1
|
Mihealsick E, Word A, Scully EP. The impact of sex on HIV immunopathogenesis and therapeutic interventions. J Clin Invest 2024; 134:e180075. [PMID: 39286972 PMCID: PMC11405047 DOI: 10.1172/jci180075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Globally, the majority of people living with HIV are women or girls, but they have been a minority of participants in clinical trials and observational studies of HIV. Despite this underrepresentation, differences in the pathogenesis of HIV have been observed between men and women, with contributions from both gender- and sex-based factors. These include differences in the risk of HIV acquisition, in viral load set point and immune activation in responses to viremia, and differences in HIV reservoir maintenance. These differences obligate adequate study in both males and females in order to optimize treatments, but also provide a powerful leverage point for delineating the mechanisms of HIV pathogenesis. The shifts in exposure to sex steroid hormones across a lifespan introduce additional complexity, which again can be used to focus on either genetic or hormonal influences as the driver of an outcome. In this Review, we discuss consistent and reproducible differences by sex across the spectrum of HIV, from acquisition through pathogenesis, treatment, and cure, and explore potential mechanisms and gaps in knowledge.
Collapse
Affiliation(s)
| | | | - Eileen P Scully
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Tipoe T, Ogbe A, Lee M, Brown H, Robinson N, Hall R, Petersen C, Lewis H, Thornhill J, Ryan F, Fox J, Fidler S, Frater J. Impact of antiretroviral therapy during primary HIV infection on T-cell immunity after treatment interruption. Eur J Immunol 2024:e2451200. [PMID: 39138621 DOI: 10.1002/eji.202451200] [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: 04/16/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024]
Abstract
This study aims to understand the impact of early antiretroviral therapy (ART) on HIV-specific T-cell responses measured after treatment interruption may inform strategies to deliver ART-free immune-mediated viral suppression. HIV-specific T-cell immunity was analysed using gamma interferon enzyme-linked immunospot assays in two studies. SPARTAC included individuals with primary HIV infection randomised to 48 weeks of ART (n = 24) or no immediate therapy (n = 37). The PITCH (n = 7) cohort started antiretroviral therapy in primary infection for at least one year, followed by TI. In SPARTAC, participants treated in PHI for 48 weeks followed by TI for 12 weeks, and those who remained untreated for 60 weeks made similar HIV Gag-directed responses (both magnitude and breadth) at week 60. However, the treated group made a greater proportion of novel HIV Gag-directed responses by Week 60, suggestive of a greater reserve to produce new potentially protective responses. In the more intensively followed PITCH study, 6/7 participants showed dominant Gag and/or Pol-specific responses post-TI compared with pre-TI. Although early ART in PHI was not associated with major differences in HIV-specific immunity following TI compared with untreated participants, the potential to make more new Gag-directed responses warrants further investigation as this may inform strategies to achieve ART-free control.
Collapse
Affiliation(s)
- Timothy Tipoe
- Peter Medawar Building for Pathogen Research, Nuffield Dept of Medicine, University of Oxford, Oxford, UK
| | - Ane Ogbe
- Peter Medawar Building for Pathogen Research, Nuffield Dept of Medicine, University of Oxford, Oxford, UK
| | - Ming Lee
- Peter Medawar Building for Pathogen Research, Nuffield Dept of Medicine, University of Oxford, Oxford, UK
- Department of HIV Medicine, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Helen Brown
- Peter Medawar Building for Pathogen Research, Nuffield Dept of Medicine, University of Oxford, Oxford, UK
| | - Nicola Robinson
- Peter Medawar Building for Pathogen Research, Nuffield Dept of Medicine, University of Oxford, Oxford, UK
| | - Rebecca Hall
- Department of Infectious Disease, Imperial College London, London, UK
| | - Claire Petersen
- Department of Infectious Disease, Imperial College London, London, UK
| | - Heather Lewis
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Fiona Ryan
- Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Julie Fox
- Guy's and St Thomas' NHS Foundation Trust, London, UK
- NIHR Clinical Research Facility, Guys and St Thomas' NHS Trust, London, UK
| | - Sarah Fidler
- Department of HIV Medicine, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
- NIHR Imperial College Biomedical Research Centre, London, UK
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Dept of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
3
|
Grinsztejn E, Cardoso SW, Velasque L, Hoagland B, dos Santos DG, Coutinho C, Cruz Silva SDC, Nazer SC, Ferreira ACG, Castilho J, Grinsztejn B, Veloso VG. Impact of Latent M. tuberculosis Infection Treatment on Time to CD4/CD8 Recovery in Acute, Recent, and Chronic HIV Infection. J Acquir Immune Defic Syndr 2023; 94:355-363. [PMID: 37595204 PMCID: PMC10609716 DOI: 10.1097/qai.0000000000003284] [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: 03/16/2023] [Accepted: 08/01/2023] [Indexed: 08/20/2023]
Abstract
INTRODUCTION In people living with HIV, active and latent tuberculosis (TB) coinfections are associated with immune activation that correlate with HIV progression and mortality. We investigated the effect of initiating antiretroviral therapy (ART) during acute (AHI), recent (RHI), or chronic HIV infection (CHI) on CD4/CD8 ratio normalization and associated factors, the impact of latent TB infection treatment, and prior/concomitant TB diagnosis at the time of ART initiation. METHODS We included sex with men and transgender women individuals initiating ART with AHI, RHI and CHI between 2013 and 2019, from a prospective cohort in Brazil. We compared time from ART initiation to the first normal CD4/CD8 ratio (CD4/CD8 ≥1) using Kaplan-Meier curves and multivariable Cox proportional hazards models. Sociodemographic and clinical variables were explored. Variables with P -values <0.20 in univariable analyses were included in multivariable analyses. RESULTS Five hundred fifty participants were included, 11.8% classified as AHI and 6.4% as RHI, 46.7% with CHI-CD4 cell counts ≥350 cells/mm 3 and 35.1% with CHI-CD4 cell counts <350 cells/mm 3 . Time to normalization was shortest among AHI patients, followed by RHI and CHI individuals with higher baseline CD4. In the multivariable model, AHI was associated with a six-fold increased likelihood of achieving a CD4/CD8 ratio ≥1 (hazard ratio [HR]: 6.03; 95% confidence interval [CI]: 3.70 to 9.82; P < 0.001), RHI with HR: 4.47 (95% CI: 2.57 to 7.76; P < 0.001), and CHI CD4 ≥350 cells/mm 3 with HR: 1.87 (95% CI: 1.24 to 2.84; P = 0.003). Latent TB infection treatment was significantly associated with a higher likelihood of the outcome (HR: 1.79; 95% CI: 1.22 to 2.62; P = 0.003). Previous history or concomitant active TB at ART initiation was associated with a lower likelihood of the outcome (HR: 0.41; 95% CI: 0.16 to 1.02; P = 0.054). CONCLUSIONS Initiating ART early during AHI may offer an opportunity to mitigate immune damage. Efforts to implement HIV diagnosis and ART initiation during AHI are critical to amplify ART benefits.
Collapse
Affiliation(s)
- Eduarda Grinsztejn
- Department of Hematology-Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH
| | | | - Luciane Velasque
- Departamento de Matemática e Estatística, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, RJ, Brazil
| | - Brenda Hoagland
- Instituto de Pesquisa Clínica Evandro Chagas-Fiocruz, Rio de Janeiro, Brazil
| | | | - Carolina Coutinho
- Instituto de Pesquisa Clínica Evandro Chagas-Fiocruz, Rio de Janeiro, Brazil
| | | | | | | | | | - Beatriz Grinsztejn
- Instituto de Pesquisa Clínica Evandro Chagas-Fiocruz, Rio de Janeiro, Brazil
| | - Valdilea G. Veloso
- Instituto de Pesquisa Clínica Evandro Chagas-Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Doshi RK, Hull S, Broun A, Boyani S, Moch D, Visconti AJ, Castel AD, Baral S, Colasanti J, Rodriguez AE, Jones J, Coffey S, Monroe AK. Lessons learned from U.S. rapid antiretroviral therapy initiation programs. Int J STD AIDS 2023; 34:945-955. [PMID: 37461333 PMCID: PMC11000141 DOI: 10.1177/09564624231185622] [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: 08/03/2023]
Abstract
BACKGROUND Rapid antiretroviral therapy initiation (R-ART) for treatment of HIV has been recommended since 2017, however it has not been adopted widely across the US. PURPOSE The study purpose was to understand facilitators and barriers to R-ART implementation in the U.S. RESEARCH DESIGN This was a qualitative design involving semi-structured interviews. STUDY SAMPLE The study sample was comprised of the medical leadership of nine US HIV clinics that were early implementers of R-ART. DATA COLLECTION AND ANALYSIS In-depth, semi-structured interviews were performed. The Consolidated Framework for Implementation Research (CFIR) was used to guide thematic analysis. RESULTS We identified three main content areas: strong scientific rationale for R-ART, buy-in from multiple key stakeholders, and the condensed timeline of R-ART. The CFIR construct of Evidence Strength and Quality was cited as an important factor in R-ART implementation. Buy-in from key stakeholders and immediate access to medications ensured the success of R-ART implementation. Patient acceptance of the condensed timeline for ART initiation was facilitated when presented in a patient-centered manner, including empathetic communication and addressing other patient needs concurrently. The condensed timeline of R-ART presented logistical challenges and opportunities for the development of intense patient-provider relationships. CONCLUSIONS Results from the analysis showed that R-ART implementation should address the following: 1) logistical planning to implement HIV treatment with a condensed timeline 2) patients' mixed reactions to a new HIV diagnosis and 3) the high cost of HIV medications.
Collapse
Affiliation(s)
- Rupali K Doshi
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
- The HIV/AIDS, Hepatitis, STD and TB Administration (HAHSTA), District of Columbia Department of Health, Washington, DC, USA
| | - Shawnika Hull
- Rutgers University School of Communication and Information, New Brunswick, NJ, USA
| | - Aaron Broun
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Saanjh Boyani
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Darryl Moch
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Adam J Visconti
- The HIV/AIDS, Hepatitis, STD and TB Administration (HAHSTA), District of Columbia Department of Health, Washington, DC, USA
| | - Amanda D Castel
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Stefan Baral
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | | | | | - Joyce Jones
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susa Coffey
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Anne K Monroe
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| |
Collapse
|
5
|
Abstract
Posttreatment controllers (PTCs) are rare HIV-infected individuals who can limit viral rebound after antiretroviral therapy interruption (ATI), but the mechanisms of this remain unclear. To investigate these mechanisms, we quantified various HIV RNA transcripts (via reverse transcription droplet digital PCR [RT-ddPCR]) and cellular transcriptomes (via RNA-seq) in blood cells from PTCs and noncontrollers (NCs) before and two time points after ATI. HIV transcription initiation did not significantly increase after ATI in PTCs or in NCs, whereas completed HIV transcripts increased at early ATI in both groups and multiply-spliced HIV transcripts increased only in NCs. Compared to NCs, PTCs showed lower levels of HIV DNA, more cell-associated HIV transcripts per total RNA at all times, no increase in multiply-spliced HIV RNA at early or late ATI, and a reduction in the ratio of completed/elongated HIV RNA after early ATI. NCs expressed higher levels of the IL-7 pathway before ATI and expressed higher levels of multiple cytokine, inflammation, HIV transcription, and cell death pathways after ATI. Compared to the baseline, the NCs upregulated interferon and cytokine (especially TNF) pathways during early and late ATI, whereas PTCs upregulated interferon and p53 pathways only at early ATI and downregulated gene translation during early and late ATI. In NCs, viral rebound after ATI is associated with increases in HIV transcriptional completion and splicing, rather than initiation. Differences in HIV and cellular transcription may contribute to posttreatment control, including an early limitation of spliced HIV RNA, a delayed reduction in completed HIV transcripts, and the differential expression of the IL-7, p53, and TNF pathways. IMPORTANCE The findings presented here provide new insights into how HIV and cellular gene expression change after stopping ART in both noncontrollers and posttreatment controllers. Posttreatment control is associated with an early ability to limit increases in multiply-spliced HIV RNA, a delayed (and presumably immune-mediated) ability to reverse an initial rise in processive/completed HIV transcripts, and multiple differences in cellular gene expression pathways. These differences may represent correlates or mechanisms of posttreatment control and may provide insight into the development and/or monitoring of therapeutic strategies that are aimed at a functional HIV cure.
Collapse
|
6
|
Zhao J, Chen H, Wan Z, Yu T, Liu Q, Shui J, Wang H, Peng J, Tang S. Evaluation of antiretroviral therapy effect and prognosis between HIV-1 recent and long-term infection based on a rapid recent infection testing algorithm. Front Microbiol 2022; 13:1004960. [PMID: 36483196 PMCID: PMC9722761 DOI: 10.3389/fmicb.2022.1004960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/14/2022] [Indexed: 08/30/2023] Open
Abstract
Early diagnosis of HIV-1 infection and immediate initiation of combination antiretroviral therapy (cART) are important for achieving better virological suppression and quicker immune reconstitution. However, no serological HIV-1 recency testing assay has been approved for clinical use, and the real-world clinical outcomes remain to be explored for the subjects with HIV-1 recent infection (RI) or long-term infection (LI) when antiretroviral therapy is initiated. In this study, a HIV-1 rapid recent-infection testing strip (RRITS) was developed and incorporated into the recent infection testing algorithms (RITAs) to distinguish HIV-1 RI and LI and to assess their clinical outcomes including virological response, the recovery of CD4+ T-cell count and CD4/CD8 ratio and the probability of survival. We found that the concordance between our RRITS and the commercially available LAg-Avidity EIA was 97.13% and 90.63% when detecting the longitudinal and cross-sectional HIV-1 positive samples, respectively. Among the 200 HIV-1 patients analyzed, 22.5% (45/200) of them were RI patients and 77.5% (155/200) were chronically infected and 30% (60/200) of them were AIDS patients. After cART, 4.1% (5/155) of the LI patients showed virological rebound, but none in the RI group. The proportion of CD4+ T-cell count >500 cells/mm3 was significantly higher in RI patients than in LI after 2 years of cART with a hazard ratio (HR) of 2.6 (95% CI: 1.9, 3.6, p < 0.0001) while the probability of CD4/CD8 = 1 was higher in RI than in LI group with a HR of 3.6 (95% CI: 2.2, 5.7, p < 0.0001). Furthermore, the immunological recovery speed was 16 cells/mm3/month for CD4+ T-cell and 0.043/month for the ratio of CD4/CD8 in the RI group, and was bigger in the RI group than in the LI patients (p < 0.05) during the 1st year of cART. The survival probability for LI patients was significantly lower than that for RI patients (p < 0.001). Our results indicated that RRITS combined with RITAs could successfully distinguish HIV-1 RI and LI patients whose clinical outcomes were significantly different after cART. The rapid HIV-1 recency test provides a feasible assay for diagnosing HIV-1 recent infection and a useful tool for predicting the outcomes of HIV-1 patients.
Collapse
Affiliation(s)
- Jianhui Zhao
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hongjie Chen
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhengwei Wan
- Department of Health Management and Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Yu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Quanxun Liu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jingwei Shui
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Haiying Wang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jie Peng
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shixing Tang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW The quest for HIV-1 cure could take advantage of the study of rare individuals that control viral replication spontaneously (elite controllers) or after an initial course of antiretroviral therapy (posttreatment controllers, PTCs). In this review, we will compare back-to-back the immunological and virological features underlying viral suppression in elite controllers and PTCs, and explore their possible contributions to the HIV-1 cure research. RECENT FINDINGS HIV-1 control in elite controllers shows hallmarks of an effective antiviral response, favored by genetic background and possibly associated to residual immune activation. The immune pressure in elite controllers might select against actively transcribing intact proviruses, allowing the persistence of a small and poorly inducible reservoir. Evidence on PTCs is less abundant but preliminary data suggest that antiviral immune responses may be less pronounced. Therefore, these patients may rely on distinct mechanisms, not completely elucidated to date, suppressing HIV-1 transcription and replication. SUMMARY PTCs and elite controllers may control HIV replication using distinct pathways, the elucidation of which may contribute to design future interventional strategies aiming to achieve a functional cure.
Collapse
|
8
|
Nothelle S, Chamberlain AM, Jacobson D, Green AR, Boyd CM, Rocca WA, Fan C, St. Sauver JL. Prevalence of co-occurring serious illness diagnoses and association with health care utilization at the end of life. J Am Geriatr Soc 2022; 70:2621-2629. [PMID: 35593458 PMCID: PMC9489605 DOI: 10.1111/jgs.17881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 04/19/2022] [Accepted: 04/23/2022] [Indexed: 12/30/2022]
Abstract
INTRODUCTION End-of-life care differs by serious illness diagnosis. Cancer and dementia are serious illnesses that have been associated with less intensive end-of-life health care use. It is not known how health care utilization varies in the presence of >1 serious illness. METHODS We used the Rochester Epidemiology Project to identify persons living in a midwestern area who died on July 1, 2017-June 30, 2018 at age ≥65 years, and were seriously ill. We examined the number of emergency department (ED), hospital, and intensive care unit (ICU) stays in the last 6 months and the last 30 days of life. We used Poisson regression to determine the incidence rate ratio for ED, hospital, and ICU stay in the last 6 months and 30 days of life by number of serious illness diagnoses. For cancer and dementia, we examined the effect of an additional serious illness. RESULTS We included a population of 1372 adults who were, on average, 84 years, 52% female, and 96% white. Approximately 41% had multiple serious illnesses. Compared to older adults with 1 serious illness diagnosis, rates of hospitalization, and ICU stay for adults with 2 or ≥3 serious illness diagnoses were at least 1.5 times higher in the last 6 months and the last 30 days of life. Rates of ED visits were significantly higher for older adults with 2 or ≥3 serious illness diagnoses in the last 6 months of life, but only higher for those with ≥3 versus 1 serious illness diagnosis in the last 30 days of life. For both cancer and dementia, rates of ED visits, hospitalization and ICU stay were lower for the condition alone than when an additional serious illness diagnosis was present. CONCLUSION Having multiple serious illnesses increases the risk of health care utilization at the end of life.
Collapse
Affiliation(s)
- Stephanie Nothelle
- Center for Transformative Geriatrics Research, Division of Geriatric Medicine and GerontologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Alanna M. Chamberlain
- Division of Epidemiology, Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA,Robert D. and Patricia E. Kern Center for the Science of Health Care DeliveryMayo ClinicRochesterMinnesotaUSA
| | - Debra Jacobson
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Ariel R. Green
- Center for Transformative Geriatrics Research, Division of Geriatric Medicine and GerontologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Cynthia M. Boyd
- Center for Transformative Geriatrics Research, Division of Geriatric Medicine and GerontologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Walter A. Rocca
- Division of Epidemiology, Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA,Department of NeurologyMayo ClinicRochesterMinnesotaUSA,Women's Health Research CenterMayo ClinicRochesterMinnesotaUSA
| | - Chun Fan
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Jennifer L. St. Sauver
- Division of Epidemiology, Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA,Robert D. and Patricia E. Kern Center for the Science of Health Care DeliveryMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
9
|
Pace M, Ogbe A, Hurst J, Robinson N, Meyerowitz J, Olejniczak N, Thornhill JP, Jones M, Waters A, Lwanga J, Kuldanek K, Hall R, Zacharopoulou P, Martin GE, Brown H, Nwokolo N, Peppa D, Fox J, Fidler S, Frater J. Impact of antiretroviral therapy in primary HIV infection on natural killer cell function and the association with viral rebound and HIV DNA following treatment interruption. Front Immunol 2022; 13:878743. [PMID: 36110857 PMCID: PMC9468877 DOI: 10.3389/fimmu.2022.878743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Natural Killer (NK) cells play a key role in controlling HIV replication, with potential downstream impact on the size of the HIV reservoir and likelihood of viral rebound after antiretroviral therapy (ART) cessation. It is therefore important to understand how primary HIV infection (PHI) disrupts NK cell function, and how these functions are restored by early ART. We examined the impact of commencing ART during PHI on phenotypic and functional NK cell markers at treatment initiation (baseline), 3 months, 1 year, and 2 years in seven well-characterised participants in comparison to HIV seronegative volunteers. We then examined how those NK cell properties differentially impacted by ART related to time to viral rebound and HIV DNA levels in 44 individuals from the SPARTAC trial who stopped ART after 48 weeks treatment, started during PHI. NK cell markers that were significantly different between the seven people with HIV (PWH) treated for 2 years and HIV uninfected individuals included NKG2C levels in CD56dim NK cells, Tim-3 expression in CD56bright NK cells, IFN-γ expressed by CD56dim NK cells after IL-12/IL-18 stimulation and the fraction of Eomes-/T-bet+ in CD56dim and CD56bright NK cells. When exploring time to viral rebound after stopping ART among the 44 SPARTAC participants, no single NK phenotypic marker correlated with control. Higher levels of IL-12/IL-18 mediated NK cell degranulation at baseline were associated with longer times to viral rebound after treatment interruption (P=0.028). Additionally, we found higher fractions of CD56dim NK cells in individuals with lower levels of HIV DNA (P=0.048). NKG2A and NKp30 levels in CD56neg NK cells were higher in patients with lower HIV DNA levels (p=0.00174, r=-0.49 and p=0.03, r= -0.327, respectively) while CD27 levels were higher in those with higher levels of HIV DNA (p=0.026). These data show NK cell functions are heterogeneously impacted by HIV infection with a mixed picture of resolution on ART, and that while NK cells may affect HIV DNA levels and time to viral rebound, no single NK cell marker defined delayed viral rebound.
Collapse
Affiliation(s)
- Matthew Pace
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ane Ogbe
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jacob Hurst
- Etcembly Ltd, Harwell Campus, Didcot, United Kingdom
| | - Nicola Robinson
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jodi Meyerowitz
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Natalia Olejniczak
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - John P. Thornhill
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mathew Jones
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Anele Waters
- Department of Infection, Guys and St Thomas’ National Health Service (NHS) Trust, London, United Kingdom
| | - Julianne Lwanga
- Department of Infection, Guys and St Thomas’ National Health Service (NHS) Trust, London, United Kingdom
| | - Kristen Kuldanek
- Department of HIV Medicine, St Mary’s Hospital, Imperial College Healthcare National Health Service (NHS) Trust, London, United Kingdom
| | - Rebecca Hall
- Department of HIV Medicine, St Mary’s Hospital, Imperial College Healthcare National Health Service (NHS) Trust, London, United Kingdom
| | | | - Genevieve E. Martin
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| | - Helen Brown
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nneka Nwokolo
- Department of HIV/GUM, Chelsea and Westminster Hospital, London, United Kingdom
| | - Dimitra Peppa
- Division of Infection and Immunity, University College, London, United Kingdom
| | - Julie Fox
- Department of Infection, Guys and St Thomas’ National Health Service (NHS) Trust, London, United Kingdom
| | - Sarah Fidler
- Department of HIV Medicine, St Mary’s Hospital, Imperial College Healthcare National Health Service (NHS) Trust, London, United Kingdom
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- National Institute for Health and Care Research (NIHR) Imperial College Biomedical Research Centre, London, United Kingdom
| | - John Frater
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
10
|
Lau CY, Adan MA, Earhart J, Seamon C, Nguyen T, Savramis A, Adams L, Zipparo ME, Madeen E, Huik K, Grossman Z, Chimukangara B, Wulan WN, Millo C, Nath A, Smith BR, Ortega-Villa AM, Proschan M, Wood BJ, Hammoud DA, Maldarelli F. Imaging and biopsy of HIV-infected individuals undergoing analytic treatment interruption. Front Med (Lausanne) 2022; 9:979756. [PMID: 36072945 PMCID: PMC9441850 DOI: 10.3389/fmed.2022.979756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background HIV persistence during antiretroviral therapy (ART) is the principal obstacle to cure. Lymphoid tissue is a compartment for HIV, but mechanisms of persistence during ART and viral rebound when ART is interrupted are inadequately understood. Metabolic activity in lymphoid tissue of patients on long-term ART is relatively low, and increases when ART is stopped. Increases in metabolic activity can be detected by 18F-fluorodeoxyglucose Positron Emission Tomography (FDG-PET) and may represent sites of HIV replication or immune activation in response to HIV replication. Methods FDG-PET imaging will be used to identify areas of high and low metabolic uptake in lymphoid tissue of individuals undergoing long-term ART. Baseline tissue samples will be collected. Participants will then be randomized 1:1 to continue or interrupt ART via analytic treatment interruption (ATI). Image-guided biopsy will be repeated 10 days after ATI initiation. After ART restart criteria are met, image-guided biopsy will be repeated once viral suppression is re-achieved. Participants who continued ART will have a second FDG-PET and biopsies 12–16 weeks after the first. Genetic characteristics of HIV populations in areas of high and low FDG uptake will be assesed. Optional assessments of non-lymphoid anatomic compartments may be performed to evaluate HIV populations in distinct anatomic compartments. Anticipated results We anticipate that PET standardized uptake values (SUV) will correlate with HIV viral RNA in biopsies of those regions and that lymph nodes with high SUV will have more viral RNA than those with low SUV within a patient. Individuals who undergo ATI are expected to have diverse viral populations upon viral rebound in lymphoid tissue. HIV populations in tissues may initially be phylogenetically diverse after ATI, with emergence of dominant viral species (clone) over time in plasma. Dominant viral species may represent the same HIV population seen before ATI. Discussion This study will allow us to explore utility of PET for identification of HIV infected cells and determine whether high FDG uptake respresents areas of HIV replication, immune activation or both. We will also characterize HIV infected cell populations in different anatomic locations. The protocol will represent a platform to investigate persistence and agents that may target HIV populations. Study protocol registration Identifier: NCT05419024.
Collapse
Affiliation(s)
- Chuen-Yen Lau
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
- *Correspondence: Chuen-Yen Lau
| | - Matthew A. Adan
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Jessica Earhart
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Cassie Seamon
- Critical Care Medicine Department, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Thuy Nguyen
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Ariana Savramis
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Lindsey Adams
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Mary-Elizabeth Zipparo
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Erin Madeen
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Kristi Huik
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Zehava Grossman
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Benjamin Chimukangara
- Critical Care Medicine Department, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Wahyu Nawang Wulan
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Corina Millo
- PET Department, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Avindra Nath
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Bryan R. Smith
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Ana M. Ortega-Villa
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Michael Proschan
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Bradford J. Wood
- Interventional Radiology, Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Dima A. Hammoud
- Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| |
Collapse
|
11
|
Chéret A, Bauer R, Meiffrédy V, Lopez P, Ajana F, Lacombe K, Morlat P, Lascoux C, Reynes J, Calin R, Abel S, Goujard C, Rouzioux C, Avettand-Fenoel V, Meyer L. Once-daily dolutegravir versus darunavir plus cobicistat in adults at the time of primary HIV-1 infection: the OPTIPRIM2-ANRS 169 randomized, open-label, Phase 3 trial. J Antimicrob Chemother 2022; 77:2506-2515. [PMID: 35762503 DOI: 10.1093/jac/dkac207] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Whether integrase strand transfer inhibitors (INSTIs) can decrease HIV-1 DNA levels more rapidly than boosted PIs during primary HIV-1 infection (PHI) is unknown. We hypothesized that once-daily dolutegravir/tenofovir/emtricitabine could reduce the viral reservoir through rapid viral replication control further than once-daily darunavir/cobicistat/tenofovir/emtricitabine. METHODS The OPTIPRIM2-ANRS 169 study was a randomized (1:1), open-label, multicentre trial in adults with ≤5 or ≤3 HIV antibodies detected, respectively, by western blot or immunoblot in the last 10 days. The primary endpoint was total HIV-1 DNA levels in PBMCs at Week 48 (W48) adjusted for baseline levels. The main secondary endpoint was HIV-1 RNA level decrease. RESULTS Between April 2017 and August 2018, 101 patients were included from 31 hospitals. Most patients were men (93%), the median age was 36 years and 17% were Fiebig stage ≤3. The median (IQR) plasma HIV-1 RNA and DNA levels were, respectively, 5.8 (5.0-6.6) and 3.87 (3.52-4.15) log10 copies/million PBMCs. The median (IQR) decreases in HIV-1 DNA levels at W48 were -1.48 (-1.74 to -1.06) and -1.39 (-1.55 to -0.98) log10 copies/million PBMCs in the dolutegravir and darunavir/cobicistat groups, respectively (P = 0.52). Plasma HIV-1 RNA levels were <50 copies/mL in 24% versus 0% of patients in the dolutegravir and darunavir/cobicistat groups at W4, 55% versus 2% at W8, 67% versus 17% at W12, and 94% versus 90% at W48, respectively. CONCLUSIONS Dolutegravir-based and darunavir-based regimens initiated during PHI strongly and similarly decreased the blood reservoir size. Considering the rapid viral suppression during a period of high HIV-1 transmission risk, dolutegravir-based regimens are a major first-line option.
Collapse
Affiliation(s)
- Antoine Chéret
- Service de Médecine Interne, APHP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM, U1016, CNRS, UMR8104, Institut Cochin, Paris, France
| | | | | | - Pauline Lopez
- INSERM, U1016, CNRS, UMR8104, Institut Cochin, Paris, France.,APHP, Laboratoire de Microbiologie Clinique, Hôpital Necker-Enfants Malades, Paris, France.,Université de Paris, Faculté de Médecine, Paris, France
| | - Faïza Ajana
- Service de Maladies Infectieuses et Tropicales, Hôpital Dron, Tourcoing, France
| | - Karine Lacombe
- Service de Maladies Infectieuses et Tropicales, Hôpital St Antoine, APHP, Paris, France.,Sorbonne Université, IPLESP Inserm UMR, Hôpital St Antoine, APHP, Paris, France
| | - Philippe Morlat
- Service de Médecine Interne et Maladies Infectieuses, CHU Saint-André, Université de Bordeaux, Bordeaux, France
| | - Caroline Lascoux
- Service de Maladies Infectieuses et Tropicales, Hôpital Saint-Louis, APHP, Paris, France
| | - Jacques Reynes
- Département de Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire de Montpellier, Montpellier, France.,TransVIHMI, Université de Montpellier, IRD, INSERM, Montpellier, France
| | - Ruxandra Calin
- Service de Maladies Infectieuses et Tropicales, Hôpital Tenon, APHP, Paris, France
| | - Sylvie Abel
- Service de maladies Infectieuses et Tropicales, CHU de Martinique, Fort-de-France, France.,Pathogenesis and Control of Chronic Infections, Montpellier University, Antilles University, INSERM, EFS, Montpellier, France
| | - Cécile Goujard
- Service de Médecine Interne, APHP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,INSERM, CESP U1018, Université Paris Saclay, APHP, Le Kremlin-Bicêtre, France
| | | | - Véronique Avettand-Fenoel
- INSERM, U1016, CNRS, UMR8104, Institut Cochin, Paris, France.,APHP, Laboratoire de Microbiologie Clinique, Hôpital Necker-Enfants Malades, Paris, France.,Université de Paris, Faculté de Médecine, Paris, France
| | - Laurence Meyer
- INSERM SC10-US19, Villejuif, France.,INSERM, CESP U1018, Université Paris Saclay, APHP, Le Kremlin-Bicêtre, France
| |
Collapse
|
12
|
Nothelle S, Kelley AS, Zhang T, Roth DL, Wolff JL, Boyd C. Fragmentation of care in the last year of life: Does dementia status matter? J Am Geriatr Soc 2022; 70:2320-2329. [PMID: 35488709 PMCID: PMC9378534 DOI: 10.1111/jgs.17827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 12/01/2022]
Abstract
Background Care at the end of life is commonly fragmented; however, little is known about commonly used measures of fragmentation of care in the last year of life (LYOL). We sought to understand differences in fragmentation of care by dementia status among seriously ill older adults in the LYOL. Methods We analyzed data from adults ≥65 years in the National Health and Aging Trends Study who died and had linked 2011–2017 Medicare fee‐for‐service claims for ≥12 months before death. We categorized older adults as having serious illness due to dementia (hereafter dementia), non‐dementia serious illness or no serious illness. For outpatient fragmentation, we calculated the Bice–Boxerman continuity of care index (COC), which measures care concentration, and the known provider of care index (KPC), which measures the proportion of clinicians who were previously seen. For acute care fragmentation, we divided the number of hospitals and emergency departments visited by the total number of visits. We built separate multivariable quantile regression models for each measure of fragmentation. Results Of 1793 older adults, 42% had dementia, 53% non‐dementia serious illness and 5% neither. Older adults with dementia had fewer hospitalizations than older adults with non‐dementia serious illness but more than older adults without serious illness (mean 1.9 vs 2.3 vs 1, p = 0.002). In adjusted models, compared to older adults with non‐dementia serious illness, those with dementia had significantly less fragmented care across all quantiles of COC (range 0.016–0.110) but a lower predicted 90th percentile of KPC, meaning more older adults with dementia had extremely fragmented care on the KPC measure. There was no significant difference in acute care fragmentation. Conclusions In the LYOL, older adults with dementia have fewer healthcare encounters and less fragmentation of care by the COC index than older adults with non‐dementia serious illness.
Collapse
Affiliation(s)
- Stephanie Nothelle
- Center for Transformative Geriatric Research, Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Roger C. Lipitz Center for Integrated Health Care, Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Amy S Kelley
- Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Talan Zhang
- Center on Aging and Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - David L Roth
- Center for Transformative Geriatric Research, Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Center on Aging and Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jennifer L Wolff
- Center for Transformative Geriatric Research, Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Roger C. Lipitz Center for Integrated Health Care, Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Center on Aging and Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Cynthia Boyd
- Center for Transformative Geriatric Research, Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Roger C. Lipitz Center for Integrated Health Care, Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Center on Aging and Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Azamar-Alonso A, Mbuagbaw L, Smaill F, Bautista-Arredondo SA, Costa AP, Tarride JE. Virologic failure in people living with HIV in 1st line ART: A 10-year Mexican population-based study. Int J STD AIDS 2022; 33:363-373. [PMID: 35118929 PMCID: PMC8958557 DOI: 10.1177/09564624211067036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background In Mexico, the number of people living with HIV (PLWH) receiving antiretroviral therapy (ART) has increased in the last 20 years. The elimination of a CD4 threshold to initiate publicly funded ART was a major policy implemented in 2014. The study objective was to assess the determinants of Virologic Failure (VF) in Mexican PLWH on first-line ART between 2008 and 2017 and to evaluate the effects of changes following the 2014 policy. Methods A 10-year patient-level data analysis was conducted using the Mexican SALVAR database. The main outcome was the proportion of PLWH with VF. A multivariable logistic regression was conducted to identify the association between covariates and VF before and after the 2014 policy implementation. Results We found a lower proportion of people with VF in 2014–2017 compared with 2008–2013 (50% vs 33%, p<0.001). The multivariable analysis showed a reduction in the odds of virologic failure after 2014 (Odds ratio: 0.50 [95% CI: 0.48–0.51]). Place of treatment and level of deprivation were significant predictors of VF in during 2014–2017, but not before. Conclusion This study indicates that, by lowering threshold levels of CD4 required for treatment initiation in Mexico, a higher number of PLWH initiated treatment during 2014–2017, compared to 2008–2013 and the odds of VF were reduced.
Collapse
Affiliation(s)
- Amilcar Azamar-Alonso
- Department of Health Research Methods, Evidence, and Impact, 3710McMaster University, Hamilton, Ontario, Canada.,Gilead Sciences Inc., Foster City, CA, United States
| | - Lawrence Mbuagbaw
- Department of Health Research Methods, Evidence, and Impact, 3710McMaster University, Hamilton, Ontario, Canada.,Biostatistics Unit, Father Sean O'Sullivan Research Centre, Hamilton, ON, Canada
| | - Fiona Smaill
- ChB Department of Pathology and Molecular Medicine, Faculty of Health Sciences, 3710McMaster University, Hamilton, Ontario, Canada
| | | | - Andrew P Costa
- Department of Health Research Methods, Evidence, and Impact, 3710McMaster University, Hamilton, Ontario, Canada.,Centre for Health Economics and Policy Analysis, 3710McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, 3710McMaster University, Hamilton, Ontario, Canada
| | - Jean-Eric Tarride
- Department of Health Research Methods, Evidence, and Impact, 3710McMaster University, Hamilton, Ontario, Canada.,Centre for Health Economics and Policy Analysis, 3710McMaster University, Hamilton, Ontario, Canada.,The Research Institute of St Joe's Hamilton, Hamilton, ON, Canada.,McMaster Chair in Health Technology Management Hamilton, Hamilton, ON, Canada
| |
Collapse
|
14
|
Zacharopoulou P, Marchi E, Ogbe A, Robinson N, Brown H, Jones M, Parolini L, Pace M, Grayson N, Kaleebu P, Rees H, Fidler S, Goulder P, Klenerman P, Frater J. Expression of type I interferon-associated genes at antiretroviral therapy interruption predicts HIV virological rebound. Sci Rep 2022; 12:462. [PMID: 35013427 PMCID: PMC8748440 DOI: 10.1038/s41598-021-04212-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/08/2021] [Indexed: 12/25/2022] Open
Abstract
Although certain individuals with HIV infection can stop antiretroviral therapy (ART) without viral load rebound, the mechanisms under-pinning 'post-treatment control' remain unclear. Using RNA-Seq we explored CD4 T cell gene expression to identify evidence of a mechanism that might underpin virological rebound and lead to discovery of associated biomarkers. Fourteen female participants who received 12 months of ART starting from primary HIV infection were sampled at the time of stopping therapy. Two analysis methods (Differential Gene Expression with Gene Set Enrichment Analysis, and Weighted Gene Co-expression Network Analysis) were employed to interrogate CD4+ T cell gene expression data and study pathways enriched in post-treatment controllers versus early rebounders. Using independent analysis tools, expression of genes associated with type I interferon responses were associated with a delayed time to viral rebound following treatment interruption (TI). Expression of four genes identified by Cox-Lasso (ISG15, XAF1, TRIM25 and USP18) was converted to a Risk Score, which associated with rebound (p < 0.01). These data link transcriptomic signatures associated with innate immunity with control following stopping ART. The results from this small sample need to be confirmed in larger trials, but could help define strategies for new therapies and identify new biomarkers for remission.
Collapse
Affiliation(s)
- P Zacharopoulou
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - E Marchi
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - A Ogbe
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - N Robinson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - H Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - M Jones
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - L Parolini
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - M Pace
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - N Grayson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - P Kaleebu
- Medical Research Council/Uganda Virus Research Institute, Entebbe, Uganda
| | - H Rees
- Wits Reproductive Health and HIV Institute of the University of the Witwatersrand in Johannesburg, Johannesburg, South Africa
| | - S Fidler
- Division of Medicine, Wright Fleming Institute, Imperial College, London, UK
- Imperial College NIHR Biomedical Research Centre, London, UK
| | - P Goulder
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - P Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- National Institute of Health Research Biomedical Research Centre, Oxford, UK
| | - J Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- National Institute of Health Research Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
15
|
Liu A, Wei Q, Lin H, Ding Y, Sun YV, Zhao D, He J, Ma Z, Li F, Zhou S, Chen X, Shen W, Gao M, He N. Baseline Characteristics of Mitochondrial DNA and Mutations Associated With Short-Term Posttreatment CD4+T-Cell Recovery in Chinese People With HIV. Front Immunol 2022; 12:793375. [PMID: 34970271 PMCID: PMC8712318 DOI: 10.3389/fimmu.2021.793375] [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: 10/12/2021] [Accepted: 11/16/2021] [Indexed: 11/25/2022] Open
Abstract
Background Mitochondrial DNA (mtDNA) profiles and contributions of mtDNA variants to CD4+T-cell recovery in Euramerican people living with HIV (PLWH) may not be transferred to East-Asian PLWH, highlighting the need to consider more regional studies. We aimed to identify mtDNA characteristics and mutations that explain the variability of short-term CD4+T-cell recovery in East-Asian PLWH. Method Eight hundred fifty-six newly reported antiretroviral therapy (ART)-naïve Chinese PLWH from the Comparative HIV and Aging Research in Taizhou (CHART) cohort (Zhejiang Province, Eastern China) were enrolled. MtDNA was extracted from peripheral whole blood of those PLWH at HIV diagnosis, amplified, and sequenced using polymerase chain reaction and gene array. Characterization metrics such as mutational diversity and momentum were developed to delineate baseline mtDNA mutational patterns in ART-naïve PLWH. The associations between mtDNA genome-wide single nucleotide variants and CD4+T-cell recovery after short-term (within ~48 weeks) ART in 724 PLWH were examined using bootstrapping median regressions. Results Of 856 participants, 74.18% and 25.82% were male and female, respectively. The median age was 37 years; 94.51% were of the major Han ethnicity, and 69.04% and 28.62% were of the heterosexual and homosexual transmission, respectively. We identified 2,352 types of mtDNA mutations and mtDNA regions D-loop, ND5, CYB, or RNR1 with highest mutational diversity or volume. Female PLWH rather than male PLWH at the baseline showed remarkable age-related uptrends of momentum and mutational diversity as well as correlations between CD4+T <200 (cells/μl) and age-related uptrends of mutational diversity in many mtDNA regions. After adjustments of important sociodemographic and clinical variables, m.1005T>C, m.1824T>C, m.3394T>C, m.4491G>A, m.7828A>G, m.9814T>C, m.10586G>A, m.12338T>C, m.13708G>A, and m.14308T>C (at the Bonferroni-corrected significance) were negatively associated with short-term CD4+T-cell recovery whereas m.93A>G, m.15218A>G, and m.16399A>G were positively associated with short-term CD4+T-cell recovery. Conclusion Our baseline mtDNA characterization stresses the attention to East-Asian female PLWH at risk of CD4+T-cell loss-related aging and noncommunicable chronic diseases. Furthermore, mtDNA variants identified in regression analyses account for heterogeneity in short-term CD4+T-cell recovery of East-Asian PLWH. These results may help individualize the East-Asian immune recovery strategies under complicated HIV management caused by CD4+T-cell loss.
Collapse
Affiliation(s)
- Anni Liu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China.,Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.,Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, United States
| | - Qian Wei
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China.,Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Haijiang Lin
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China.,Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.,Department of AIDS/STD Control and Prevention, Taizhou City Center for Disease Control and Prevention, Taizhou, China
| | - Yingying Ding
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China.,Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States.,Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, GA, United States
| | - Dan Zhao
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China.,Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Jiayu He
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China.,Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Zhonghui Ma
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China.,Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Feihu Li
- School of Mathematical Sciences, Fudan University, Shanghai, China
| | - Sujuan Zhou
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China.,Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Xiaoxiao Chen
- Department of AIDS/STD Control and Prevention, Taizhou City Center for Disease Control and Prevention, Taizhou, China
| | - Weiwei Shen
- Department of AIDS/STD Control and Prevention, Taizhou City Center for Disease Control and Prevention, Taizhou, China
| | - Meiyang Gao
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China.,Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Na He
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China.,Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.,Key Laboratory of Health Technology Assessment, National Commission of Health, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Phetsouphanh C, Phalora P, Hackstein CP, Thornhill J, Munier CML, Meyerowitz J, Murray L, VanVuuren C, Goedhals D, Drexhage L, Russell RA, Sattentau QJ, Mak JYW, Fairlie DP, Fidler S, Kelleher AD, Frater J, Klenerman P. Human MAIT cells respond to and suppress HIV-1. eLife 2021; 10:e50324. [PMID: 34951583 PMCID: PMC8752121 DOI: 10.7554/elife.50324] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/23/2021] [Indexed: 11/13/2022] Open
Abstract
Human MAIT cells sit at the interface between innate and adaptive immunity, are polyfunctional and are capable of killing pathogen infected cells via recognition of the Class IB molecule MR1. MAIT cells have recently been shown to possess an antiviral protective role in vivo and we therefore sought to explore this in relation to HIV-1 infection. There was marked activation of MAIT cells in vivo in HIV-1-infected individuals, which decreased following ART. Stimulation of THP1 monocytes with R5 tropic HIVBAL potently activated MAIT cells in vitro. This activation was dependent on IL-12 and IL-18 but was independent of the TCR. Upon activation, MAIT cells were able to upregulate granzyme B, IFNγ and HIV-1 restriction factors CCL3, 4, and 5. Restriction factors produced by MAIT cells inhibited HIV-1 infection of primary PBMCs and immortalized target cells in vitro. These data reveal MAIT cells to be an additional T cell population responding to HIV-1, with a potentially important role in controlling viral replication at mucosal sites.
Collapse
Affiliation(s)
- Chansavath Phetsouphanh
- Peter Medawar Building for Pathogen Research, University of OxfordOxfordUnited Kingdom
- The Kirby Institute, University of New South WalesSydneyAustralia
| | - Prabhjeet Phalora
- Peter Medawar Building for Pathogen Research, University of OxfordOxfordUnited Kingdom
| | | | | | | | - Jodi Meyerowitz
- Peter Medawar Building for Pathogen Research, University of OxfordOxfordUnited Kingdom
| | - Lyle Murray
- Peter Medawar Building for Pathogen Research, University of OxfordOxfordUnited Kingdom
| | | | - Dominique Goedhals
- Division of Virology, University of the Free State/National Health Laboratory ServiceFree StateSouth Africa
| | - Linnea Drexhage
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
| | - Rebecca A Russell
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
| | - Quentin J Sattentau
- Sir William Dunn School of Pathology, University of OxfordOxfordUnited Kingdom
| | - Jeffrey YW Mak
- ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of QueenslandBrisbaneAustralia
- ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of QueenslandBrisbaneAustralia
| | - David P Fairlie
- ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of QueenslandBrisbaneAustralia
- ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of QueenslandBrisbaneAustralia
| | | | | | - John Frater
- Peter Medawar Building for Pathogen Research, University of OxfordOxfordUnited Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
17
|
Granger LA, Huettner I, Debeljak F, Kaleebu P, Schechter M, Tambussi G, Weber J, Miro JM, Phillips R, Babiker A, Cooper DA, Fisher M, Ramjee G, Fidler S, Frater J, Fox J, Doores KJ. Broadly neutralizing antibody responses in the longitudinal primary HIV-1 infection Short Pulse Anti-Retroviral Therapy at Seroconversion cohort. AIDS 2021; 35:2073-2084. [PMID: 34127581 PMCID: PMC8505148 DOI: 10.1097/qad.0000000000002988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/11/2021] [Accepted: 03/02/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Development of immunogens that elicit an anti-HIV-1 broadly neutralizing antibody (bnAb) response will be a key step in the development of an effective HIV-1 vaccine. Although HIV-1 bnAb epitopes have been identified and mechanisms of action studied, current HIV-1 envelope-based immunogens do not elicit HIV-1 bnAbs in humans or animal models. A better understanding of how HIV-1 bnAbs arise during infection and the clinical factors associated with bnAb development may be critical for HIV-1 immunogen design efforts. DESIGN AND METHODS Longitudinal plasma samples from the treatment-naive control arm of the Short Pulse Anti-Retroviral Therapy at Seroconversion (SPARTAC) primary HIV-1 infection cohort were used in an HIV-1 pseudotype neutralization assay to measure the neutralization breadth, potency and specificity of bnAb responses over time. RESULTS In the SPARTAC cohort, development of plasma neutralization breadth and potency correlates with duration of HIV infection and high viral loads, and typically takes 3-4 years to arise. bnAb activity was mostly directed to one or two bnAb epitopes per donor and more than 60% of donors with the highest plasma neutralization having bnAbs targeted towards glycan-dependent epitopes. CONCLUSION This study highlights the SPARTAC cohort as an important resource for more in-depth analysis of bnAb developmental pathways.
Collapse
Affiliation(s)
- Luke A. Granger
- Department of Infectious Diseases, King's College London, Guy's Hospital, Great Maze Pond, London, UK
- Department of Infectious Disease, Imperial College London
| | - Isabella Huettner
- Department of Infectious Diseases, King's College London, Guy's Hospital, Great Maze Pond, London, UK
| | - Franka Debeljak
- Department of Infectious Diseases, King's College London, Guy's Hospital, Great Maze Pond, London, UK
| | - Pontiano Kaleebu
- Medical Research Council/Uganda Virus Research Institute, Entebbe, Uganda
| | - Mauro Schechter
- Projeto Praça Onze, Hospital Escola São Francisco de Assis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giuseppe Tambussi
- Department of Infectious Diseases, Ospedale San Raffaele, Milan, Italy
| | | | - Jose M. Miro
- Infectious Diseases Service. Hospital Clinic–Institut d’investigacions Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Rodney Phillips
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, UK
| | - Abdel Babiker
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology
| | - David A. Cooper
- St Vincent's Centre for Applied Medical Research and The Kirby Institute, UNSW Australia, Sydney, NSW, Australia
| | - Martin Fisher
- Brighton and Sussex University Hospitals, Brighton, UK
| | - Gita Ramjee
- HIV Prevention Research Unit, South African Medical Research Council, Durban, South Africa
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London
- NIHR Imperial Biomedical Research Centre, London
| | - John Frater
- Nuffield Department of Medicine, Oxford University
- Oxford NIHR Biomedical Research Centre, Oxford
| | - Julie Fox
- Department of Infectious Diseases, King's College London, Guy's Hospital, Great Maze Pond, London, UK
- King's College NIHR Research Biomedical Research Centre, London, UK
| | - Katie J. Doores
- Department of Infectious Diseases, King's College London, Guy's Hospital, Great Maze Pond, London, UK
| |
Collapse
|
18
|
Naidoo KK, Ndumnego OC, Ismail N, Dong KL, Ndung'u T. Antigen Presenting Cells Contribute to Persistent Immune Activation Despite Antiretroviral Therapy Initiation During Hyperacute HIV-1 Infection. Front Immunol 2021; 12:738743. [PMID: 34630420 PMCID: PMC8498034 DOI: 10.3389/fimmu.2021.738743] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/09/2021] [Indexed: 12/26/2022] Open
Abstract
Human immunodeficiency virus (HIV)-induced changes in immune cells during the acute phase of infection can cause irreversible immunological damage and predict the rate of disease progression. Antiretroviral therapy (ART) remains the most effective strategy for successful immune restoration in immunocompromised people living with HIV and the earlier ART is initiated after infection, the better the long-term clinical outcomes. Here we explored the effect of ART on peripheral antigen presenting cell (APC) phenotype and function in women with HIV-1 subtype C infection who initiated ART in the hyperacute phase (before peak viremia) or during chronic infection. Peripheral blood mononuclear cells obtained longitudinally from study participants were used for immunophenotyping and functional analysis of monocytes and dendritic cells (DCs) using multiparametric flow cytometry and matched plasma was used for measurement of inflammatory markers IL-6 and soluble CD14 (sCD14) by enzyme-linked immunosorbent assay. HIV infection was associated with expansion of monocyte and plasmacytoid DC (pDC) frequencies and perturbation of monocyte subsets compared to uninfected persons despite antiretroviral treatment during hyperacute infection. Expression of activation marker CD69 on monocytes and pDCs in early treated HIV was similar to uninfected individuals. However, despite early ART, HIV infection was associated with elevation of plasma IL-6 and sCD14 levels which correlated with monocyte activation. Furthermore, HIV infection with or without early ART was associated with downmodulation of the co-stimulatory molecule CD86. Notably, early ART was associated with preserved toll-like receptor (TLR)-induced IFN-α responses of pDCs. Overall, this data provides evidence of the beneficial impact of ART initiated in hyperacute infection in preservation of APC functional cytokine production activity; but also highlights persistent inflammation facilitated by monocyte activation even after prolonged viral suppression and suggests the need for therapeutic interventions that target residual immune activation.
Collapse
Affiliation(s)
- Kewreshini K Naidoo
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | - Nasreen Ismail
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Krista L Dong
- Females Rising Through Education, Support and Health, Durban, South Africa.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States
| | - Thumbi Ndung'u
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa.,Africa Health Research Institute, Durban, South Africa.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States.,Max Planck Institute for Infection Biology, Berlin, Germany.,Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
19
|
Petkov S, Chiodi F. Distinct transcriptomic profiles of naïve CD4+ T cells distinguish HIV-1 infected patients initiating antiretroviral therapy at acute or chronic phase of infection. Genomics 2021; 113:3487-3500. [PMID: 34425224 DOI: 10.1016/j.ygeno.2021.08.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023]
Abstract
We analyzed the whole transcriptome characteristics of blood CD4+ T naïve (TN) cells isolated from HIV-1 infected patients starting ART at acute (early ART = EA; n = 13) or chronic (late ART = LA; n = 11) phase of infection and controls (C; n = 15). RNA sequencing revealed 389 differentially expressed genes (DEGs) in EA and 810 in LA group in relation to controls. Comparison of the two groups of patients showed 183 DEGs. We focused on DEGs involved in apoptosis, inflammation and immune response. Clustering showed a poor separation of EA from C suggesting that these two groups present a similar transcriptomic profile of CD4+ TN cells. The comparison of EA and LA patients resulted in a high cluster purity revealing that different biological dysfunctions characterize EA and LA patients. The upregulated expression of several inflammatory chemokine genes distinguished the patient groups from C; CCL2 and CCL7, however, were downregulated in EA compared to LA patients. BCL2, an anti-apoptotic factor pivotal for naïve T cell homeostasis, distinguished both EA and LA from C. The expression of several DEGs involved in different inflammatory processes (TLR4, PTGS2, RAG1, IFNA16) was lower in EA compared LA. We conclude that although the transcriptome of CD4+ TN cells isolated from patients initiating ART at acute infection reveals a more quiescent phenotype, the survival profile of these cells still appears to be affected. Our results show that the detrimental process of inflammation is under more efficient control in EA patients.
Collapse
Affiliation(s)
- Stefan Petkov
- Department of Microbiology, Tumor and Cell Biology at Biomedicum, Karolinska Institutet, Solna, Sweden.
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology at Biomedicum, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
20
|
Yates KB, Tonnerre P, Martin GE, Gerdemann U, Al Abosy R, Comstock DE, Weiss SA, Wolski D, Tully DC, Chung RT, Allen TM, Kim AY, Fidler S, Fox J, Frater J, Lauer GM, Haining WN, Sen DR. Epigenetic scars of CD8 + T cell exhaustion persist after cure of chronic infection in humans. Nat Immunol 2021; 22:1020-1029. [PMID: 34312547 DOI: 10.1038/s41590-021-00979-1] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
T cell exhaustion is an induced state of dysfunction that arises in response to chronic infection and cancer. Exhausted CD8+ T cells acquire a distinct epigenetic state, but it is not known whether that chromatin landscape is fixed or plastic following the resolution of a chronic infection. Here we show that the epigenetic state of exhaustion is largely irreversible, even after curative therapy. Analysis of chromatin accessibility in HCV- and HIV-specific responses identifies a core epigenetic program of exhaustion in CD8+ T cells, which undergoes only limited remodeling before and after resolution of infection. Moreover, canonical features of exhaustion, including super-enhancers near the genes TOX and HIF1A, remain 'epigenetically scarred.' T cell exhaustion is therefore a conserved epigenetic state that becomes fixed and persists independent of chronic antigen stimulation and inflammation. Therapeutic efforts to reverse T cell exhaustion may require new approaches that increase the epigenetic plasticity of exhausted T cells.
Collapse
Affiliation(s)
- Kathleen B Yates
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pierre Tonnerre
- Division of Gastroenterology, Liver Center, Massachusetts General Hospital, Boston, MA, USA.,Inserm U976, Institut de Recherche Saint-Louis, Paris, France
| | - Genevieve E Martin
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Ulrike Gerdemann
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rose Al Abosy
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dawn E Comstock
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Sarah A Weiss
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - David Wolski
- Division of Gastroenterology, Liver Center, Massachusetts General Hospital, Boston, MA, USA
| | - Damien C Tully
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.,Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Raymond T Chung
- Division of Gastroenterology, Liver Center, Massachusetts General Hospital, Boston, MA, USA
| | - Todd M Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Arthur Y Kim
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Sarah Fidler
- Division of Medicine, Wright Fleming Institute, Imperial College, London, UK.,Imperial College National Institute for Health Research Biomedical Research Centre, London, UK
| | - Julie Fox
- Department of Genitourinary Medicine and Infectious Disease, Guy's and St Thomas' NHS Foundation Trust, London, UK.,King's College National Institute for Health Research Biomedical Research Centre, London, UK
| | - John Frater
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Oxford National Institute for Health Research Biomedical Research Centre, Oxford, UK
| | - Georg M Lauer
- Division of Gastroenterology, Liver Center, Massachusetts General Hospital, Boston, MA, USA
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Merck Research Laboratories, Boston, MA, USA.
| | - Debattama R Sen
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
21
|
Martin GE, Sen DR, Pace M, Robinson N, Meyerowitz J, Adland E, Thornhill JP, Jones M, Ogbe A, Parolini L, Olejniczak N, Zacharopoulou P, Brown H, Willberg CB, Nwokolo N, Fox J, Fidler S, Haining WN, Frater J. Epigenetic Features of HIV-Induced T-Cell Exhaustion Persist Despite Early Antiretroviral Therapy. Front Immunol 2021; 12:647688. [PMID: 34149690 PMCID: PMC8213372 DOI: 10.3389/fimmu.2021.647688] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/09/2021] [Indexed: 01/03/2023] Open
Abstract
T cell dysfunction occurs early following HIV infection, impacting the emergence of non-AIDS morbidities and limiting curative efforts. ART initiated during primary HIV infection (PHI) can reverse this dysfunction, but the extent of recovery is unknown. We studied 66 HIV-infected individuals treated from early PHI with up to three years of ART. Compared with HIV-uninfected controls, CD4 and CD8 T cells from early HIV infection were characterised by T cell activation and increased expression of the immune checkpoint receptors (ICRs) PD1, Tim-3 and TIGIT. Three years of ART lead to partial – but not complete – normalisation of ICR expression, the dynamics of which varied for individual ICRs. For HIV-specific cells, epigenetic profiling of tetramer-sorted CD8 T cells revealed that epigenetic features of exhaustion typically seen in chronic HIV infection were already present early in PHI, and that ART initiation during PHI resulted in only a partial shift of the epigenome to one with more favourable memory characteristics. These findings suggest that although ART initiation during PHI results in significant immune reconstitution, there may be only partial resolution of HIV-related phenotypic and epigenetic changes.
Collapse
Affiliation(s)
- Genevieve E Martin
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| | - Debattama R Sen
- Department of Immunology, Harvard Medical School, Boston, MA, United States.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Matthew Pace
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicola Robinson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jodi Meyerowitz
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Emily Adland
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - John P Thornhill
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,Division of Medicine, Wright Fleming Institute, Imperial College, London, United Kingdom
| | - Mathew Jones
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ane Ogbe
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lucia Parolini
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Natalia Olejniczak
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Panagiota Zacharopoulou
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Helen Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Christian B Willberg
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,Oxford National Institute of Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Nneka Nwokolo
- Chelsea and Westminster Hospital, London, United Kingdom
| | - Julie Fox
- Department of Genitourinary Medicine and Infectious Disease, Guys and St Thomas' National Health Service (NHS) Trust, London, United Kingdom.,King's College National Institute of Health Research (NIHR) Biomedical Research Centre, London, United Kingdom
| | - Sarah Fidler
- Division of Medicine, Wright Fleming Institute, Imperial College, London, United Kingdom.,Imperial College NIHR Biomedical Research Centre, London, United Kingdom
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Discovery Oncology and Immunology, Merck Research Laboratories, Boston, MA, United States
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,Oxford National Institute of Health Research Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
22
|
Launching a multidisciplinary European collaboration towards a cure for HIV: The EU2Cure Consortium. J Virus Erad 2021; 7:100045. [PMID: 34141442 PMCID: PMC8184646 DOI: 10.1016/j.jve.2021.100045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 11/23/2022] Open
Abstract
We felt the urgency to launch the EU2Cure Consortium to support research and find a cure for the human immunodeficiency virus (HIV) infection through intensified collaboration within Europe. This consortium is open to stakeholders on cure in Europe from academia and the community to connect. The aim of this consortium is to intensify the research collaboration amongst European HIV cure groups and the community and facilitate interactions with other academic and community cure consortia, private parties, and policy makers. Our main aim is to create a European research agenda, data sharing, and development of best practice for clinical and translational science to achieve breakthroughs with clinically feasible HIV cure strategies. This consortium should also enable setting up collaborative studies accessible to a broader group of people living with HIV. Besides reservoir studies, we have identified three overlapping scientific interests in the consortium that provide a starting point for further research within a European network: developing “shock and kill” cure strategies, defining HIV cure biomarkers, and connecting cure cohorts. This strategy should aid stakeholders to sustain progress in HIV cure research regardless of coincidental global health or political crises.
Collapse
|
23
|
Dunn K, Rogers R, Simonson RB, Luo D, Sheng S, Kassam PT, Seyedkazemi S, Hardy H. Rapid initiation of darunavir/cobicistat/emtricitabine/tenofovir alafenamide in acute and early HIV-1 infection: a DIAMOND subgroup analysis. HIV Res Clin Pract 2021; 22:55-61. [PMID: 33999786 DOI: 10.1080/25787489.2021.1915652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Treatment during acute or early human immunodeficiency virus (HIV)-1 infection is associated with immunologic and virologic benefits. OBJECTIVE To evaluate darunavir/cobicistat/emtricitabine/tenofovir alafenamide (D/C/F/TAF) efficacy/safety among patients with acute or early HIV-1 infection who rapidly initiate treatment. METHODS DIAMOND (ClinicalTrials.gov Identifier: NCT03227861), a phase 3 study, evaluated the efficacy/safety of D/C/F/TAF 800/150/200/10 mg in rapid initiation. Adults aged ≥18 years began D/C/F/TAF within 14 days of diagnosis, prior to the availability of screening/baseline laboratory results. In this subgroup analysis, virologic response (HIV-1 RNA <50 copies/mL) was assessed at Week 48 by intent-to-treat FDA snapshot (ITT-FDA snapshot) and observed (excluding patients with missing data) analyses in patients with acute (HIV-1 antibody negative and HIV-1 RNA positive/p24 positive) or early (HIV-1 antibody positive and suspected infection ≤6 months before screening/baseline) infection. RESULTS Among 109 patients, 13 had acute and 43 had early HIV-1 infection. High rates of virologic response were demonstrated at Week 48 by ITT-FDA snapshot (acute: 10/13 [76.9%]; early: 37/43 [86.0%]) and observed (acute: 10/11 [90.9%]; early: 37/38 [97.4%]) analyses. No patients discontinued or required regimen change due to baseline resistance or lack of efficacy, or developed protocol-defined virologic failure. Through Week 48, 7 (53.8%) acute and 22 (51.2%) early infection patients had a D/C/F/TAF-related adverse event (AE); none had a D/C/F/TAF-related grade 4 or serious AE. CONCLUSIONS High rates of viral suppression during acute/early infection were achieved with D/C/F/TAF rapid initiation, no treatment-emergent resistant mutations were observed, and D/C/F/TAF was safe and well tolerated.
Collapse
Affiliation(s)
- Keith Dunn
- Janssen Scientific Affairs, LLC, Titusville, NJ, USA
| | - Rachel Rogers
- Janssen Scientific Affairs, LLC, Titusville, NJ, USA
| | | | - Donghan Luo
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Shubin Sheng
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | | | | | - Hélène Hardy
- Janssen Research & Development, LLC, Titusville, NJ, USA
| |
Collapse
|
24
|
Treatment of primary HIV infection: new data for a new era. AIDS 2020; 34:1855-1856. [PMID: 32889855 DOI: 10.1097/qad.0000000000002635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Abstract
PURPOSE OF REVIEW Although cities present opportunities for infectious pathogens such as HIV to spread, public health infrastructure within these cities also provides opportunities to design effective approaches to eliminate transmission of these pathogens. The HIV Transmission Elimination AMsterdam (H-TEAM) Initiative, a consortium of relevant stakeholders involved in HIV prevention and care, designed an integrated approach to curb the HIV epidemic in Amsterdam, including providing preexposure prophylaxis (PrEP), increasing awareness of acute HIV infection, offering same-day test and treat, and improving indicator disease-driven HIV testing. RECENT FINDINGS In 2013, approximately 230 people in Amsterdam were newly diagnosed with HIV, largely belonging to one of two key affected populations, namely MSM and people with a migration background. Since the start of H-TEAM in 2014, a decrease in new diagnoses was observed (130 in 2017), with an increasing proportion of MSM who had been diagnosed with a recent infection. SUMMARY The H-TEAM shows that a city-based concerted effort is feasible. However, major challenges remain, such as reducing the number of late HIV diagnoses, and identifying and providing appropriate services to a diminishing group of individuals who are likely the source of transmission.
Collapse
|
26
|
Taramasso L, Fabbiani M, Nozza S, De Benedetto I, Bruzzesi E, Mastrangelo A, Pinnetti C, Calcagno A, Ferrara M, Bozzi G, Focà E, Quiros-Roldan E, Ripamonti D, Campus M, Celesia BM, Torti C, Cosco L, Di Biagio A, Rusconi S, Marchetti G, Mussini C, Gulminetti R, Cingolani A, d'Ettorre G, Madeddu G, Franco A, Orofino G, Squillace N, Muscatello A, Gori A, Antinori A, Tambussi G, Bandera A. Predictors of incomplete viral response and virologic failure in patients with acute and early HIV infection. Results of Italian Network of ACuTe HIV InfectiON (INACTION) cohort. HIV Med 2020; 21:523-535. [PMID: 32578947 DOI: 10.1111/hiv.12885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the factors that can influence an incomplete viral response (IVR) after acute and early HIV infection (AEHI). METHODS This was a retrospective, observational study including patients with AEHI (Fiebig stages I-V) diagnosed between January 2008 and December 2014 at 20 Italian centres. IVR was defined by: (1) viral blip (51-1000 HIV-1 RNA copies/mL after achievement of < 50 HIV-1 RNA copies/mL); (2) virologic failure [> 1000 copies/mL after achievement of < 200 copies/mL, or ≥ 200 copies/mL after 24 weeks on an antiretroviral therapy (ART)]; (3) suboptimal viral response (> 50 copies/mL after 48 weeks on ART or two consecutive HIV-1 RNA levels with ascending trend during ART). Cox regression analysis was used to calculate the hazard ratios (HRs) and 95% confidence intervals (95% CIs) for IVR. RESULTS In all, 263 patients were studied, 227 (86%) males, with a median [interquartile range (IQR)] age of 38 (30-46) years. During a median follow-up of 13.0 (5.7-31.1) months, 38 (14.4%) had IVR. The presence of central nervous system (CNS) symptoms was linked to a higher risk of IVR (HR = 4.70, 95% CI: 1.56-14.17), while a higher CD4/CD8 cell count ratio (HR = 0.13, 95% CI: 0.03-0.51 for each point increase) and first-line ART with three-drug regimens recommended by current guidelines (HR = 0.40, 95% CI: 0.18-0.91 compared with other regimens including four or five drugs, older drugs or non-standard backbones) were protective against IVR. CONCLUSIONS Patients with lower CD4/CD8 ratio and CNS symptoms could be at a higher risk of IVR after AEHI. The use of recommended ART may be relevant for improving short-term viral efficacy in this group of patients.
Collapse
Affiliation(s)
- L Taramasso
- Infectious Diseases Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda, Policlinico Maggiore Hospital, Milan, Italy
| | - M Fabbiani
- Infectious Diseases Unit, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - S Nozza
- Clinic of Infectious Diseases, San Raffaele Hospital, University Vita Salute, Milan, Italy
| | - I De Benedetto
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, Turin, Italy
| | - E Bruzzesi
- Clinic of Infectious Diseases, San Raffaele Hospital, University Vita Salute, Milan, Italy
| | - A Mastrangelo
- Clinic of Infectious Diseases, San Raffaele Hospital, University Vita Salute, Milan, Italy
| | - C Pinnetti
- National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - A Calcagno
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, Turin, Italy
| | - M Ferrara
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, Turin, Italy
| | - G Bozzi
- Infectious Diseases Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda, Policlinico Maggiore Hospital, Milan, Italy
| | - E Focà
- Division of Infectious and Tropical Diseases, ASST Spedali Civili Hospital, University of Brescia, Brescia, Italy
| | - E Quiros-Roldan
- Division of Infectious and Tropical Diseases, ASST Spedali Civili Hospital, University of Brescia, Brescia, Italy
| | - D Ripamonti
- Infectious Disease Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - M Campus
- Infectious Diseases Unit, SS Trinità Hospital, ASSL Cagliari, Cagliari, Italy
| | - B M Celesia
- Unit of Infectious Diseases, Garibaldi Hospital, Catania, Italy
| | - C Torti
- Unit of Infectious Diseases, Department of Medical and Surgical Sciences, University "Magna Graecia", Catanzaro, Italy
| | - L Cosco
- Infectious Diseases Unit, "Pugliese-Ciaccio" Hospital, Catanzaro, Italy
| | - A Di Biagio
- Department of Infectious Diseases, Policlinico San Martino Hospital, Genoa, Italy
| | - S Rusconi
- Infectious Diseases Unit, Department of Biomedical and Clinical Sciences, "Luigi Sacco" Hospital, University of Milan, Milan, Italy
| | - G Marchetti
- Clinic of Infectious Diseases, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milan, Italy
| | - C Mussini
- Clinic of Infectious Diseases, Modena Hospital, University of Modena and Reggio Emilia, Modena, Italy
| | - R Gulminetti
- Infectious Diseases Unit, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - A Cingolani
- Institute of Clinical Infectious Diseases, Agostino Gemelli Hospital, Catholic University of Sacred Heart, Rome, Italy
| | - G d'Ettorre
- Infectious Diseases Unit, Umberto I Hospital, La Sapienza University, Rome, Italy
| | - G Madeddu
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - A Franco
- Infectious Diseases Unit, ASP Siracusa, Siracusa, Italy
| | - G Orofino
- Unit of Infectious Diseases, Divisione A, Amedeo di Savoia Hospital, Turin, Italy
| | - N Squillace
- Infectious Diseases Unit, Department of Internal Medicine, ASST San Gerardo, Monza, Italy.,University of Milano-Bicocca, Milan, Italy
| | - A Muscatello
- Infectious Diseases Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda, Policlinico Maggiore Hospital, Milan, Italy
| | - A Gori
- Infectious Diseases Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda, Policlinico Maggiore Hospital, Milan, Italy.,School of Medicine and Surgery, University of Milan, Milan, Italy
| | - A Antinori
- National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - G Tambussi
- Clinic of Infectious Diseases, San Raffaele Hospital, University Vita Salute, Milan, Italy
| | - A Bandera
- Infectious Diseases Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda, Policlinico Maggiore Hospital, Milan, Italy.,School of Medicine and Surgery, University of Milan, Milan, Italy
| | | |
Collapse
|
27
|
Abstract
Therapeutic approaches towards a functional cure or eradication of HIV have gained renewed momentum upon encouraging data emerging from studies in SIV monkey models and recent results from human clinical studies. However, a multitude of questions remain to be addressed, including how to deal with the latent viral reservoir, how to boost the host immune response to the virus and what the hurdles are to reach relevant viral compartments in the body. Advances have been made especially with regard to identifying agents that can reactivate the latent virus in vivo and boost the cellular and humoral immunity, but it remains largely unclear whether any of these strategies can awaken a sufficiently large fraction of the viral reservoir and whether the boosted immunity can prevent rapid viral replication once antiretroviral treatments are stopped.
Collapse
Affiliation(s)
- Lucia Bailon
- Fundació Lluita contra la Sida, Infectious Disease Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Beatriz Mothe
- Fundació Lluita contra la Sida, Infectious Disease Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain
| | | | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.
- Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.
- AELIX Therapeutics, Barcelona, Spain.
- ICREA, Pg. Lluis Companys 23, Barcelona, Spain.
| |
Collapse
|
28
|
Boyd MA, Boffito M, Castagna A, Estrada V. Rapid initiation of antiretroviral therapy at HIV diagnosis: definition, process, knowledge gaps. HIV Med 2020; 20 Suppl 1:3-11. [PMID: 30724450 DOI: 10.1111/hiv.12708] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2018] [Indexed: 01/14/2023]
Abstract
Initiating antiretroviral therapy (ART) as early as the day of HIV diagnosis is a strategy of increasing global interest to control the HIV epidemic and optimize the health of people living with HIV (PLWH). No detrimental effects of rapid-start ART have been identified in randomized controlled trials undertaken in low- or middle-income countries, or in cohort studies performed in high-income countries. Rapid-start ART may be a key approach in reaching the 2020 Joint United Nations Programme on HIV/AIDS goal of 90% of all PLWH knowing their status, 90% of those diagnosed receiving sustained ART, and 90% of those receiving ART achieving viral suppression; it may also be important for achieving the suggested fourth "90%" goal: improving health-related quality-of-life in PLWH. Presently there is insufficient broad evidence for guidelines to recommend universal test-and-treat strategies for all people, in all settings, at HIV diagnosis; consequently, there is a pressing need to conduct high-quality studies that investigate immediate ART initiation. This article evaluates global evidence regarding rapid-start ART, including same-day start, with particular focus on the implementation of this strategy in high-income countries.
Collapse
Affiliation(s)
- M A Boyd
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - M Boffito
- Chelsea and Westminster Hospital, London, UK.,Imperial College London, London, UK
| | - A Castagna
- Clinic of Infectious Diseases, San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy
| | - V Estrada
- Hospital Clinico San Carlos, Universidad Complutense, Madrid, Spain
| |
Collapse
|
29
|
Muscatello A, Nozza S, Fabbiani M, De Benedetto I, Ripa M, Dell'acqua R, Antinori A, Pinnetti C, Calcagno A, Ferrara M, Focà E, Quiros-Roldan E, Ripamonti D, Campus M, Maurizio Celesia B, Torti C, Cosco L, Di Biagio A, Rusconi S, Marchetti G, Mussini C, Gulminetti R, Cingolani A, D'ettorre G, Madeddu G, Franco A, Orofino G, Squillace N, Gori A, Tambussi G, Bandera A. Enhanced Immunological Recovery With Early Start of Antiretroviral Therapy During Acute or Early HIV Infection-Results of Italian Network of ACuTe HIV InfectiON (INACTION) Retrospective Study. Pathog Immun 2020; 5:8-33. [PMID: 32258852 PMCID: PMC7104556 DOI: 10.20411/pai.v5i1.341] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/20/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Viral load peak and immune activation occur shortly after exposure during acute or early HIV infection (AEHI). We aimed to define the benefit of early start of antiretroviral treatment (ART) during AEHI in terms of immunological recovery, virological suppression, and treatment discontinuation. SETTING Patients diagnosed with AEHI (Fiebig stages I-V) during 2008-2014 from an analysis of 20 Italian centers. METHODS This was an observational, retrospective, and multicenter study. We investigated the effect of early ART (defined as initiation within 3 months from AEHI diagnosis) on time to virological suppression, optimal immunological recovery (defined as CD4 count ≥500/µL, CD4 ≥30%, and CD4/CD8 ≥1), and first-line ART regimen discontinuation by Cox regression analysis. RESULTS There were 321 patients with AEHI included in the study (82.9% in Fiebig stage III-V). At diagnosis, the median viral load was 5.67 log10 copies/mL and the median CD4 count was 456 cells/µL. Overall, 70.6% of patients started early ART (median time from HIV diagnosis to ART initiation 12 days, IQR 6-27). Higher baseline viral load and AEHI diagnosis during 2012-2014 were independently associated with early ART. HBV co-infection, baseline CD4/CD8 ≥1, lower baseline HIV-RNA, and AEHI diagnosis in recent years (2012-2014) were independently associated with a shorter time to virological suppression. Early ART emerged as an independent predictor of optimal immunological recovery after adjustment for baseline CD4 (absolute and percentage count) and CD4/CD8 ratio. The only independent predictor of first-line ART discontinuation was an initial ART regimen including > 3 drugs. CONCLUSIONS In a large cohort of well-characterized patients with AEHI, we confirmed the beneficial role of early ART on CD4+ T-cell recovery and on rates of CD4/CD8 ratio normalization. Moreover, we recognized baseline CD4/CD8 ratio as an independent factor influencing time to virological response in the setting of AEHI, thus giving new insights into research of immunological markers associated with virological control.
Collapse
Affiliation(s)
- Antonio Muscatello
- Infectious Diseases Unit; Department of Internal Medicine; IRCCS Ca' Granda Foundation Maggiore Hospital; Milan, Italy
| | - Silvia Nozza
- Clinic of Infectious Diseases; San Raffaele Hospital; University Vita Salute; Milan, Italy
| | - Massimiliano Fabbiani
- Infectious and Tropical Diseases Unit; Azienda Ospedaliero-Universitaria Senese; Siena, Italy
| | - Ilaria De Benedetto
- Department of Medical Sciences; Unit of Infectious Diseases; University of Turin; Amedeo di Savoia Hospital; Turin, Italy
| | - Marco Ripa
- Clinic of Infectious Diseases; San Raffaele Hospital; University Vita Salute; Milan, Italy
| | - Raffaele Dell'acqua
- Clinic of Infectious Diseases; San Raffaele Hospital; University Vita Salute; Milan, Italy
| | - Andrea Antinori
- National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS; Rome, Italy
| | - Carmela Pinnetti
- National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS; Rome, Italy
| | - Andrea Calcagno
- Department of Medical Sciences; Unit of Infectious Diseases; University of Turin; Amedeo di Savoia Hospital; Turin, Italy
| | - Micol Ferrara
- Department of Medical Sciences; Unit of Infectious Diseases; University of Turin; Amedeo di Savoia Hospital; Turin, Italy
| | - Emanuele Focà
- Division of Infectious and Tropical Diseases; University of Brescia; ASST Spedali Civili Hospital; Brescia, Italy
| | - Eugenia Quiros-Roldan
- Division of Infectious and Tropical Diseases; University of Brescia; ASST Spedali Civili Hospital; Brescia, Italy
| | - Diego Ripamonti
- Infectious Disease Unit; ASST Papa Giovanni XXIII; Bergamo, Italy
| | - Marco Campus
- Infectious Diseases Unit; SS Trinità Hospital; ASSL Cagliari, Italy
| | | | - Carlo Torti
- Unit of Infectious Diseases; Department of Medical and Surgical Sciences; University “Magna Graecia;” Catanzaro, Italy
| | - Lucio Cosco
- Infectious Diseases Unit; “Pugliese-Ciaccio” Hospital; Catanzaro, Italy
| | - Antonio Di Biagio
- Department of Infectious Diseases; IRCCS AOU San Martino IST; (DISSAL); University of Genoa; Genoa, Italy
| | - Stefano Rusconi
- Infectious Diseases Unit; Department of Biomedical and Clinical Sciences “Luigi Sacco” Hospital; University of Milan, Italy
| | - Giulia Marchetti
- Clinic of Infectious Diseases; Department of Health Sciences; University of Milan; ASST Santi Paolo e Carlo; Milan, Italy
| | - Cristina Mussini
- Clinic of Infectious Diseases; University of Modena and Reggio Emilia; Modena Hospital; Italy
| | - Roberto Gulminetti
- Department of Medical Sciences; Unit of Infectious Diseases; University of Turin; Amedeo di Savoia Hospital; Turin, Italy
| | - Antonella Cingolani
- Institute of Clinical Infectious Diseases; Agostino Gemelli Hospital; Catholic University of Sacred Heart; Rome, Italy
| | - Gabriella D'ettorre
- Infectious Diseases Unit; Umberto I Hospital; La Sapienza University; Rome, Italy
| | - Giordano Madeddu
- Department of Clinical and Experimental Medicine; Unit of Infectious Diseases; University of Sassari, Italy
| | | | - Giancarlo Orofino
- Unit of Infectious Diseases; Divisione A; Amedeo di Savoia Hospital; Turin, Italy
| | - Nicola Squillace
- Infectious Diseases Unit; Department of Internal Medicine; ASST San Gerardo; Monza, Italy, University of Milano-Bicocca; Milan, Italy
| | - Andrea Gori
- Infectious Diseases Unit; Department of Internal Medicine; IRCCS Ca' Granda Foundation Maggiore Hospital; Milan, Italy
- School of Medicine and Surger; University of Milan, Italy
| | - Giuseppe Tambussi
- Clinic of Infectious Diseases; San Raffaele Hospital; University Vita Salute; Milan, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit; Department of Internal Medicine; IRCCS Ca' Granda Foundation Maggiore Hospital; Milan, Italy
- School of Medicine and Surger; University of Milan, Italy
| | - On Behalf Of Inaction Study Group.
- Infectious Diseases Unit; Department of Internal Medicine; IRCCS Ca' Granda Foundation Maggiore Hospital; Milan, Italy
- Clinic of Infectious Diseases; San Raffaele Hospital; University Vita Salute; Milan, Italy
- Infectious and Tropical Diseases Unit; Azienda Ospedaliero-Universitaria Senese; Siena, Italy
- Department of Medical Sciences; Unit of Infectious Diseases; University of Turin; Amedeo di Savoia Hospital; Turin, Italy
- National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS; Rome, Italy
- Division of Infectious and Tropical Diseases; University of Brescia; ASST Spedali Civili Hospital; Brescia, Italy
- Infectious Disease Unit; ASST Papa Giovanni XXIII; Bergamo, Italy
- Infectious Diseases Unit; SS Trinità Hospital; ASSL Cagliari, Italy
- Unit of Infectious Diseases; Garibaldi Hospital; Catania, Italy
- Unit of Infectious Diseases; Department of Medical and Surgical Sciences; University “Magna Graecia;” Catanzaro, Italy
- Infectious Diseases Unit; “Pugliese-Ciaccio” Hospital; Catanzaro, Italy
- Department of Infectious Diseases; IRCCS AOU San Martino IST; (DISSAL); University of Genoa; Genoa, Italy
- Infectious Diseases Unit; Department of Biomedical and Clinical Sciences “Luigi Sacco” Hospital; University of Milan, Italy
- Clinic of Infectious Diseases; Department of Health Sciences; University of Milan; ASST Santi Paolo e Carlo; Milan, Italy
- Clinic of Infectious Diseases; University of Modena and Reggio Emilia; Modena Hospital; Italy
- Institute of Clinical Infectious Diseases; Agostino Gemelli Hospital; Catholic University of Sacred Heart; Rome, Italy
- Infectious Diseases Unit; Umberto I Hospital; La Sapienza University; Rome, Italy
- Department of Clinical and Experimental Medicine; Unit of Infectious Diseases; University of Sassari, Italy
- Infectious Diseases Unit; ASP Siracusa, Italy
- Unit of Infectious Diseases; Divisione A; Amedeo di Savoia Hospital; Turin, Italy
- Infectious Diseases Unit; Department of Internal Medicine; ASST San Gerardo; Monza, Italy, University of Milano-Bicocca; Milan, Italy
- School of Medicine and Surger; University of Milan, Italy
| |
Collapse
|
30
|
Estimated dates of detectable infection (EDDIs) as an improvement upon Fiebig staging for HIV infection dating. Epidemiol Infect 2020; 148:e53. [PMID: 32070438 PMCID: PMC7078584 DOI: 10.1017/s0950268820000503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Accurate methods for determining the duration of HIV infection at the individual level are valuable in many settings, including many critical research studies and in clinical practice (especially for acute infection). Since first published in 2003, the ‘Fiebig staging system’ has been used as the primary way of classifying early HIV infection into five sequential stages based on HIV test result patterns in newly diagnosed individuals. However, Fiebig stages can only be assigned to individuals who produce both a negative and a positive test result on the same day, on specific pairs of tests of varying ‘sensitivity’. Further, in the past 16 years HIV-testing technology has evolved substantially, and three of the five key assays used to define Fiebig stages are no longer widely used. To address these limitations, we developed an improved and more general framework for estimating the duration of HIV infection by interpreting any combination of diagnostic test results, whether obtained on single or multiple days, into an estimated date of detectable infection, or EDDI. A key advantage of the EDDI method over Fiebig staging is that it allows for the generation of a point estimate, as well as an associated credibility interval for the date of first detectable infection, for any person who has at least one positive and one negative HIV test of any kind. The tests do not have to be run on the same day; they do not have to be run during the acute phase of infection and the method does not rely on any special pairing of tests to define ‘stages’ of infection. The size of the interval surrounding the EDDI (and therefore the precision of the estimate itself) depends largely on the length of time between negative and positive tests. The EDDI approach is also flexible, seamlessly incorporating any assay for which there is a reasonable diagnostic delay estimate. An open-source, free online tool includes a user-updatable curated database of published diagnostic delays. HIV diagnostics have evolved tremendously since that original publication more than 15 years ago, and it is time to similarly evolve the methods used to estimate timing of infection. The EDDI method is a flexible and rigorous way to estimate the timing of HIV infection in a continuously evolving diagnostic landscape.
Collapse
|
31
|
Eriksen J, Carlander C, Albert J, Flamholc L, Gisslén M, Navér L, Svedhem V, Yilmaz A, Sönnerborg A. Antiretroviral treatment for HIV infection: Swedish recommendations 2019. Infect Dis (Lond) 2020; 52:295-329. [PMID: 31928282 DOI: 10.1080/23744235.2019.1707867] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The Swedish Reference Group for Antiviral Therapy (RAV) published recommendations for the treatment of HIV infection in this journal most recently in 2017. An expert group under the guidance of RAV here provides updated recommendations. The most important updates in the present guidelines are the following: (a) The risk of HIV transmission through condomless sex from individuals with fully suppressed HIV viral load is effectively zero. (b) Pre-exposure prophylaxis (PrEP) is recommended for groups with a high risk of HIV infection. (c) Since the last update, two new substances have been registered: bictegravir and doravirine. (d) Dual treatment may be an alternative in selected patients, using lamivudine + dolutegravir or lamivudine + boosted darunavir/atazanavir. As with previous publications, recommendations are evidence-graded in accordance with the Oxford Centre for Evidence Based Medicine. This document does not cover treatment of opportunistic infections and tumours.
Collapse
Affiliation(s)
- Jaran Eriksen
- Unit of Infectious Diseases/Venhälsan, Södersjukhuset, Stockholm, Sweden.,Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Christina Carlander
- Department of Infectious Diseases, Västmanland County Hospital, Västerås, Sweden.,Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jan Albert
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Leo Flamholc
- Department of Infectious Diseases, Skåne University Hospital, Malmö, Sweden
| | - Magnus Gisslén
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lars Navér
- Division of Paediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden.,Department of Paediatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Veronica Svedhem
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Aylin Yilmaz
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Sönnerborg
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden.,Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
32
|
Patel V, Spouge JL. Estimating the basic reproduction number of a pathogen in a single host when only a single founder successfully infects. PLoS One 2020; 15:e0227127. [PMID: 31923263 PMCID: PMC6953795 DOI: 10.1371/journal.pone.0227127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 12/12/2019] [Indexed: 11/27/2022] Open
Abstract
If viruses or other pathogens infect a single host, the outcome of infection may depend on the initial basic reproduction number R0, the expected number of host cells infected by a single infected cell. This article shows that sometimes, phylogenetic models can estimate the initial R0, using only sequences sampled from the pathogenic population during its exponential growth or shortly thereafter. When evaluated by simulations mimicking the bursting viral reproduction of HIV and simultaneous sampling of HIV gp120 sequences during early viremia, the estimated R0 displayed useful accuracies in achievable experimental designs. Estimates of R0 have several potential applications to investigators interested in the progress of infection in single hosts, including: (1) timing a pathogen’s movement through different microenvironments; (2) timing the change points in a pathogen’s mode of spread (e.g., timing the change from cell-free spread to cell-to-cell spread, or vice versa, in an HIV infection); (3) quantifying the impact different initial microenvironments have on pathogens (e.g., in mucosal challenge with HIV, quantifying the impact that the presence or absence of mucosal infection has on R0); (4) quantifying subtle changes in infectability in therapeutic trials (either human or animal), even when therapies do not produce total sterilizing immunity; and (5) providing a variable predictive of the clinical efficacy of prophylactic therapies.
Collapse
Affiliation(s)
- Vruj Patel
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John L. Spouge
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
33
|
Nourmohammad A, Otwinowski J, Łuksza M, Mora T, Walczak AM. Fierce Selection and Interference in B-Cell Repertoire Response to Chronic HIV-1. Mol Biol Evol 2020; 36:2184-2194. [PMID: 31209469 PMCID: PMC6759071 DOI: 10.1093/molbev/msz143] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
During chronic infection, HIV-1 engages in a rapid coevolutionary arms race with the host's adaptive immune system. While it is clear that HIV exerts strong selection on the adaptive immune system, the characteristics of the somatic evolution that shape the immune response are still unknown. Traditional population genetics methods fail to distinguish chronic immune response from healthy repertoire evolution. Here, we infer the evolutionary modes of B-cell repertoires and identify complex dynamics with a constant production of better B-cell receptor (BCR) mutants that compete, maintaining large clonal diversity and potentially slowing down adaptation. A substantial fraction of mutations that rise to high frequencies in pathogen-engaging CDRs of BCRs are beneficial, in contrast to many such changes in structurally relevant frameworks that are deleterious and circulate by hitchhiking. We identify a pattern where BCRs in patients who experience larger viral expansions undergo stronger selection with a rapid turnover of beneficial mutations due to clonal interference in their CDR3 regions. Using population genetics modeling, we show that the extinction of these beneficial mutations can be attributed to the rise of competing beneficial alleles and clonal interference. The picture is of a dynamic repertoire, where better clones may be outcompeted by new mutants before they fix.
Collapse
Affiliation(s)
- Armita Nourmohammad
- Max Planck Institute for Dynamics and Self-organization, Göttingen, Germany.,Department of Physics, University of Washington, Seattle, WA
| | - Jakub Otwinowski
- Max Planck Institute for Dynamics and Self-organization, Göttingen, Germany
| | - Marta Łuksza
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Thierry Mora
- Laboratoire de Physique Statistique, CNRS, Sorbonne University, Paris-Diderot University, École Normale Supérieure (PSL), Paris, France
| | - Aleksandra M Walczak
- Laboratoire de Physique Théorique, CNRS, Sorbonne University, École Normale Supérieure (PSL), Paris, France
| |
Collapse
|
34
|
Jongen VW, van Santen DK, Alberts CJ, Schim van der Loeff MF. Estimating incidence rates of grouped HPV types: A systematic review and comparison of the impact of different epidemiological assumptions. PAPILLOMAVIRUS RESEARCH (AMSTERDAM, NETHERLANDS) 2019; 8:100187. [PMID: 31600572 PMCID: PMC6804437 DOI: 10.1016/j.pvr.2019.100187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 10/04/2019] [Accepted: 10/06/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Some studies on human papillomavirus (HPV) provide not only type-specific incidence rates (IR), but also IRs of HPV groupings (e.g. the nonavalent grouping). We made an inventory of the different approaches used to calculate such IRs and assessed their impact on the estimated IRs of HPV groupings. METHODS We performed a systematic review assessing all approaches used in literature to estimate IRs. Subsequently we applied these approaches to data of a Dutch cohort study on HPV in men who have sex with men (H2M). IRs were estimated for six different HPV groupings. RESULTS The systematic review yielded six different approaches (A-F) for estimating the IRs, varying in exclusion criteria at baseline, and the definitions of an incident event and person-time. Applying these approaches to the H2M dataset (n = 749), we found differences in the number of participants at risk, number of incidents events, person-time, and IR. For example, for the nonavalent grouping, depending on the approach chosen, the IR varied between 3.09 and 6.54 per 100 person-months. CONCLUSION In published studies different epidemiological assumptions are used to estimate IRs of grouped HPV types, leading to widely differing estimates of IRs. IRs between different studies may therefore not be comparable.
Collapse
Affiliation(s)
- Vita W Jongen
- Department of Infectious Diseases, Public Health Service Amsterdam, Amsterdam, the Netherlands
| | - Daniëla K van Santen
- Department of Infectious Diseases, Public Health Service Amsterdam, Amsterdam, the Netherlands
| | - Catharina J Alberts
- Department of Infectious Diseases, Public Health Service Amsterdam, Amsterdam, the Netherlands; International Agency for Research on Cancer, Lyon, France
| | - Maarten F Schim van der Loeff
- Department of Infectious Diseases, Public Health Service Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, Univ of Amsterdam, Internal Medicine, Amsterdam Infection & Immunity Institute (AIII), Amsterdam, Netherlands.
| |
Collapse
|
35
|
Time to viral rebound and safety after antiretroviral treatment interruption in postpartum women compared with men. AIDS 2019; 33:2149-2156. [PMID: 31373919 DOI: 10.1097/qad.0000000000002334] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE(S) The short-term safety of treatment interruptions, a necessary part of cure studies, is not well established, particularly in women. We explored viral rebound kinetics and safety in a group of postpartum women discontinuing ART and compared results to men in historical interruption trials. DESIGN Prospective evaluation of time to virologic rebound. METHODS One thousand and seventy-six asymptomatic, virally suppressed, postpartum women living with HIV enrolled in the PROMISE trial with baseline CD4 cell counts at least 350 cells/μl underwent antiretroviral treatment (ART) discontinuation. Proportion with virologic suppression at weeks 4 and 12 were compared with participants in ACTG treatment interruption trials (91% male population). RESULTS In PROMISE, using interval censored methods, the estimated median time to HIV viral rebound was 2 weeks. An estimated 6% of women would remain virally suppressed at 30 weeks. Of those who had viral rebound by 30 weeks (N = 993), less than 4% experienced grade 3 or higher laboratory events, and 1% experienced WHO stage 2 or higher clinical events. Overall, less than 1% of participants progressed from WHO Stage 1 to Stage 2 or higher after discontinuation of ART, and 3.9% experienced a decline in CD4 cell count to less than 350 cells/μl or local treatment guidelines. A significantly higher proportion of women in PROMISE (25.4%) were virologically suppressed (<400 copies/ml) at 12 weeks compared with ACTG NWCS 371 participants (6.4%). CONCLUSION Temporary treatment interruptions in healthy, HIV-infected women with high CD4 cell counts can be well tolerated. Potential sex differences need to be considered in cure studies examining time to virologic rebound.
Collapse
|
36
|
Novelli S, Lécuroux C, Avettand-Fenoel V, Seng R, Essat A, Morlat P, Viard JP, Rouzioux C, Meyer L, Goujard C. Long-term Therapeutic Impact of the Timing of Antiretroviral Therapy in Patients Diagnosed With Primary Human Immunodeficiency Virus Type 1 Infection. Clin Infect Dis 2019; 66:1519-1527. [PMID: 29211834 DOI: 10.1093/cid/cix1068] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022] Open
Abstract
Background We aimed to determine the consequences of delayed human immunodeficiency virus type 1 (HIV-1) infection diagnosis by comparing long-term outcomes depending on the time of combination antiretroviral therapy (cART) initiation in patients diagnosed during primary HIV infection (PHI). Methods We selected patients from the French National Agency for Research on AIDS and Viral Hepatitis (ANRS) PRIMO cohort, treated for ≥36 months, with sustained HIV RNA <50 copies/mL: 77 treated within 1 month following PHI diagnosis (immediate ART) and 73 treated >12 months after infection (deferred ART). We measured inflammatory biomarkers from PHI through the last visit on cART, and CD4+ and CD8+ T-cell activation and plasma ultrasensitive HIV RNA at the last visit. Inflammation/activation levels were compared with those of uninfected controls. We modeled CD4+ count, CD4:CD8 ratio, and HIV DNA dynamics on cART. Results The decrease of HIV DNA levels was more marked in the immediate than deferred ART group, leading to a sustained mean difference of -0.6 log10 copies/106 peripheral blood mononuclear cells. Immediate ART led to improved CD4+ T-cell counts and CD4:CD8 ratios over the first 4 years of cART. At the last visit (median, 82 months), there was no difference between groups in CD4+ counts, CD4:CD8 ratio, ultrasensitive HIV RNA, or inflammation/activation marker levels. Long-term suppressive cART failed to normalize inflammation levels, which were not associated with immunovirological markers. Conclusions Antiretroviral therapy initiated during PHI promotes long-term reduction of HIV reservoir size. In patients with sustained virologic suppression, inflammation may be driven by non-HIV-related factors.
Collapse
Affiliation(s)
- Sophie Novelli
- National Institute of Health and Medical Research (INSERM), Centre for Research in Epidemiology and Population Health (CESP), Paris-Sud University, France
| | - Camille Lécuroux
- Commissariat à l'énergie atomique et aux énergies alternatives (CEA) -Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases (IMVA), Infectious Disease Models and Innovative Therapie (IDMIT), Institut de biologie François Jacob (IBJF), Direction de la recherche fondamentale (DRF), Fontenay-aux-Roses, France
| | - Véronique Avettand-Fenoel
- Paris Descartes University, EA 7327, Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris (AP-HP), Necker Hospital, Virology Department, France
| | - Rémonie Seng
- INSERM, CESP, U1018, Paris-Sud University, AP-HP, Bicêtre Hospital, Department of Public Health and Epidemiology, Le Kremlin-Bicêtre, France
| | - Asma Essat
- National Institute of Health and Medical Research (INSERM), Centre for Research in Epidemiology and Population Health (CESP), Paris-Sud University, France
| | - Philippe Morlat
- Bordeaux University Hospital, Saint André Hospital, Department of Internal Medicine, France
| | - Jean-Paul Viard
- Paris Descartes University, EA 7327, Sorbonne Paris Cité, AP-HP, Hôtel Dieu Hospital, Unit of Therapeutics in Immunology and Infectiology, France
| | - Christine Rouzioux
- Paris Descartes University, EA 7327, Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris (AP-HP), Necker Hospital, Virology Department, France
| | - Laurence Meyer
- INSERM, CESP, U1018, Paris-Sud University, AP-HP, Bicêtre Hospital, Department of Public Health and Epidemiology, Le Kremlin-Bicêtre, France
| | - Cécile Goujard
- INSERM, CESP, U1018, Paris-Sud University, AP-HP, Bicêtre Hospital, Department of Internal Medicine, Le Kremlin-Bicêtre, France
| |
Collapse
|
37
|
Binda F, Monge E, Simonetti FR, Zanchetta N, Galli M, Milazzo L, Corbellino M, Antinori S. Primary HIV infection in patients with acute hepatitis B: a report of two cases. Antivir Ther 2019; 23:197-200. [PMID: 29022881 DOI: 10.3851/imp3201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2017] [Indexed: 02/07/2023]
Abstract
We describe two patients admitted to our institution with a diagnosis of sexually acquired acute hepatitis B who also had underlying hyper acute HIV infection. Both individuals reported high rates of condomless sex. Antiviral therapy active against HBV and HIV was started within days after diagnosis. Treatment was well tolerated and led to a rapid control of both infections and hepatitis B surface antibody seroconversion. The efficacy and safety of contemporary antiretroviral drug combinations suggest that treatment of acute HIV infection is feasible in patients with acute hepatitis B.
Collapse
Affiliation(s)
- Francesca Binda
- Department of Biomedical and Clinical Sciences 'Luigi Sacco', University of Milan, Milan, Italy
| | - Elisa Monge
- Department of Biomedical and Clinical Sciences 'Luigi Sacco', University of Milan, Milan, Italy
| | | | - Nadia Zanchetta
- Clinical Microbiology, Virology and Bioemergence Diagnostics, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Massimo Galli
- Department of Biomedical and Clinical Sciences 'Luigi Sacco', University of Milan, Milan, Italy.,III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Laura Milazzo
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Mario Corbellino
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Spinello Antinori
- Department of Biomedical and Clinical Sciences 'Luigi Sacco', University of Milan, Milan, Italy.,III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| |
Collapse
|
38
|
Schuster C, Mayer FJ, Wohlfahrt C, Marculescu R, Skoll M, Strassl R, Pavo N, Popow-Kraupp T, Hülsmann M, Bauer M, Aichelburg MC, Rieger A, Goliasch G. Acute HIV Infection Results in Subclinical Inflammatory Cardiomyopathy. J Infect Dis 2019; 218:466-470. [PMID: 29608697 DOI: 10.1093/infdis/jiy183] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 03/28/2018] [Indexed: 11/13/2022] Open
Abstract
The impact of excess viral RNA on myocardial function and morphology in the setting of acute human immunodeficiency virus (HIV) infection remains unknown. In this study, 49 patients with acute HIV infection showed increased levels of N-terminal prohormone of brain natriuretic peptide, a surrogate of myocardial function, which decreased with viral suppression and normalization of systemic inflammation (79 pg/mL vs 28 pg/mL; P < .001). A comparable change was seen with levels of troponin T, a marker of morphologic myocardial damage (4.9 ng/L vs 1.5 ng/L; P < .001). In conclusion, we observed significant functional and morphological myocardial impairment during acute HIV infection, fueled by inflammatory activation and extensive viral replication, resulting in a reversible subclinical inflammatory cardiomyopathy.
Collapse
Affiliation(s)
| | - Florian J Mayer
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Corinna Wohlfahrt
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Rodrig Marculescu
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Michael Skoll
- Department of Dermatology, Medical University of Vienna, Austria
| | - Robert Strassl
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Noemi Pavo
- Department of Internal Medicine II, Medical University of Vienna, Austria
| | | | - Martin Hülsmann
- Department of Internal Medicine II, Medical University of Vienna, Austria
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Austria
| | | | - Armin Rieger
- Department of Dermatology, Medical University of Vienna, Austria
| | - Georg Goliasch
- Department of Internal Medicine II, Medical University of Vienna, Austria
| |
Collapse
|
39
|
Sneller MC, Clarridge KE, Seamon C, Shi V, Zorawski MD, Justement JS, Blazkova J, Huiting ED, Proschan MA, Mora JR, Shetzline M, Moir S, Lane HC, Chun TW, Fauci AS. An open-label phase 1 clinical trial of the anti-α 4β 7 monoclonal antibody vedolizumab in HIV-infected individuals. Sci Transl Med 2019; 11:scitranslmed.aax3447. [PMID: 31488581 DOI: 10.1126/scitranslmed.aax3447] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022]
Abstract
Despite the substantial clinical benefits of antiretroviral therapy (ART), complete eradication of HIV has not been possible. The gastrointestinal tract and associated lymphoid tissues may play an important role in the pathogenesis of HIV infection. The integrin α4β7 facilitates homing of T lymphocytes to the gut by binding to the mucosal addressin cell adhesion molecule-1 (MAdCAM-1) expressed on venules in gut-associated lymphoid tissue. CD4+ T cells with increased expression of α4β7 are susceptible to HIV infection and may be key players in subsequent virus dissemination. Data from nonhuman primate models infected with simian immunodeficiency virus (SIV) have suggested that blockade of the α4β7/MAdCAM-1 interaction may be effective at preventing SIV infection and may have beneficial effects in animals with established viral infection. To explore whether these findings could be reproduced in HIV-infected individuals after interruption of ART, we conducted an open-label phase 1 clinical trial of vedolizumab, a monoclonal antibody against α4β7 integrin. Vedolizumab infusions in 20 HIV-infected individuals were well tolerated with no serious adverse events related to the study drug. After interruption of ART, the median time to meeting protocol criteria to restart therapy was 13 weeks. The median duration of plasma viremia of <400 copies/ml was 5.4 weeks. Only a single subject in the trial experienced prolonged suppression of plasma viremia after interruption of ART. These results suggest that blockade of α4β7 may not be an effective strategy for inducing virological remission in HIV-infected individuals after ART interruption.
Collapse
Affiliation(s)
- Michael C Sneller
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Katherine E Clarridge
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Catherine Seamon
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA
| | - Victoria Shi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Marek D Zorawski
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jesse Shawn Justement
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jana Blazkova
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Erin D Huiting
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | | | | | | | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Henry Clifford Lane
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Anthony S Fauci
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
40
|
Phetsouphanh C, Aldridge D, Marchi E, Munier CML, Meyerowitz J, Murray L, Van Vuuren C, Goedhals D, Fidler S, Kelleher A, Klenerman P, Frater J. Maintenance of Functional CD57+ Cytolytic CD4+ T Cells in HIV+ Elite Controllers. Front Immunol 2019; 10:1844. [PMID: 31440240 PMCID: PMC6694780 DOI: 10.3389/fimmu.2019.01844] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/22/2019] [Indexed: 11/29/2022] Open
Abstract
Cytolytic CD4+ T cells play a prominent role in chronic viral infection. CD4+ CTLs clones specific for HIV-1 Nef and Gag are capable of killing HIV-1 infected CD4+ T cells and macrophages. Additionally, HIV-specific cytolytic CD4+ T cell responses in acute HIV infection are predictive of disease progression. CD57 expression on CD4s identifies cytolytic cells. These cells were dramatically increased in chronic HIV infection. CD57 expression correlated with cytolytic granules, granzyme B and perforin expression. They express lower CCR5 compared to CD57- cells, have less HIV total DNA, and were a minor component of the HIV reservoir. A small percentage of CD57+ CD4+ CTLs from EC were HIV-specific, could upregulate IFNγ with Gag peptide stimulation, express cytolytic granule markers and maintain TbethighEomes+ transcription factor phenotype. This was not observed in viraemic controllers. The maintenance of HIV-specific CD4 cytolytic function in Elite controllers together with CD8 CTLs may be important for the control of HIV viraemia and of potential relevance to cure strategies.
Collapse
Affiliation(s)
| | - Daniel Aldridge
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Emanuele Marchi
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - C. Mee Ling Munier
- Department of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Jodi Meyerowitz
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Lyle Murray
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | | | - Dominique Goedhals
- National Health Laboratory Service, Division of Virology, University of the Free State, Bloemfontein, South Africa
| | | | - Anthony Kelleher
- Department of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - John Frater
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
41
|
Teira R, Gutierrez M, Galindo P, Martínez E, Muñoz P, de la Fuente B, Téllez F, Montero M. Integrase strand-transfer inhibitors for treatment of early HIV infection: A case series. Medicine (Baltimore) 2019; 98:e16866. [PMID: 31464915 PMCID: PMC6736467 DOI: 10.1097/md.0000000000016866] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
This study evaluated whether the interval from the first clinic visit until the start of antiretroviral treatment (ART) was correlated with common parameters of immunological recovery among patients with early HIV infection (EHI).We reviewed the medical records of patients with EHI who started ART using integrase strand-transfer inhibitors (ISTIs) within the first 6 months after diagnosis. Simple linear regression analyses were performed to determine whether the interval from the first visit to the start of ART was correlated with 1-year changes in CD4+ cell count, CD8+ cell count, CD4+ percentage, and CD4+/CD8+ ratio.Fifty-three patients with probable or definite EHI started ART using ISTIs between April 2014 and August 2016. Forty-nine patients completed 1 year of follow-up, including 48 men. The routes of HIV transmission were 1 case of needle sharing, 5 cases of heterosexual activity, and 43 cases of men who had sex with men. None of the immunological recovery parameters were correlated with time to the start of ART (CD4+ cell count: R = .12, P = .42; CD8+ cell count: R = .107, P = .5; CD4+ percentage: R = .14, P = .34; CD4+/CD8+ ratio: R = .23, P = .14). Furthermore, subgroup sensitivity analyses failed to detect significant correlations based on definite or probable diagnoses, treatment using elvitegravir or dolutegravir, or the time from HIV diagnosis to ART initiation.This series of EHI cases indicate that using ART with ISTI-based regimens is efficacious and well-tolerated. However, earlier initiation of treatment was not significantly correlated with common parameters of immunological recovery.
Collapse
Affiliation(s)
- Ramón Teira
- Hospital Universitario de Sierrallana, Torrelavega
| | | | | | - Elisa Martínez
- Complejo Hospitalario Universitario de Albacete, Albacete
| | - Pepa Muñoz
- Hospital Universitario de Basurto, Bilbao
| | | | | | - Marta Montero
- Hospital Universitario y Politécnico la Fe, Valencia, Spain
| |
Collapse
|
42
|
Palmer DS, Turner I, Fidler S, Frater J, Goedhals D, Goulder P, Huang KHG, Oxenius A, Phillips R, Shapiro R, Vuuren CV, McLean AR, McVean G. Mapping the drivers of within-host pathogen evolution using massive data sets. Nat Commun 2019; 10:3017. [PMID: 31289267 PMCID: PMC6616926 DOI: 10.1038/s41467-019-10724-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 05/20/2019] [Indexed: 11/09/2022] Open
Abstract
Differences among hosts, resulting from genetic variation in the immune system or heterogeneity in drug treatment, can impact within-host pathogen evolution. Genetic association studies can potentially identify such interactions. However, extensive and correlated genetic population structure in hosts and pathogens presents a substantial risk of confounding analyses. Moreover, the multiple testing burden of interaction scanning can potentially limit power. We present a Bayesian approach for detecting host influences on pathogen evolution that exploits vast existing data sets of pathogen diversity to improve power and control for stratification. The approach models key processes, including recombination and selection, and identifies regions of the pathogen genome affected by host factors. Our simulations and empirical analysis of drug-induced selection on the HIV-1 genome show that the method recovers known associations and has superior precision-recall characteristics compared to other approaches. We build a high-resolution map of HLA-induced selection in the HIV-1 genome, identifying novel epitope-allele combinations.
Collapse
Affiliation(s)
- Duncan S Palmer
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK.
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK.
- Institute for Emerging Infections, The Oxford Martin School, Oxford, OX1 3BD, UK.
| | - Isaac Turner
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Sarah Fidler
- Division of Medicine, Wright Fleming Institute, Imperial College, London, W2 1PG, UK
| | - John Frater
- Institute for Emerging Infections, The Oxford Martin School, Oxford, OX1 3BD, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, OX1 3SY, UK
- Oxford NIHR Biomedical Research Centre, Oxford, OX3 7LE, UK
| | - Dominique Goedhals
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, 4013, South Africa
| | - Philip Goulder
- Division of Infectious Diseases, University of the Free State, and 3 Military Hospital, Bloemfontein, 9300, South Africa
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, OX1 3SY, UK
| | - Kuan-Hsiang Gary Huang
- Nuffield Department of Clinical Medicine, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, OX1 3SY, UK
- Einstein Medical Center Philadelphia, 5501 Old York Road, PA, 19141, USA
| | - Annette Oxenius
- Institute of Microbiology, Swiss Federal Institute of Technology Zurich, 8093, Zurich, Switzerland
| | - Rodney Phillips
- Institute for Emerging Infections, The Oxford Martin School, Oxford, OX1 3BD, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, OX1 3SY, UK
- Oxford NIHR Biomedical Research Centre, Oxford, OX3 7LE, UK
- Faculty of Medicine, UNSW Sydney, NSW, 2052, Australia
| | - Roger Shapiro
- Botswana Harvard AIDS Institute Partnership, Gaborone, BO 320, Botswana
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, MA, 02215, USA
| | - Cloete van Vuuren
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, 4013, South Africa
| | - Angela R McLean
- Institute for Emerging Infections, The Oxford Martin School, Oxford, OX1 3BD, UK
- Zoology Department, University of Oxford, South Parks Road, Oxford, OX1 3PS, UK
| | - Gil McVean
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| |
Collapse
|
43
|
The benefit of immediate compared with deferred antiretroviral therapy on CD4+ cell count recovery in early HIV infection. AIDS 2019; 33:1335-1344. [PMID: 31157663 DOI: 10.1097/qad.0000000000002219] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To assess the impact of immediate vs. deferred antiretroviral therapy (ART) on CD4 recovery among individuals early in HIV infection. DESIGN Using serologic markers of early infection together with self-reported dates of infection and HIV diagnosis, ART-naive participants who were randomized to immediate vs. deferred ART in the Strategic Timing of Antiretroviral Treatment trial were classified into subgroups of duration of HIV infection at baseline. CD4 cell count recovery over follow-up according to duration of HIV infection was investigated. METHODS Three subgroups were defined: first, infected 6 months or less (n = 373); second, infected 6-24 months (n = 2634); and third, infected 24 months or longer (n = 1605). Follow-up CD4, CD8, and CD4 : CD8 ratio for the immediate and deferred ART groups were compared by subgroup using linear models. For the deferred ART group, decline to CD4 less than 350 cells/μl or AIDS according to infection duration was compared using time-to-event methods. RESULTS Follow-up CD4 cell count differences (immediate minus deferred) were greater for those recently infected (+231 cells/μl) compared with the two other subgroups (202 and 171 cells/μl; P < 0.001). CD4 : CD8 ratio treatment differences varied significantly (P < 0.001) according to duration of infection. In the deferred ART group, decline to CD4 less than 350 cells/μl or AIDS was greater among those recently infected (16.1 vs. 13.2 and 10.5 per 100 person years for those infected 6-24 and ≥24 months; P = 0.002). CONCLUSION In this randomized comparison of immediate vs. deferred ART, the CD4 cell count difference was greatest for those recently infected with HIV, emphasizing the importance of immediate ART initiation.
Collapse
|
44
|
A generalizable method for estimating duration of HIV infections using clinical testing history and HIV test results. AIDS 2019; 33:1231-1240. [PMID: 30870196 DOI: 10.1097/qad.0000000000002190] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To determine the precision of new and established methods for estimating duration of HIV infection. DESIGN A retrospective analysis of HIV testing results from serial samples in commercially available panels, taking advantage of extensive testing previously conducted on 53 seroconverters. METHODS We initially investigated four methods for estimating infection timing: method 1, 'Fiebig stages' based on test results from a single specimen; method 2, an updated '4th gen' method similar to Fiebig stages but using antigen/antibody tests in place of the p24 antigen test; method 3, modeling of 'viral ramp-up' dynamics using quantitative HIV-1 viral load data from antibody-negative specimens; and method 4, using detailed clinical testing history to define a plausible interval and best estimate of infection time. We then investigated a 'two-step method' using data from both methods 3 and 4, allowing for test results to have come from specimens collected on different days. RESULTS Fiebig and '4th gen' staging method estimates of time since detectable viremia had similar and modest correlation with observed data. Correlation of estimates from both new methods (3 and 4), and from a combination of these two ('two-step method') was markedly improved and variability significantly reduced when compared with Fiebig estimates on the same specimens. CONCLUSION The new 'two-step' method more accurately estimates timing of infection and is intended to be generalizable to more situations in clinical medicine, research, and surveillance than previous methods. An online tool is now available that enables researchers/clinicians to input data related to method 4, and generate estimated dates of detectable infection.
Collapse
|
45
|
Lundgren JD, Borges AH, Neaton JD. Serious Non-AIDS Conditions in HIV: Benefit of Early ART. Curr HIV/AIDS Rep 2019; 15:162-171. [PMID: 29504063 DOI: 10.1007/s11904-018-0387-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Optimal control of HIV can be achieved by early diagnosis followed by the initiation of antiretroviral therapy (ART). Two large randomised trials (TEMPRANO and START) have recently been published documenting the clinical benefits to HIV-positive adults of early ART initiation. Main findings are reviewed with a focus on serious non-AIDS (SNA) conditions. RECENT FINDINGS Data from the two trials demonstrated that initiating ART early in the course of HIV infection resulted in marked reductions in the risk of opportunistic diseases and invasive bacterial infections. This indicates that HIV causes immune impairment in early infection that is remedied by controlling viral replication. Intriguingly, in START, a marked reduction in risk of cancers, both infection-related and unrelated types of cancers, was observed. Like the findings for opportunistic infections, this anti-cancer effect of early ART shows how the immune system influences important pro-oncogenic processes. In START, there was also some evidence suggesting that early ART initiation preserved kidney function, although the clinical consequence of this remains unclear. Conversely, while no adverse effects were evident, the trials did not demonstrate a clear effect on metabolic-related disease outcomes, pulmonary disease, or neurocognitive function. HIV causes immune impairment soon after acquisition of infection. ART reverses this harm at least partially. The biological nature of the immune impairment needs further elucidation, as well as mechanisms and clinical impact of innate immune activation. Based on the findings from TEMPRANO and START, and because ART lowers the risk of onward transmission, ART initiation should be offered to all persons following their diagnosis of HIV.
Collapse
Affiliation(s)
- Jens D Lundgren
- Centre of Excellence for Health, Immunity and Infections (CHIP), Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Esther Møllers Vej 6, 2100, Copenhagen Ø, Denmark.
| | - Alvaro H Borges
- Centre of Excellence for Health, Immunity and Infections (CHIP), Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Esther Møllers Vej 6, 2100, Copenhagen Ø, Denmark
| | - James D Neaton
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
46
|
Mothe B, Manzardo C, Sanchez-Bernabeu A, Coll P, Morón-López S, Puertas MC, Rosas-Umbert M, Cobarsi P, Escrig R, Perez-Alvarez N, Ruiz I, Rovira C, Meulbroek M, Crook A, Borthwick N, Wee EG, Yang H, Miró JM, Dorrell L, Clotet B, Martinez-Picado J, Brander C, Hanke T. Therapeutic Vaccination Refocuses T-cell Responses Towards Conserved Regions of HIV-1 in Early Treated Individuals (BCN 01 study). EClinicalMedicine 2019; 11:65-80. [PMID: 31312806 PMCID: PMC6610778 DOI: 10.1016/j.eclinm.2019.05.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Strong and broad antiviral T-cell responses targeting vulnerable sites of HIV-1 will likely be a critical component for any effective cure strategy. METHODS BCN01 trial was a phase I, open-label, non-randomized, multicenter study in HIV-1-positive individuals diagnosed and treated during early HIV-1 infection to evaluate two vaccination regimen arms, which differed in the time (8 versus 24 week) between the ChAdV63.HIVconsv prime and MVA.HIVconsv boost vaccinations. The primary outcome was safety. Secondary endpoints included frequencies of vaccine-induced IFN-γ+ CD8+ T cells, in vitro virus-inhibitory capacity, plasma HIV-1 RNA and total CD4+ T-cells associated HIV-1 DNA. (NCT01712425). FINDINGS No differences in safety, peak magnitude or durability of vaccine-induced responses were observed between long and short interval vaccination arms. Grade 1/2 local and systemic post-vaccination events occurred in 22/24 individuals and resolved within 3 days. Weak responses to conserved HIV-1 regions were detected in 50% of the individuals before cART initiation, representing median of less than 10% of their total HIV-1-specific T cells. All participants significantly elevated these subdominant T-cell responses, which after MVA.HIVconsv peaked at median (range) of 938 (73-6,805) IFN-γ SFU/106 PBMC, representing on average 58% of their total anti-HIV-1 T cells. The decay in the size of the HIV-1 reservoir was consistent with the first year of early cART initiation in both arms. INTERPRETATION Heterologous prime-boost vaccination with ChAdV63-MVA/HIVconsv was well-tolerated and refocused pre-cART T-cell responses towards more protective epitopes, in which immune escape is frequently associated with reduced HIV-1 replicative fitness and which are common to most global HIV-1 variants. FUNDING HIVACAT Catalan research program for an HIV vaccine and Fundació Gloria Soler. Vaccine manufacture was jointly funded by the Medical Research Council (MRC) UK and the UK Department for International Development (DFID) under the MRC/DFID Concordat agreements (G0701669. RESEARCH IN CONTEXT Evidence Before this Study: T cells play an important role in the control of HIV infection and may be particularly useful for HIV-1 cure by killing cells with reactivated HIV-1. Evidence is emerging that not all T-cell responses are protective and mainly only those targeting conserved regions of HIV-1 proteins are effective, but typically immunologically subdominant, while those recognizing hypervariable, easy-to-escape immunodominant 'decoys' do not control viremia and do not protect from a loss of CD4 T cells. We pioneered a vaccine strategy focusing T-cell responses on the most conserved regions of the HIV-1 proteome using an immunogen designated HIVconsv. T cells elicited by the HIVconsv vaccines in HIV-uninfected UK and Kenyan adults inhibited in vitro replication of HIV-1 isolates from 4 major global clades A, B, C and D.Added Value of this Study: The present study demonstrated the concept that epitopes subdominant in natural infection, when taken out of the context of the whole HIV-1 proteome and presented to the immune system by a potent simian adenovirus prime-poxvirus MVA boost regimen, can induce strong responses in patients on antiretroviral treatment and efficiently refocus HIV-1-specific T-cells to the protective epitopes delivered by the vaccine.Implications of all the Available Evidence: Nearly all HIV-1 vaccine strategies currently emphasize induction of broadly neutralizing Abs. The HIVconsv vaccine is one of a very few approaches focussing exclusively on elicitation of T cells and, therefore, can complement antibody induction for better prevention and cure. Given the cross-clade reach on the HIVconsv immunogen design, if efficient, the HIVconsv vaccines could be deployed globally. Effective vaccines will likely be a necessary component in combination with other available preventive measures for halting the HIV-1/AIDS epidemic.
Collapse
Affiliation(s)
- Beatriz Mothe
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Universitat de Vic - Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
- Corresponding author at: IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Crta Canyet s/n, 08916, Badalona, Barcelona, Spain.
| | | | | | - Pep Coll
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | | | - Miriam Rosas-Umbert
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Patricia Cobarsi
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Roser Escrig
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Núria Perez-Alvarez
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Technical University of Catalonia, Barcelona, Spain
| | - Irene Ruiz
- Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Cristina Rovira
- Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | - Alison Crook
- The Jenner Institute, University of Oxford, Oxford, UK
| | | | - Edmund G. Wee
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Hongbing Yang
- Nuffield Department of Medicine and Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Jose M. Miró
- Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Lucy Dorrell
- Nuffield Department of Medicine and Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Universitat de Vic - Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
- Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Universitat de Vic - Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
- ICREA, Pg. Luis Companys 23, Barcelona, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Universitat de Vic - Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
- ICREA, Pg. Luis Companys 23, Barcelona, Spain
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford, Oxford, UK
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
47
|
Lau JS, Smith MZ, Lewin SR, McMahon JH. Clinical trials of antiretroviral treatment interruption in HIV-infected individuals. AIDS 2019; 33:773-791. [PMID: 30883388 DOI: 10.1097/qad.0000000000002113] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
: Despite the benefits of antiretroviral therapy (ART) for people living with HIV, there has been a long-standing research interest in interrupting ART as a strategy to minimize adverse effects of ART as well as to test interventions aiming to achieve a degree of virological control without ART. We performed a systematic review of HIV clinical studies involving treatment interruption from 2000 to 2017 to describe the differences between treatment interruption in studies that contained and didn't contain an intervention. We assessed differences in monitoring strategies, threshold to restart ART, duration and adverse outcomes of treatment interruption, and factors aimed at minimizing transmission. We found that treatment interruption has been incorporated into 159 clinical studies since 2000 and is increasingly being included in trials to assess the efficacy of interventions to achieve sustained virological remission off ART. Great heterogeneity was noted in immunological, virological and clinical monitoring strategies, as well as in thresholds to recommence ART. Treatment interruption in recent intervention studies were more closely monitored, had more conservative thresholds to restart ART and had a shorter treatment interruption duration, compared with older treatment interruption studies that didn't include an intervention.
Collapse
|
48
|
Khan S, Parrillo M, Gutierrez AH, Terry FE, Moise L, Martin WD, De Groot AS. Immune escape and immune camouflage may reduce the efficacy of RTS,S vaccine in Malawi. Hum Vaccin Immunother 2019; 16:214-227. [PMID: 30614773 PMCID: PMC7062414 DOI: 10.1080/21645515.2018.1560772] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The RTS,S/AS01 malaria vaccine will undergo a pilot vaccination study in sub-Saharan Africa beginning in 2019. RTS,S/AS01 Phase III trials reported an efficacy of 28.3% (children 5–17 months) and 18.3% (infants 6–12 weeks), with substantial variability across study sites. We postulated that the relatively low efficacy of the RTS,S vaccine and variability across sites may be due to lack of T-cell epitopes in the vaccine antigen, and due to the HLA distribution of the vaccinated population, and/or due to ‘immune camouflage’, an immune escape mechanism. To examine these hypotheses, we used immunoinformatics tools to compare T helper epitopes contained in RTS,S vaccine antigens with Plasmodium falciparum circumsporozoite protein (CSP) variants isolated from infected individuals in Malawi. The prevalence of epitopes restricted by specific HLA-DRB1 alleles was inversely associated with prevalence of the HLA-DRB1 allele in the Malawi study population, suggesting immune escape. In addition, T-cell epitopes in the CSP of strains circulating in Malawi were more often restricted by low-frequency HLA-DRB1 alleles in the population. Furthermore, T-cell epitopes that were highly conserved across CSP variants in Malawi possessed TCR-facing residues that were highly conserved in the human proteome, potentially reducing T-cell help through tolerance. The CSP component of the RTS,S vaccine also exhibited a low degree of T-cell epitope relatedness to circulating variants. These results suggest that RTS,S vaccine efficacy may be impacted by low T-cell epitope content, reduced presentation of T-cell epitopes by prevalent HLA-DRB1, high potential for human-cross-reactivity, and limited conservation with the CSP of circulating malaria strains.
Collapse
Affiliation(s)
- Sundos Khan
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA
| | - Matthew Parrillo
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA
| | | | | | - Leonard Moise
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA.,EpiVax, Inc., Providence, RI, USA
| | | | - Anne S De Groot
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA.,EpiVax, Inc., Providence, RI, USA
| |
Collapse
|
49
|
In-vitro viral suppressive capacity correlates with immune checkpoint marker expression on peripheral CD8+ T cells in treated HIV-positive patients. AIDS 2019; 33:387-398. [PMID: 30702513 DOI: 10.1097/qad.0000000000002068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To determine whether viral suppressive capacity (VSC) of CD8+ T cells can be boosted by stimulation with HIV-1 peptides and whether the ability to control HIV-1 replication correlates with immunological (cytokine production and CD8+ T-cell phenotype) and viral reservoir measures (total HIV-1 DNA and cell-associated RNA) in well treated HIV-infected chronic progressors. DESIGN We compared VSC of peripheral CD8+ T cells to cytokine production profile in response to peptide stimulation, detailed phenotype (17-color flow-cytometry), reservoir size (total HIV-1 DNA), basal viral transcription (unspliced cell-associated RNA) and inducible viral transcription (tat/rev induced limiting dilution assay) in 36 HIV+ patients on cART and six healthy donors. RESULTS We found that the VSC of CD8+ T cells can be increased by prior stimulation with a pool of consensus HIV-1 gag peptides in a significant proportion of progressor patients. We also found that VSC after peptide stimulation was correlated with higher expression of immune checkpoint markers on subsets of terminally differentiated effector memory (TEMRA) CD8 T cells as well as with production of IFN-γ, TNF-α and IL-10. We did not find a correlation between VSC and viral reservoir measures. CONCLUSION These results add to a small body of evidence that the capacity of CD8+ T cells to suppress viral replication is increased after stimulation with HIV-1 peptides. Interestingly, this VSC was correlated with expression of immune checkpoint markers, which are generally considered to be markers of exhaustion. Our findings may guide further investigations into immune phenotypes correlated with viral suppression.
Collapse
|
50
|
Bekele Y, Lakshmikanth T, Chen Y, Mikes J, Nasi A, Petkov S, Hejdeman B, Brodin P, Chiodi F. Mass cytometry identifies distinct CD4+ T cell clusters distinguishing HIV-1-infected patients according to antiretroviral therapy initiation. JCI Insight 2019; 4:125442. [PMID: 30728327 DOI: 10.1172/jci.insight.125442] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/20/2018] [Indexed: 01/13/2023] Open
Abstract
Recent guidelines recommend antiretroviral therapy (ART) to be administered as early as possible during HIV-1 infection. Few studies addressed the immunological benefit of commencing ART during the acute phase of infection. We used mass cytometry to characterize blood CD4+ T cells from HIV-1-infected patients who initiated ART during acute or chronic phase of infection. Using this method, we analyzed a large number of markers on millions of individual immune cells. The results revealed that CD4+ T cell clusters with high expression of CD27, CD28, CD127, and CD44, whose function involves T cell migration to inflamed tissues and survival, are more abundant in healthy controls and patients initiating ART during the acute phase; on the contrary, CD4+ T cell clusters in patients initiating ART during the chronic phase had reduced expression of these markers. The results are suggestive of a better preserved immune function in HIV-1-infected patients initiating ART during acute infection.
Collapse
Affiliation(s)
- Yonas Bekele
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, and
| | - Tadepally Lakshmikanth
- Science for Life Laboratory, Division of Clinical Pediatrics, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Yang Chen
- Science for Life Laboratory, Division of Clinical Pediatrics, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Jaromir Mikes
- Science for Life Laboratory, Division of Clinical Pediatrics, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Aikaterini Nasi
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, and
| | - Stefan Petkov
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, and
| | - Bo Hejdeman
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, and Unit of Infectious Diseases, Venhälsan, Södersjukhuset, Stockholm, Sweden
| | - Petter Brodin
- Science for Life Laboratory, Division of Clinical Pediatrics, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Newborn Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, and
| |
Collapse
|