González E, Wasmuth JC, Sönnerborg A, De Wit S, Chkhartishvili N, Stephan C, Petoumenos K, Jaschinski N, Vannappagari V, Gallant J, Young L, Volny Anne A, Greenberg L, Martín-Iguacel R, Poveda E, Llibre JM. Plasma Human Immunodeficiency Virus 1 RNA and CD4+ T-Cell Counts Are Determinants of Virological Nonsuppression Outcomes With Initial Integrase Inhibitor-Based Regimens: A Prospective RESPOND Cohort Study. Clin Infect Dis 2023; 77:593-605. [PMID: 37052343 PMCID: PMC10893964 DOI: 10.1093/cid/ciad219] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND There are conflicting data regarding baseline determinants of virological nonsuppression outcomes in persons with human immunodeficiency virus (HIV) starting antiretroviral treatment (ART). We evaluated the impact of different baseline variables in the RESPOND cohort. METHODS We included treatment-naive participants aged ≥18 who initiated 3-drug ART, in 2014-2020. We assessed the odds of virological suppression (VS) at weeks 48 and 96 using logistic regression. Viral blips, low-level viremia (LLV), residual viremia (RV), and virological failure (VF) rates were assessed using Cox regression. RESULTS Of 4310 eligible participants, 72% started integrase strand transfer inhibitor (INSTI)-based regimens. At 48 and 96 weeks, 91.0% and 93.3% achieved VS, respectively. At 48 weeks, Kaplan-Meier estimates of rates were 9.6% for viral blips, 2.1% for LLV, 22.2% for RV, and 2.1% for VF. Baseline HIV-1 RNA levels >100 000 copies/mL and CD4+ T-cell counts ≤200/µL were negatively associated with VS at weeks 48 (adjusted odds ratio, 0.51 [95% confidence interval, .39-.68] and .40 [.27-.58], respectively) and 96 and with significantly higher rates of blips, LLV, and RV. CD4+ T-cell counts ≤200/µL were associated with higher risk of VF (adjusted hazard ratio, 3.12 [95% confidence interval, 2.02-4.83]). Results were consistent in those starting INSTIs versus other regimens and those starting dolutegravir versus other INSTIs. CONCLUSIONS Initial high HIV-1 RNA and low CD4+ T-cell counts are associated with lower rates of VS at 48 and 96 weeks and higher rates of viral blips, LLV, and RV. Low baseline CD4+ T-cell counts are associated with higher VF rates. These associations remain with INSTI-based and specifically with dolutegravir-based regimens. These findings suggest that the impact of these baseline determinants is independent of the ART regimen initiated.
Collapse
Affiliation(s)
- Hortensia Álvarez
- Department of Internal Medicine, Infectious Diseases Unit, Complexo Hospitalario Universitario de Ferrol, Ferrol, SERGAS-A Coruña, Spain
- Department of Biochemistry, Genetics and Immunology, Universidade de Vigo, Vigo, Spain
| | - Amanda Mocroft
- CHIP, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation, Institute for Global Health, University College London, London, United Kingdom
| | - Lene Ryom
- CHIP, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Hvidovre University Hospital, Copenhagen, Denmark
| | | | - Simon Edwards
- Department of HIV, Mortimer Market Centre, London, United Kingdom
| | - Veronica Svedhem
- Department of Medicine, Medical Unit Infectious Diseases, Karolinska University Hospital, Karolinska Institutet, Huddinge, Sweden
| | - Huldrych F Günthard
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich and Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Robert Zangerle
- Austrian HIV Cohort Study, Medizinische Universität Innsbruck, Innsbruck, Austria
| | - Colette Smith
- The Royal Free HIV Cohort Study, Royal Free Hospital, University College London, London, United Kingdom
| | - Antonella Castagna
- San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milano, Italy
| | | | - Ferdinand Wit
- AIDS Therapy Evaluation in the Netherlands (ATHENA) cohort, HIV Monitoring Foundation, Amsterdam, The Netherlands
| | - Melanie Stecher
- Division of Infectious Diseases, Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Clara Lehman
- Division of Infectious Diseases, Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Cristina Mussini
- Modena HIV Cohort, Università degli Studi di Modena, Modena, Italy
| | - Eric Fontas
- Nice HIV Cohort, Université Côte d´Azur et Centre Hospitalier Universitaire, Nice, France
| | - Eva González
- PISCIS Cohort Study, Centre Estudis Epidemologics de ITS i VIH de Catalunya, Badalona, Spain
| | | | - Anders Sönnerborg
- Swedish InfCare HIV Cohort, Karolinska University Hospital, Stockholm, Sweden
| | - Stéphane De Wit
- CHU Saint-Pierre, Université Libre de Bruxelles, Brussels, Belgium
| | - Nikoloz Chkhartishvili
- Georgian National AIDS Health Information System, Infectious Diseases, AIDS and Clinical Immunology Research Center, Tbilisi, Georgia
| | - Christoph Stephan
- Frankfurt HIV Cohort Study, University Hospital Frankfurt, Goethe-University, Infectious Diseases Unit, Frankfurt, Germany
| | - Kathy Petoumenos
- The Kirby Institute, University of New South Wales, Sydney, Australia
| | | | | | | | | | | | - Lauren Greenberg
- CHIP, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation, Institute for Global Health, University College London, London, United Kingdom
| | | | - Eva Poveda
- Group of Virology and Pathogenesis, Galicia Sur Health Research Institute (IIS Galicia Sur)–Complexo Hospitalario Universitario de Vigo, Vigo, SERGAS-UVigo, Spain
| | - Josep M Llibre
- Infectious Diseases Division and Fight Infections Foundation, University Hospital Germans Trias i Pujol, Barcelona, Spain
| |
Collapse
|
39
|
Broyles LN, Luo R, Boeras D, Vojnov L. The risk of sexual transmission of HIV in individuals with low-level HIV viraemia: a systematic review. Lancet 2023; 402:464-471. [PMID: 37490935 PMCID: PMC10415671 DOI: 10.1016/s0140-6736(23)00877-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 07/27/2023]
Abstract
BACKGROUND The risk of sexual transmission of HIV from individuals with low-level HIV viraemia receiving antiretroviral therapy (ART) has important public health implications, especially in resource-limited settings that use alternatives to plasma-based viral load testing. This Article summarises the evidence related to sexual transmission of HIV at varying HIV viral load levels to inform messaging for people living with HIV, their partners, their health-care providers, and the wider public. METHODS We conducted a systematic review and searched PubMed, MEDLINE, Cochrane Central Register of Controlled Trials, Embase, Conference Proceedings Citation Index-Science, and WHO Global Index Medicus, for work published from Jan 1, 2010 to Nov 17, 2022. Studies were included if they pertained to sexual transmission between serodiscordant couples at various levels of viraemia, the science behind undetectable=untransmittable, or the public health impact of low-level viraemia. Studies were excluded if they did not specify viral load thresholds or a definition for low-level viraemia or did not provide quantitative viral load information for transmission outcomes. Reviews, non-research letters, commentaries, and editorials were excluded. Risk of bias was evaluated using the ROBINS-I framework. Data were extracted and summarised with a focus on HIV sexual transmission at varying HIV viral loads. FINDINGS 244 studies were identified and eight were included in the analysis, comprising 7762 serodiscordant couples across 25 countries. The certainty of evidence was moderate; the risk of bias was low. Three studies showed no HIV transmission when the partner living with HIV had a viral load less than 200 copies per mL. Across the remaining four prospective studies, there were 323 transmission events; none were in patients considered stably suppressed on ART. Among all studies there were two cases of transmission when the index patient's (ie, patient with previously diagnosed HIV infection) most recent viral load was less than 1000 copies per mL. However, interpretation of both cases was complicated by long intervals (ie, 50 days and 53 days) between the transmission date and the most recent index viral load result. INTERPRETATION There is almost zero risk of sexual transmission of HIV with viral loads of less than 1000 copies per mL. These data provide a powerful opportunity to destigmatise HIV and promote adherence to ART through dissemination of this positive public health message. These findings can also promote access to viral load testing in resource-limited settings for all people living with HIV by facilitating uptake of alternative sample types and technologies. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
| | - Robert Luo
- Global Health Impact Group, Atlanta, GA, USA
| | - Debi Boeras
- Global Health Impact Group, Atlanta, GA, USA
| | | |
Collapse
|
40
|
Mayasi Ngongo N, Kamangu Ntambwe E, Situakibanza Nani-Tuma H, Mbula Mambimbi M, Mandina Ndona M, Longokolo Mashi M, Bepouka Izizag B, Lukiana T, Odio Ossam J, Mangala Sonzi D, Maes N, Moutschen M, El Moussaoui M, Darcis G. Human Immunodeficiency Virus Viral Load Monitoring and Rate of Virologic Suppression Among Patients Receiving Antiretroviral Therapy in Democratic Republic of the Congo, 2013-2020. Open Forum Infect Dis 2023; 10:ofad242. [PMID: 37363052 PMCID: PMC10287095 DOI: 10.1093/ofid/ofad242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/02/2023] [Indexed: 06/28/2023] Open
Abstract
Background Antiretroviral therapy (ART) expansion and viral load as a treatment monitoring approach have increased the demand for viral load testing. Many hurdles affect the coverage, quality, and use of viral load results. Estimates of viral load monitoring and viral suppression rates are needed to assess the performance of ART programs and improve human immunodeficiency virus (HIV) management outcomes. Methods People with HIV (PWH) viral load monitoring data were routinely collected in 84 health facilities in Kinshasa, Democratic Republic of the Congo (DRC), between 2013 and 2020. The number of PWH under ART, the number of participants with at least 1 viral load test result, the rate of viral suppression (defined as ≤1000 HIV ribonucleic acid copies per mL), and the mean turnaround time from sample collection to release of viral load test results were collected together with clinical data. Results A total of 14 057 PWH were included in the analysis. People with HIV were mainly enrolled after the "test and treat" implementation. The patients were followed for a median period of 27 months. The proportion of PWH with at least 1 available viral load largely increased in recent years. The delay from sample collection to release of viral load test results decreased overtime, from 35 days in 2018 to 16 days in 2020. Pregnancy and advanced HIV disease were associated with a lower chance of viral suppression. Conclusions There has been considerable success in increasing viral load access for all PWH under therapy in DRC. Nevertheless, viral load testing should be intensified with a particular effort to be made in groups at higher risk of viral failure.
Collapse
Affiliation(s)
- Nadine Mayasi Ngongo
- Correspondence: Gilles Darcis, MD, PhD, Avenue de l’hôpital 1, 4000 Liège, Belgium (); Nadine Mayasi Ngongo, MD, PhD, Avenue de l’hôpital 1, 4000 Liège, Belgium (nadiamayas02@gmail)
| | - Erick Kamangu Ntambwe
- Department of Molecular Biology of Basic Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Hippolyte Situakibanza Nani-Tuma
- Department of Internal Medicine, Infectious and Tropical Diseases, University Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Marcel Mbula Mambimbi
- Department of Internal Medicine, Infectious and Tropical Diseases, University Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Madone Mandina Ndona
- Department of Internal Medicine, Infectious and Tropical Diseases, University Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Murielle Longokolo Mashi
- Department of Internal Medicine, Infectious and Tropical Diseases, University Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Ben Bepouka Izizag
- Department of Internal Medicine, Infectious and Tropical Diseases, University Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Tuna Lukiana
- Department of Internal Medicine, Infectious and Tropical Diseases, University Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Jérôme Odio Ossam
- Department of Internal Medicine, Infectious and Tropical Diseases, University Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Donatien Mangala Sonzi
- Department of Internal Medicine, Infectious and Tropical Diseases, University Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Nathalie Maes
- Biostatistics and Research Method Center (B-STAT), University Hospital of Liège, Liège, Belgium
| | - Michel Moutschen
- Department of Internal Medicine and Infectious Diseases, Liège University Hospital, Liège, Belgium
- AIDS Reference Laboratory, University of Liège, Liège, Belgium
| | - Majdouline El Moussaoui
- Department of Internal Medicine and Infectious Diseases, Liège University Hospital, Liège, Belgium
| | - Gilles Darcis
- Correspondence: Gilles Darcis, MD, PhD, Avenue de l’hôpital 1, 4000 Liège, Belgium (); Nadine Mayasi Ngongo, MD, PhD, Avenue de l’hôpital 1, 4000 Liège, Belgium (nadiamayas02@gmail)
| |
Collapse
|
41
|
Lane T, Makarov V, Nelson JAE, Meeker RB, Sanna G, Riabova O, Kazakova E, Monakhova N, Tsedilin A, Urbina F, Jones T, Suchy A, Ekins S. N-Phenyl-1-(phenylsulfonyl)-1 H-1,2,4-triazol-3-amine as a New Class of HIV-1 Non-nucleoside Reverse Transcriptase Inhibitor. J Med Chem 2023; 66:6193-6217. [PMID: 37130343 PMCID: PMC10269403 DOI: 10.1021/acs.jmedchem.2c02055] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Highly active antiretroviral therapy (HAART) has revolutionized human immunodeficiency virus (HIV) healthcare, turning it from a terminal to a potentially chronic disease, although some patients can develop severe comorbidities. These include neurological complications, such as HIV-associated neurocognitive disorders (HAND), which result in cognitive and/or motor function symptoms. We now describe the discovery, synthesis, and evaluation of a new class of N-phenyl-1-(phenylsulfonyl)-1H-1,2,4-triazol-3-amine HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTI) aimed at avoiding HAND. The most promising molecule, 12126065, exhibited antiviral activity against wild-type HIV-1 in TZM cells (EC50 = 0.24 nM) with low in vitro cytotoxicity (CC50 = 4.8 μM) as well as retained activity against clinically relevant HIV mutants. 12126065 also demonstrated no in vivo acute or subacute toxicity, good in vivo brain penetration, and minimal neurotoxicity in mouse neurons up to 10 μM, with a 50% toxicity concentration (TC50) of >100 μM, well below its EC50.
Collapse
Affiliation(s)
- Thomas Lane
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC 27606, USA
| | - Vadim Makarov
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow 119071, Russia
| | - Julie A. E. Nelson
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Rick B. Meeker
- Department of Neurology, University of North Carolina, NC 27514, USA
| | - Giuseppina Sanna
- Department of Biomedical Science, University of Cagliari, Monserrato, 09042, Italy
| | - Olga Riabova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow 119071, Russia
| | - Elena Kazakova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow 119071, Russia
| | - Natalia Monakhova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow 119071, Russia
| | - Andrey Tsedilin
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow 119071, Russia
| | - Fabio Urbina
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC 27606, USA
| | - Thane Jones
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC 27606, USA
| | - Ashley Suchy
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC 27606, USA
| |
Collapse
|
42
|
Nanyeenya N, Siu G, Kiwanuka N, Makumbi F, Nasuuna E, Nakanjako D, Nakigozi G, Nabadda S, Kiyaga C, Kibira SPS. Hopes, joys and fears: Meaning and perceptions of viral load testing and low-level viraemia among people on antiretroviral therapy in Uganda: A qualitative study. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001797. [PMID: 37163527 PMCID: PMC10171654 DOI: 10.1371/journal.pgph.0001797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/12/2023] [Indexed: 05/12/2023]
Abstract
Uganda applies the World Health Organization threshold of 1,000 copies/ml to determine HIV viral non-suppression. While there is an emerging concern of low-level viraemia (≥50 to <1,000 copies/ml), there is limited understanding of how people on antiretroviral therapy perceive viral load testing and low-level viremia in resource-limited settings. This qualitative study used the health belief model to explore the meaning that people living with HIV attach to viral load testing and low-level viraemia in Uganda. We used stratified purposive sampling to select people on antiretroviral therapy from eight high volume health facilities from the Central, Eastern, Northern and Western regions of Uganda. We used an interview guide, based on the health belief model, to conduct 32 in-depth interviews, which were audio-recorded and transcribed verbatim. Thematic analysis technique was used to analyze the data with the help of ATLAS.ti 6. The descriptions of viral load testing used by the participants nearly matched the medical meaning, and many people living with HIV understood what viral load testing was. Perceived benefits for viral load testing were the ability to show; the amount of HIV in the body, how the people living with HIV take their drugs, whether the drugs are working, and also guide the next treatments steps for the patients. Participants reported HIV stigma, lack of transport, lack of awareness for viral load testing, delayed and missing viral load results and few health workers as the main barriers to viral load testing. On the contrary, most participants did not know what low-level viraemia meant, while several perceived it as having a reduced viral load that is suppressed. Many people living with HIV are unaware about low-level viraemia, and hence do not understand its associated risks. Likewise, some people living with HIV are still not aware about viral load testing. Lack of transport, HIV stigma and delayed viral load results are major barriers to viral load testing. Hence, there is an imminent need to institute more strategies to create awareness about both low-level viraemia and viral load testing, manage HIV related stigma, and improve turnaround time for viral load results.
Collapse
Affiliation(s)
- Nicholus Nanyeenya
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
- Ministry of Health Central Public Health Laboratories, Kampala Uganda
| | - Godfrey Siu
- Child Health and Development Centre, School of Medicine Makerere University College of Health Sciences, Kampala, Uganda
| | - Noah Kiwanuka
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Fredrick Makumbi
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Esther Nasuuna
- Infectious Diseases Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Damalie Nakanjako
- Department of Medicine, School of Medicine Makerere University College of Health Sciences, Kampala, Uganda
| | | | - Susan Nabadda
- Ministry of Health Central Public Health Laboratories, Kampala Uganda
| | - Charles Kiyaga
- Ministry of Health Central Public Health Laboratories, Kampala Uganda
| | - Simon P. S. Kibira
- Department of Community Health and Behavioral Sciences, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| |
Collapse
|
43
|
Chen J, He Y, Zhong H, Hu F, Li Y, Zhang Y, Zhang X, Lin W, Li Q, Xu F, Chen S, Zhang H, Cai W, Li L. Transcriptome analysis of CD4+ T cells from HIV-infected individuals receiving ART with LLV revealed novel transcription factors regulating HIV-1 promoter activity. Virol Sin 2023:S1995-820X(23)00022-6. [PMID: 36907331 DOI: 10.1016/j.virs.2023.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Some HIV-infected individuals receiving ART develop low-level viremia (LLV), with a plasma viral load of 50-1000 copies/mL. Persistent low-level viremia is associated with subsequent virologic failure. The peripheral blood CD4+ T cell pool is a source of LLV. However, the intrinsic characteristics of CD4+ T cells in LLV which may contribute to low-level viremia are largely unknown. We analyzed the transcriptome profiling of peripheral blood CD4+ T cells from healthy controls (HC) and HIV-infected patients receiving ART with either virologic suppression (VS) or LLV. To identify pathways potentially responding to increasing viral loads from HC to VS and to LLV, KEGG pathways of differentially expressed genes (DEGs) were acquired by comparing VS with HC (VS-HC group) and LLV with VS (LLV-VS group). Characterization of DEGs in key overlapping pathways showed that CD4+ T cells in LLV expressed higher levels of Th1 signature transcription factors (TBX21), toll-like receptors (TLR-4, -6, -7 and -8), anti-HIV entry chemokines (CCL3 and CCL4), and anti-IL-1β factors (ILRN and IL1R2) compared to VS. Our results also indicated activation of the NF-κB and TNF signaling pathways that could promote HIV-1 transcription. Finally, we evaluated the effects of 4 and 17 transcription factors that were upregulated in the VS-HC and LLV-VS groups, respectively, on HIV-1 promoter activity. Functional studies revealed that CXXC5 significantly increased, while SOX5 markedly suppressed HIV-1 transcription. In summary, we found that CD4+ T cells in LLV displayed a distinct mRNA profiling compared to that in VS, which promoted HIV-1 replication and reactivation of viral latency and may eventually contribute to virologic failure in patients with persistent LLV. CXXC5 and SOX5 may serve as targets for the development of latency-reversing agents.
Collapse
Affiliation(s)
- Jingliang Chen
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Yaozu He
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Huolin Zhong
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Fengyu Hu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Yonghong Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Yeyang Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Xia Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Weiyin Lin
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Quanmin Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Feilong Xu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Shaozhen Chen
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Hui Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China; Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China.
| | - Weiping Cai
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China.
| | - Linghua Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China.
| |
Collapse
|
44
|
Yuan D, Li M, Zhou Y, Shi L, Lu J, Fu G, Wang B. Influencing factors and adverse outcomes of different virologic rebound states in antiretroviral treated HIV/AIDS patients. J Virus Erad 2023; 9:100320. [PMID: 37008574 PMCID: PMC10063406 DOI: 10.1016/j.jve.2023.100320] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/06/2023] Open
Abstract
Antiretroviral therapy (ART) aims to inhibit HIV replication, decrease CD4 T cell loss, and immune function recovery in order to reduce the morbidity and mortality associated with the infection. Treatment should also, improve quality of life and control HIV spread. However, incomplete viral suppression still occurs during ART. Viral suppression and virological failure (VF) thresholds vary between studies in terms of virological rebound (VR) states using different detection thresholds. Further understanding of influencing factors and adverse outcomes in various VR states should provide important guidance for HIV treatment.
Collapse
Affiliation(s)
- Defu Yuan
- Department of Epidemiology and Health Statistics, Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Mingma Li
- Department of Epidemiology and Health Statistics, Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Ying Zhou
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Lingen Shi
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jing Lu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Gengfeng Fu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
- Corresponding author. No.172, Jiangsu Road, Gulou District, Nanjing, China.
| | - Bei Wang
- Department of Epidemiology and Health Statistics, Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
- Corresponding author. No.87, Dingjiaqiao Road, Gulou District, Nanjing, China.
| |
Collapse
|
45
|
Sokhela S, Lalla-Edward S, Siedner MJ, Majam M, Venter WDF. Roadmap for Achieving Universal Antiretroviral Treatment. Annu Rev Pharmacol Toxicol 2023; 63:99-117. [PMID: 36662580 PMCID: PMC10807407 DOI: 10.1146/annurev-pharmtox-052020-094321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Modern antiretroviral therapy safely, potently, and durably suppresses human immunodeficiency virus (HIV) that, if left untreated, predictably causes acquired immunodeficiency syndrome (AIDS), which has been responsible for tens of millions of deaths globally since it was described in 1981. In one of the most extraordinary medical success stories in modern times, a combination of pioneering basic science, innovative drug development, and ambitious public health programming resulted in access to lifesaving, safe drugs, taken as an oral tablet daily, for most of the world. However, substantial challenges remain in the fields of prevention, timely access to diagnosis, and treatment, especially in pediatric and adolescent patients. As HIV-positive adults age, treating their comorbidities will require understanding the course of different chronic diseases complicated by HIV-related and antiretroviral toxicities and finding potential treatments. Finally, new long-acting antiretrovirals on the horizon promise exciting new options in both the prevention and treatment fields.
Collapse
Affiliation(s)
- Simiso Sokhela
- Ezintsha, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa;
| | - Samanta Lalla-Edward
- Ezintsha, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa;
| | - Mark J Siedner
- Harvard Medical School and Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mohammed Majam
- Ezintsha, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa;
| | | |
Collapse
|
46
|
van Heerden A, Szpiro A, Ntinga X, Celum C, van Rooyen H, Essack Z, Barnabas R. A Sequential Multiple Assignment Randomized Trial of scalable interventions for ART delivery in South Africa: the SMART ART study. Trials 2023; 24:32. [PMID: 36647092 PMCID: PMC9842495 DOI: 10.1186/s13063-022-07025-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/15/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Of the 8 million people in South Africa living with HIV, 74% of persons living with HIV are on antiretroviral therapy (ART) and 65% are virally suppressed. Detectable viral load results in HIV-associated morbidity and mortality and HIV transmission. Patient barriers to care, such as missed wages, transport costs, and long wait times for clinic visits and ART refills, are associated with detectable viral load. HIV differentiated service delivery (DSD) has simplified ART delivery for clients who achieve viral suppression and engage in care. However, DSD needs adaptation to serve clients who are not engaged in care. METHODS A Sequential Multiple Assignment Randomized Trial will be undertaken in KwaZulu-Natal, South Africa, to test adaptive ART delivery for persons with detectable viral load and/or who are not engaged in care. The types of differentiated service delivery (DSD) which will be examined in this study are clinic-based incentives, community-based smart lockers, and home delivery. The study plans to enroll up to 900 participants-people living with HIV, eligible for ART, and who are not engaged in care. The study aims to assess the proportion of ART-eligible persons living with HIV who achieve viral suppression at 18 months. The study will also evaluate the preferences of clients and providers for differentiated service delivery and evaluate the cost-effectiveness of adaptive HIV treatment for those who are not engaged in care. DISCUSSION To increase population-level viral suppression, persons with detectable viral load need responsive DSD interventions. A Sequential Multiple Assignment Randomized Trial (SMART) design facilitates the evaluation of a stepped, adaptive approach to achieving viral suppression with "right-sized" interventions for patients most in need of effective and efficient HIV care delivery strategies. TRIAL REGISTRATION ClinicalTrials.gov NCT05090150. Registered on October 22, 2021.
Collapse
Affiliation(s)
- Alastair van Heerden
- grid.417715.10000 0001 0071 1142Centre for Community Based Research, Human Sciences Research Council, Pietermaritzburg, South Africa ,grid.11951.3d0000 0004 1937 1135MRC/Wits Developmental Pathways for Health Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Adam Szpiro
- grid.34477.330000000122986657Department of Global Health, University of Washington, Seattle, WA USA ,grid.34477.330000000122986657Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA USA
| | - Xolani Ntinga
- grid.417715.10000 0001 0071 1142Centre for Community Based Research, Human Sciences Research Council, Pietermaritzburg, South Africa
| | - Connie Celum
- grid.34477.330000000122986657Department of Global Health, University of Washington, Seattle, WA USA ,grid.34477.330000000122986657Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA USA
| | - Heidi van Rooyen
- grid.11951.3d0000 0004 1937 1135MRC/Wits Developmental Pathways for Health Research Unit, University of the Witwatersrand, Johannesburg, South Africa ,grid.417715.10000 0001 0071 1142Human Sciences Research Council, Pietermaritzburg, South Africa
| | - Zaynab Essack
- grid.417715.10000 0001 0071 1142Centre for Community Based Research, Human Sciences Research Council, Pietermaritzburg, South Africa
| | - Ruanne Barnabas
- grid.32224.350000 0004 0386 9924Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| |
Collapse
|
47
|
Nanyeenya N, Chang LW, Kiwanuka N, Nasuuna E, Nakanjako D, Nakigozi G, Kibira SPS, Nabadda S, Kiyaga C, Makumbi F. The association between low-level viraemia and subsequent viral non-suppression among people living with HIV/AIDS on antiretroviral therapy in Uganda. PLoS One 2023; 18:e0279479. [PMID: 36638086 PMCID: PMC9838846 DOI: 10.1371/journal.pone.0279479] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/08/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Uganda's efforts to end the HIV epidemic by 2030 are threatened by the increasing number of PLHIV with low-level viraemia (LLV). We conducted a study to determine the prevalence of LLV and the association between LLV and subsequent viral non-suppression from 2016 to 2020 among PLHIV on ART in Uganda. METHOD This was a retrospective cohort study, using the national viral load (VL) program data from 2016 to 2020. LLV was defined as a VL result of at least 50 copies/ml, but less than 1,000 copies/ml. Multivariable logistic regression was used to determine the factors associated with LLV, and cox proportional hazards regression model was used to determine the association between LLV and viral non-suppression. RESULTS A cohort of 17,783 PLHIV, of which 1,466 PLHIV (8.2%) had LLV and 16,317 (91.8%) had a non-detectable VL was retrospectively followed from 2016 to 2020. There were increasing numbers of PLHIV with LLV from 2.0% in 2016 to 8.6% in 2020; and LLV was associated with male sex, second line ART regimen and being of lower age. 32.5% of the PLHIV with LLV (476 out of 1,466 PLHIV) became non-suppressed, as compared to 7.7% of the PLHIV (1,254 out of 16,317 PLHIV) with a non-detectable viral load who became non-suppressed during the follow-up period. PLHIV with LLV had 4.1 times the hazard rate of developing viral non-suppression, as compared to PLHIV with a non-detectable VL (adjusted hazard ratio was 4.1, 95% CI: 3.7 to 4.7, p < 0.001). CONCLUSION Our study indicated that PLHIV with LLV increased from 2.0% in 2016 to 8.6% in 2020, and PLHIV with LLV had 4.1 times the hazard rate of developing viral non-suppression, as compared to PLHIV with a non-detectable VL. Hence the need to review the VL testing algorithm and also manage LLV in Uganda.
Collapse
Affiliation(s)
- Nicholus Nanyeenya
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
- Department of National Health Laboratory Services, Ministry of Health, Kampala Uganda
| | - Larry William Chang
- Department of Epidemiology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Noah Kiwanuka
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Esther Nasuuna
- Infectious Diseases Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Damalie Nakanjako
- Department of Medicine, School of Medicine Makerere University College of Health Sciences, Kampala, Uganda
| | | | - Simon P. S. Kibira
- Department of Community Health and Behavioral Sciences, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Susan Nabadda
- Department of National Health Laboratory Services, Ministry of Health, Kampala Uganda
| | - Charles Kiyaga
- Department of National Health Laboratory Services, Ministry of Health, Kampala Uganda
| | - Fredrick Makumbi
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| |
Collapse
|
48
|
Yu H, Yang Y, Cao D, Zhao Y, Jin C, Sun H, Cao Y, Ye R, Yao S, Duan S, Wu Z. Association of low-level viremia with mortality among people living with HIV on antiretroviral therapy in Dehong, Southwest China: A retrospective cohort study. HIV Med 2023; 24:37-45. [PMID: 35578387 DOI: 10.1111/hiv.13320] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 04/12/2022] [Indexed: 01/19/2023]
Abstract
OBJECTIVES To investigate the association of low-level viremia (LLV) with mortality among people living with HIV (PLHIV) on antiretroviral therapy (ART) in Dehong, Southwest China. METHODS We analysed data collected from a cohort of PLHIV on ART in Dehong. PLHIV were enrolled in this cohort after they started ART, with viral load (VL) tested once a year afterwards. Each VL level was then categorized into one of the four groups: <50, 50-199, 200-999 and ≥1000 copies/ml. VL levels of 50-199 and 200-999 copies/ml were defined as LLV. The VL level for each participant was re-categorized and fitted into an extended Cox regression model as a time-varying covariate to examine the associations of VL level with all-cause and AIDS-related deaths. RESULTS Among the included 7273 of 8762 PLHIV in this study, median age (interquartile range, IQR) was 36 (30-43) years and 59.9% were male. The patients were followed up for a median duration (IQR) of 6.2 (4.3-8.2) years. Compared with VL <50 copies/ml, LLV 200-999 copies/ml (adjusted hazard ratio [aHR] and 95% confidence interval [95% CI]: 1.56 [1.04, 2.32]) were associated with elevated risk of all-cause mortality and LLV50-199 (aHR [95% CI]: 1.00 [0.68, 1.45]) were not. Similarly, only LLV200-999 copies/ml (aHR [95% CI]: 2.37 [1.36, 4.14]) corresponded to higher risk of AIDS-related mortality. CONCLUSIONS This study suggests that PLHIV on ART may have elevated death risks even though the viremia is suppressed at a low level. Interventions targeting PLHIV with LLV should be developed to reduce their mortality.
Collapse
Affiliation(s)
- Hailiang Yu
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yuecheng Yang
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, China
| | - Dongdong Cao
- Dehong Dai and Jingpo Autonomous Prefecture People's Hospital, China
| | - Yan Zhao
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Cong Jin
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haolan Sun
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.,School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Australia
| | - Yanfen Cao
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, China
| | - Runhua Ye
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, China
| | - Shitang Yao
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, China
| | - Song Duan
- Dehong Dai and Jingpo Autonomous Prefecture Center for Disease Control and Prevention, China
| | - Zunyou Wu
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.,Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| |
Collapse
|
49
|
Nzivo MM, Waruhiu CN, Kang'ethe JM, Budambula NLM. HIV Virologic Failure among Patients with Persistent Low-Level Viremia in Nairobi, Kenya: It Is Time to Review the >1000 Virologic Failure Threshold. BIOMED RESEARCH INTERNATIONAL 2023; 2023:8961372. [PMID: 37152588 PMCID: PMC10159743 DOI: 10.1155/2023/8961372] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/31/2023] [Accepted: 02/13/2023] [Indexed: 05/09/2023]
Abstract
Persistent low-level viremia (PLLV) of 200-999 copies/ml has been reported as a risk factor for HIV virologic failure (VF). This retrospective study was aimed at characterizing patients with PLLV, determining factors associated with VF, and determining the effect of regimen change. Data were extracted from electronic medical records for HIV care and treatment. Patients' characteristics (N = 705) were as follows: a mean age of 42 years, majority female (55%), and 51% married. A majority (78.7%) had a history of opportunistic infections in their ART lifetime. To determine factors associated with VF, 187 records on patients who maintained PLLV and 12 on deceased patients at the time of data review were eliminated from the analysis, leaving 506 patient records. Out of the 506, 89% (451/506) suppressed VL to nondetectable levels while 11% (55/506) had VF, and the difference was significant (P = 0.0001). Virologic failure was significantly associated with ages 10-30 years (P < 0.05). Baseline VL ≥ 1000 (OR 3.929; P = 0.002) and 200-999 copies/ml (OR 4.062; P = 0.004) were associated with VF. During PLLV, factors associated with VF included the following: PLLV of 200-999 copies/ml (P < 0.05), viral blips (OR 4.545; P = 0.0001), mean maximum VL (P < 0.05), and age (P = 0.043). Married marital status was inversely associated with VF (OR 0.318; P = 0.026). Regimen change was not significantly associated with virologic outcomes. However, patients who switched regimens to the second line had a high risk of VF (P = 0.028; OR 3.203). Regimen change was significantly high (P < 0.05) among adolescents and patients with a start regimen of 2NRTI+1NNRTI. Most of the PLLV patients (89%) achieved nondetectable VL after their continued ART monitoring for at least 12 months. Therefore, PLLV was not an indicator of VF. However, a consistent VL of ≥200-999 copies/ml at baseline and more than 12 months of ART care and treatment were significantly associated with VF. Patients with VL 200-999 copies/ml, adolescents, and young adults require intensive ART monitoring and support.
Collapse
Affiliation(s)
- Mirriam M. Nzivo
- School of Biological Sciences, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000-00200, Nairobi, Kenya
| | - Cecilia N. Waruhiu
- The Africa Genomics Centre and Consultancy Ltd., P.O. Box 381-00517, Nairobi, Kenya
| | - James M. Kang'ethe
- Comprehensive Care Centre, Kenyatta National Hospital, P.O. Box 20723-00202, Nairobi, Kenya
| | - Nancy L. M. Budambula
- Department of Biological Sciences, University of Embu, P.O. Box 6-60100, Embu, Kenya
| |
Collapse
|
50
|
Shigayeva A, Gcwensa N, Ndlovu CD, Ntumase N, Sabela S, Ohler L, Trivino-Duran L, Kamara EF, Hlophe K, Isaakidis P, Van Cutsem G. Retention on ART and viral suppression among patients in alternative models of differentiated HIV service delivery in KwaZulu-Natal, South Africa. PLOS GLOBAL PUBLIC HEALTH 2022; 2:e0000336. [PMID: 36962695 PMCID: PMC10021436 DOI: 10.1371/journal.pgph.0000336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 11/10/2022] [Indexed: 06/18/2023]
Abstract
Differentiated models of HIV care (DMOC) aim to improve health care efficiency. We describe outcomes of five DMOC in KwaZulu-Natal, South Africa: facility adherence clubs (facility AC) and community adherence clubs (community AC), community antiretroviral treatment (ART) groups (CAG), spaced fast lane appointments (SFLA), and community pick up points (PuP). This retrospective cohort study included 8241 eligible patients enrolled into DMOC between 1/1/2012 and 31/12/2018. We assessed retention in DMOC and on ART, and viral load suppression (<1000 copies/mL). Kaplan-Meier techniques were applied to describe crude retention. Mixed effects parametric survival models with Weibull distribution and clustering on health center and individual levels were used to assess predictors for ART and DMOC attrition, and VL rebound (≥1000 copies/mL). Overall DMOC retention was 85%, 80%, and 76% at 12, 24 and 36 months. ART retention at 12, 24 and 36 months was 96%, 93%, 90%. Overall incidence rate of VL rebound was 1.9 episodes per 100 person-years. VL rebound rate was 4.9 episodes per 100 person-years among those enrolled in 2012-2015, and 0.8 episodes per 100 person-years among those enrolled in 2016-2018 (RR 0.12; 95% CI, 0.09-0.15, p<0.001). Prevalence of confirmed virological failure was 0.6% (38/6113). Predictors of attrition from DMOC and from ART were male gender, younger age, shorter duration on ART before enrollment. Low level viremia (>200-399 copies/mL) was associated with higher hazards of VL rebound and attrition from ART. Concurrent implementation of several DMOC in a large ART program is feasible and can achieve sustained retention on ART and VL suppression.
Collapse
Affiliation(s)
| | - Ntombi Gcwensa
- Médecins Sans Frontières—South Africa, Eshowe, South Africa
| | | | | | | | - Liesbet Ohler
- Médecins Sans Frontières—South Africa, Eshowe, South Africa
| | | | | | | | - Petros Isaakidis
- Southern African Medical Unit, Médecins Sans Frontières, Cape Town, South Africa
- Clinical and Molecular Epidemiology Unit, Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Gilles Van Cutsem
- Southern African Medical Unit, Médecins Sans Frontières, Cape Town, South Africa
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town, South Africa
| |
Collapse
|