1
|
Sohai DK, Keller MD, Hanley PJ, Hoq F, Kukadiya D, Datar A, Reynolds E, Copertino DC, Lazarski C, McCann CD, Tanna J, Shibli A, Lang H, Zhang A, Chansky PA, Motta C, Huynh TT, Dwyer B, Wilson A, Lynch R, Mota TM, Conce Alberto WD, Brumme ZL, Kinloch NN, Cruz CRY, MacLaren Ehui L, Henn S, Brad Jones R, Bollard CM. Autologous HIV-specific T cell therapy targeting conserved epitopes is well-tolerated in six adults with HIV: an open-label, single-arm phase 1 study. Nat Commun 2025; 16:4510. [PMID: 40374689 PMCID: PMC12081906 DOI: 10.1038/s41467-025-59810-2] [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: 06/04/2024] [Accepted: 05/06/2025] [Indexed: 05/17/2025] Open
Abstract
Novel cellular therapies may enable HIV control or cure. HIV-specific T cells targeting conserved immunogenic protein regions of HIV Gag/Pol and the entirety of HIV Nef, termed HST-NEETs, eliminate HIV infected cells in vitro. Here we enroll seven participants in an open-label, single-arm phase 1 study (NCT03485963) to evaluate the safety (primary endpoint) of two autologous administrations of HST-NEET products without prescribed lymphodepletion. Adults with well-controlled HIV on anti-retroviral therapy are eligible. Six participants completed safety monitoring. No serious product-related toxicities are observed. Secondary endpoints are to assess expansion and persistence of HIV-reactive T cell clones, and changes to the HIV reservoir for each infused participant. HIV-specific T cell and HIV anti-Env antibody responses increase in two participants after infusion two. A trend towards decreasing levels of intact proviruses is observed in 2 participants. Three participants show persistence of HIV-reactive, product-associated T cell clones for ≥40 weeks post infusions. HST-NEETs infusions are well-tolerated. Future trials are needed to evaluate the efficacy of HST-NEETs in this population.
Collapse
Affiliation(s)
- Danielle K Sohai
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Michael D Keller
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Fahmida Hoq
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Divyesh Kukadiya
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Anushree Datar
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Emily Reynolds
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Dennis C Copertino
- Infectious Diseases Division, Department of Medicine, Weill-Cornell Medicine, New York, NY, USA
| | - Christopher Lazarski
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Chase D McCann
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Jay Tanna
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Abeer Shibli
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Haili Lang
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Anqing Zhang
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Pamela A Chansky
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Cecilia Motta
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Tan T Huynh
- Infectious Diseases Division, Department of Medicine, Weill-Cornell Medicine, New York, NY, USA
| | - Bridget Dwyer
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Andrew Wilson
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Rebecca Lynch
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Talia M Mota
- Infectious Diseases Division, Department of Medicine, Weill-Cornell Medicine, New York, NY, USA
| | | | - Zabrina L Brumme
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada
| | - Natalie N Kinloch
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada
| | - Conrad Russell Y Cruz
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | | | - Sarah Henn
- Whitman-Walker Health, Washington, DC, USA
| | - R Brad Jones
- Infectious Diseases Division, Department of Medicine, Weill-Cornell Medicine, New York, NY, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA.
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA.
| |
Collapse
|
2
|
Garland JM, Mayan H, Kantor R. Treatment of Advanced HIV in the Modern Era. Drugs 2025:10.1007/s40265-025-02181-1. [PMID: 40354016 DOI: 10.1007/s40265-025-02181-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 05/14/2025]
Abstract
Antiretroviral therapy has transformed human immunodeficiency virus (HIV) infection from a fatal illness into a manageable chronic condition. However, despite remarkable progress, the HIV epidemic remains a global health challenge, with ambitious targets such as 95-95-95 by 2030 at risk of being unmet. While antiretroviral therapy availability has expanded worldwide, gaps persist, including unawareness of HIV status, inconsistent medication uptake, and limited engagement in care across diverse settings. Advanced HIV represents a particularly challenging yet underexplored aspect of HIV care. Its definition is complex, complicating efforts to address the needs of this vulnerable population. This review characterizes advanced HIV populations, defines them by spectra of immune suppression, antiretroviral therapy exposure, and drug resistance, and explores contemporary approaches to their management, with a particular focus on drug resistance and its clinical implications in modern HIV care. It highlights the unique challenges faced by individuals presenting late to care, those with limited care engagement, and aging populations with long-term exposure to HIV and antiretroviral therapy. By defining these populations, refining our understanding of advanced HIV, and addressing the diverse needs of affected individuals, providers can enhance outcomes and develop strategies to overcome barriers to care. Bridging these critical gaps is essential to advancing global efforts to end the HIV epidemic, both in the USA and worldwide.
Collapse
Affiliation(s)
- Joseph M Garland
- The Miriam Hospital, Providence, RI, USA
- Brown University, Providence, RI, USA
| | - Haim Mayan
- Sheba Medical Center, Tel Aviv, Israel
- Tel Aviv University, Tel Aviv, Israel
| | - Rami Kantor
- The Miriam Hospital, Providence, RI, USA.
- Brown University, Providence, RI, USA.
| |
Collapse
|
3
|
Ravn K, Cobuccio L, Muktupavela RA, Meisner J, Danielsen LS, Benros ME, Korneliussen TS, Sikora M, Willerslev E, Allentoft ME, Irving-Pease EK, Rasmussen S. Tracing the evolutionary history of the CCR5delta32 deletion via ancient and modern genomes. Cell 2025:S0092-8674(25)00417-9. [PMID: 40328257 DOI: 10.1016/j.cell.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/31/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025]
Abstract
The chemokine receptor variant CCR5delta32 is linked to HIV-1 resistance and other conditions. Its evolutionary history and allele frequency (10%-16%) in European populations have been extensively debated. We provide a detailed perspective of the evolutionary history of the deletion through time and space. We discovered that the CCR5delta32 allele arose on a pre-existing haplotype consisting of 84 variants. Using this information, we developed a haplotype-aware probabilistic model to screen 934 low-coverage ancient genomes and traced the origin of the CCR5delta32 deletion to at least 6,700 years before the present (BP) in the Western Eurasian Steppe region. Furthermore, we present strong evidence for positive selection acting upon the CCR5delta32 haplotype between 8,000 and 2,000 years BP in Western Eurasia and show that the presence of the haplotype in Latin America can be explained by post-Columbian genetic exchanges. Finally, we point to complex CCR5delta32 genotype-haplotype-phenotype relationships, which demand consideration when targeting the CCR5 receptor for therapeutic strategies.
Collapse
Affiliation(s)
- Kirstine Ravn
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leonardo Cobuccio
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rasa Audange Muktupavela
- Section for Molecular Ecology and Evolution, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Meisner
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen Research Centre for Mental Health, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lasse Schnell Danielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Eriksen Benros
- Copenhagen Research Centre for Mental Health, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thorfinn Sand Korneliussen
- Lundbeck Foundation GeoGenetics Centre, Globe Institute, University of Copenhagen, Copenhagen, Denmark; Centre for Ancient Environmental Genomics, University of Copenhagen, Copenhagen, Denmark
| | - Martin Sikora
- Lundbeck Foundation GeoGenetics Centre, Globe Institute, University of Copenhagen, Copenhagen, Denmark; Centre for Ancient Environmental Genomics, University of Copenhagen, Copenhagen, Denmark
| | - Eske Willerslev
- Lundbeck Foundation GeoGenetics Centre, Globe Institute, University of Copenhagen, Copenhagen, Denmark; Centre for Ancient Environmental Genomics, University of Copenhagen, Copenhagen, Denmark; Department of Genetics, University of Cambridge, Cambridge, UK; MARUM, University of Bremen, Bremen, Germany
| | - Morten E Allentoft
- Lundbeck Foundation GeoGenetics Centre, Globe Institute, University of Copenhagen, Copenhagen, Denmark; Trace and Environmental DNA (TrEnD) Laboratory, School of Molecular and Life Sciences, Curtin University, Perth, WA, Australia
| | - Evan K Irving-Pease
- Section for Molecular Ecology and Evolution, Globe Institute, University of Copenhagen, Copenhagen, Denmark; Lundbeck Foundation GeoGenetics Centre, Globe Institute, University of Copenhagen, Copenhagen, Denmark.
| | - Simon Rasmussen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Meng L, Zhao H, Chang S, Li W, Tian Y, Wang R, Wang L, Gu T, Wu J, Yu B, Wang C, Yu X. Engineering of CD8 + T cells with an HIV-specific synthetic notch receptor to secrete broadly therapeutic antibodies for combining antiviral humoral and cellular immune responses. mBio 2025; 16:e0383924. [PMID: 39998238 PMCID: PMC11980546 DOI: 10.1128/mbio.03839-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
The application of immunotherapeutic strategies, such as chimeric antigen receptor-T cells and broadly neutralizing antibodies (bNAbs), for the treatment of human immunodeficiency virus (HIV) infection is hindered by the latent reservoirs and viral escape. Achieving long-term control of viral load in the absence of antiretroviral therapy requires a combination approach utilizing these immunotherapeutic strategies. For this purpose, we developed novel anti-HIV-1 synthetic Notch (synNotch) receptor-T cells, termed CD4-17b-VN, which express both a bNAb (VRC01) and a bispecific T cell-engaging protein (N6-αCD3) with antigenic control. The synNotch receptor-expressing cells can sense the viral antigen presented on both HIV-1 particles and the surface of target cells. A human T cell line equipped with the CD4-17b-VN circuit could effectively control VRC01 and N6-αCD3 secretion upon sensitization, suppress the infection of diverse subtypes of HIV-1 strains, and mediate specific bypass cytotoxic activity against infected and latency-reactivated cells. Additionally, CD4-17b-VN CD8+ T cells exhibited long-lasting suppression of infected cells and stronger killing effect on latency-reactivated cells in vitro. Importantly, we demonstrated that the synNotch receptor did not increase susceptibility to HIV-1 infection in the engineered cells. Our study validates the concept of a synNotch platform-based T cell therapeutic approach that can deliver broadly therapeutic antibodies in an HIV-1 antigen-controlled manner, which may have important implications for the functional cure of AIDS.IMPORTANCEAdoptive transfer of effector T cells modified with a chimeric antigen receptor has been proposed as an applicable approach to treat human immunodeficiency virus (HIV) infection. The synNotch receptor (SNR) system serves as a versatile tool, enabling customized programming of input and output functions in mammalian cells. Herein, we report a novel synNotch platform-based approach for T cell engineering targeting both cell-free particles and infected cells by coupling antibody neutralization with cytotoxicity. Our findings demonstrate that the engineered CD4-17b SNR enables controllable production of functional anti-HIV-1 broadly neutralizing antibody and bispecific T cell-engaging protein upon recognition of the viral particle and cell surface antigens by the bifunctional synNotch-T cells. Human primary CD8+ T cells equipped with the bifunctional synNotch circuit CD4-17b-VN can effectively suppress long-term viral replication and reduce latency-reactivated cells in vitro, without the undesired risk of being infected by the virus, suggesting their potential candidacy for AIDS therapy with prospects for future clinical applications.
Collapse
Affiliation(s)
- Lina Meng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Haichi Zhao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Shangkun Chang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Weiting Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Yinghui Tian
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Ruihong Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Libian Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Tiejun Gu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Chu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
5
|
Xiao Q, He S, Wang C, Zhou Y, Zeng C, Liu J, Liu T, Li T, Quan X, Wang L, Zhai L, Liu Y, Li J, Zhang X, Liu Y. Deep Thought on the HIV Cured Cases: Where Have We Been and What Lies Ahead? Biomolecules 2025; 15:378. [PMID: 40149913 PMCID: PMC11940578 DOI: 10.3390/biom15030378] [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: 01/02/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Antiretroviral therapy (ART) can effectively suppress the replication of human immunodeficiency virus (HIV), but it cannot completely eradicate the virus. The persistent existence of the HIV reservoir is a major obstacle in the quest for a cure. To date, there have been a total of seven cured cases of HIV worldwide. These patients all cleared HIV while undergoing allogeneic stem cell transplantation (allo-HSCT) for hematological malignancies. However, in these cases, the specific mechanism by which allo-HSCT leads to the eradication of HIV remains unclear, so it is necessary to conduct an in-depth analysis. Due to the difficulty in obtaining donors and the risks associated with transplantation, this treatment method is not applicable to all HIV patients. There is still a need to explore new treatment strategies. In recent years, emerging therapies such as neutralizing antibody immunotherapy, chimeric antigen receptor T cell (CAR-T) therapy, gene editing, and antiviral therapies targeting the reservoir have attracted wide attention due to their ability to effectively inhibit HIV replication. This article first elaborates on the nature of the HIV reservoir, then deeply explores the treatment modalities and potential success factors of HIV cured cases, and finally discusses the current novel treatment methods, hoping to provide comprehensive and feasible strategies for achieving the cure of HIV.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Xiaomei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yao Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
6
|
Gasca-Capote C, Ruiz-Mateos E. Persistent elite controllers as the key model to identify permanent HIV remission. Curr Opin HIV AIDS 2025; 20:165-171. [PMID: 39773856 PMCID: PMC11809733 DOI: 10.1097/coh.0000000000000907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW To summarize the heterogeneity in the elite controllers population with the aim to identify a compatible profile with a persistent HIV remission, making distinction between persistent elite controllers, people with HIV (PWHIV) who permanently maintain virological control in the absence of antiretroviral treatment (ART), and transient elite controllers, PWHIV who eventually lose virological control. For this purpose, it is important to consider the mechanisms and biomarkers that have previously been associated with the maintenance and loss of the natural virological control. RECENT FINDINGS Transient elite controllers, before losing virological control, exhibit a distinct metabolomic, proteomic, microRNAs (miRNA), immunological and virological profile compared to persistent elite controllers. In addition to a reduced and less polyfunctional HIV-specific T-cell response, transient elite controllers show a greater proportion of intact proviruses integrated into genic regions. In contrast, persistent elite controllers display a privileged HIV-1 reservoir profile with absence of detected intact proviruses or low proportion of clonal intact proviruses preferentially integrated into genomic features associated with HIV-1 transcriptional repression. SUMMARY According to previous studies, the comprehensive characterization of persistent elite controllers might be crucial to identify other PWHIV with this distinct profile as spontaneously cured.
Collapse
Affiliation(s)
- Carmen Gasca-Capote
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
| | | |
Collapse
|
7
|
Lewin SR, Bansbach C, Kemps D, Mathae L, Das KT, McCune JM, Deeks SG, Ndung'u T. Target product profile for cell-based and gene-based therapies to achieve a cure for HIV. Lancet HIV 2025; 12:e154-e162. [PMID: 39761679 DOI: 10.1016/s2352-3018(24)00277-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 02/08/2025]
Abstract
This target product profile (TPP) highlights the minimal and optimal characteristics for ex-vivo and in-vivo cell and gene therapy-based products aimed at achieving an HIV cure (ie, durable antiretroviral-free viral control). The need for an effective, safe, scalable, affordable, accessible, and acceptable cure for HIV infection remains a major global priority. The possibilities for cell and gene therapy-based products for an HIV cure are rapidly expanding. In a multi-stakeholder consensus process of clinical experts and civil society, including representatives from low-income and middle-income countries, participants generally agreed on the optimal targets, whereas consensus on the minimal targets was not reached on every parameter. There was less agreement on the minimal targets for ex-vivo than in-vivo therapies given the complexity of ex-vivo interventions. The TPP is planned to be updated at regular intervals. Building a TPP, such as this one, is an important process for stakeholder engagement and aligning ambitions for the development of products that are acceptable to both clinicians and civil society.
Collapse
Affiliation(s)
- Sharon R Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| | | | | | - Lauren Mathae
- Center for Global Health Practice and Impact, Georgetown University, Washington, DC, USA
| | - Kumitaa Theva Das
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Joseph M McCune
- HIV Frontiers, Global Health Accelerator, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Steven G Deeks
- Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, CA, USA
| | - Thumbi Ndung'u
- Africa Health Research Institute, Durban, South Africa; HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA; University College London, London, UK
| |
Collapse
|
8
|
Laher F, Mahlangu N, Sibiya M. Beliefs about HIV cure: A qualitative study of people living with HIV in Soweto, South Africa. South Afr J HIV Med 2025; 26:1644. [PMID: 39967752 PMCID: PMC11830836 DOI: 10.4102/sajhivmed.v26i1.1644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/22/2024] [Indexed: 02/20/2025] Open
Abstract
Background Rare cases of HIV cure exist. Clinical trials of HIV cure are also underway. However, little is documented about how potential cures are perceived by African people living with HIV, although they are key stakeholders. Objectives We explored knowledge, beliefs, and experiences about HIV cure in Soweto, South Africa. Method We conducted qualitative research with five stratified focus groups (N = 49). Consenting adults living with HIV were eligible. Facilitators asked participants about their knowledge of HIV cure, experience of purported cures, and beliefs about cure possibilities. Transcripts from audio recordings were thematically analysed. Results Participants had knowledge of the concept of cure as eradication, not remission. Three main themes emerged about possible HIV cures. Firstly, hope and scepticism: people feared unequal access to technologies. Secondly, cultural and conventional approaches: there were beliefs in traditional healers, scepticism towards culturally purported cures (e.g. imbiza herbal tonic), and a desire for medical cures to obviate pill burdens. Thirdly, anticipated socio-behavioural effects: beliefs existed that cures might improve happiness, reduce emotional burdens of disclosure, facilitate HIV-free generations, increase risk behaviours, and reduce health checks, but not change societal attitudes to HIV. Conclusion In Soweto, South Africa, people living with HIV hope for medical technologies - such as cure and long-acting treatments - to relieve the biopsychosocial burdens of chronic treatment. Despite treatment knowledge, some people try culturally purported cures for HIV. In HIV cure trials, consent language should avoid 'cure' when remission is meant. Care should address pill burden, and counselling should address sex, substances, exercise, and nutrition.
Collapse
Affiliation(s)
- Fatima Laher
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Naledi Mahlangu
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mbalenhle Sibiya
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
9
|
Bowden-Reid E, Moles E, Kelleher A, Ahlenstiel C. Harnessing antiviral RNAi therapeutics for pandemic viruses: SARS-CoV-2 and HIV. Drug Deliv Transl Res 2025:10.1007/s13346-025-01788-x. [PMID: 39833468 DOI: 10.1007/s13346-025-01788-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
Using the knowledge from decades of research into RNA-based therapies, the COVID-19 pandemic response saw the rapid design, testing and production of the first ever mRNA vaccines approved for human use in the clinic. This breakthrough has been a significant milestone for RNA therapeutics and vaccines, driving an exponential growth of research into the field. The development of novel RNA therapeutics targeting high-threat pathogens, that pose a substantial risk to global health, could transform the future of health delivery. In this review, we provide a detailed overview of the two RNA interference (RNAi) pathways and how antiviral RNAi therapies can be used to treat acute or chronic diseases caused by the pandemic viruses SARS-CoV-2 and HIV, respectively. We also provide insights into short-interfering RNA (siRNA) delivery systems, with a focus on how lipid nanoparticles can be functionalized to achieve targeted delivery to specific sites of disease. This review will provide the current developments of SARS-CoV-2 and HIV targeted siRNAs, highlighting strategies to advance the progression of antiviral siRNA along the clinical development pathway.
Collapse
Affiliation(s)
| | - Ernest Moles
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, 2052, Australia.
- Australian Centre for Nanomedicine, Faculty of Engineering, UNSW Sydney, Sydney, 2052, Australia.
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney, 2052, Australia.
- UNSW RNA Institute, UNSW Sydney, Sydney, 2052, Australia.
| | - Anthony Kelleher
- The Kirby Institute, UNSW Sydney, Sydney, 2052, Australia
- UNSW RNA Institute, UNSW Sydney, Sydney, 2052, Australia
| | - Chantelle Ahlenstiel
- The Kirby Institute, UNSW Sydney, Sydney, 2052, Australia.
- UNSW RNA Institute, UNSW Sydney, Sydney, 2052, Australia.
| |
Collapse
|
10
|
Chhabra L, Pandey RK, Kumar R, Sundar S, Mehrotra S. Navigating the Roadblocks: Progress and Challenges in Cell-Based Therapies for Human Immunodeficiency Virus. J Cell Biochem 2025; 126:e30669. [PMID: 39485037 DOI: 10.1002/jcb.30669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/26/2024] [Accepted: 10/11/2024] [Indexed: 11/03/2024]
Abstract
Cell-based therapies represent a major advancement in the treatment and management of HIV/AIDS, with a goal to overcome the limitations of traditional antiretroviral therapy (ART). These innovative approaches not only promise a functional cure by reconstructing the immune landscape but also address the persistent viral reservoirs. For example, stem cell therapies have emerged from the foundational success of allogeneic hematopoietic stem cell transplantation in curing HIV infection in a limited number of cases. B cell therapies make use of genetically modified B cells constitutively expressing broadly neutralizing antibodies (bNAbs) against target viral particles and infected cells. Adoptive cell transfer (ACT), including TCR-T therapy, CAR-T cells, NK-CAR cells, and DC-based therapy, is adapted from cancer immunotherapy and repurposed for HIV eradication. In this review, we summarize the mechanisms through which these engineered cells recognize and destroy HIV-infected cells, the modification strategies, and their role in sustaining remission in the absence of ART. The review also addresses the challenges to cell-based therapies against HIV and discusses the recent advancements aimed at overcoming them.
Collapse
Affiliation(s)
- Lakshay Chhabra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | | | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
11
|
Cleary M, Ndhlovu LC, Sacha JB. Stem cell transplantation and allogeneic immunity: post treatment control or HIV cure? Curr Opin HIV AIDS 2025; 20:86-91. [PMID: 39484913 PMCID: PMC11620935 DOI: 10.1097/coh.0000000000000892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
PURPOSE OF REVIEW Long-lasting HIV remission has been reported in a small group of people with HIV (PWH) following allogenic hematopoietic stem cell transplants (HSCT) for the treatment of hematologic malignancies. While the mechanisms of HIV remission following release from antiretroviral therapy (ART) were not initially known, subsequent findings from clinical cases and preclinical nonhuman primate studies have implicated mechanisms of clearance. Here, we review the six currently published human cases of long-term ART-free HIV remission. RECENT FINDINGS Since the first report of ART-free HIV remission following HSCT, five subsequent cases of HSCT-induced sustained HIV remission have been published. While the pre- and posttransplant treatment conditions vary greatly, all but one received cells from donors homozygous for a 32 bp deletion in the gene that encodes CCR5 ( ccr5Δ32 ), the major HIV coreceptor. Studies in nonhuman primates and the newest published individual suggest that while CCR5 deficiency can protect donor cells from infection early posttransplant, it is not required for long term remission, as ablation of the viral reservoir is likely due to allogeneic immunity mediating a graft-versus-reservoir response. SUMMARY Studies of HSCT in PLWH and simian immunodeficiency virus (SIV)-infected monkeys show that those with durable remission are likely cured, demonstrated by complete ablation of the replication-competent HIV reservoir, gradual loss of anti-HIV immunity, and greater than 5 years of aviremia.
Collapse
Affiliation(s)
- Megan Cleary
- Division of Pathobiology and Immunology, Oregon National Primate Research Center
| | - Lishomwa C Ndhlovu
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York City, New York
| | - Jonah B Sacha
- Division of Pathobiology and Immunology, Oregon National Primate Research Center
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| |
Collapse
|
12
|
Mesquita FS, Li Y, Li JZ. Viral and immune predictors of HIV posttreatment control. Curr Opin HIV AIDS 2025; 20:54-60. [PMID: 39633539 DOI: 10.1097/coh.0000000000000898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
PURPOSE OF REVIEW This review focuses on the viral and immune factors influencing HIV posttreatment control (PTC), a rare condition where individuals maintain viral suppression after discontinuing antiretroviral therapy (ART). RECENT FINDINGS Studies demonstrate that early ART initiation leads to smaller HIV reservoirs and delayed viral rebound in PTCs. Virologically, PTCs harbor smaller HIV reservoirs and show lower levels of reservoir transcriptional activity compared with posttreatment noncontrollers. Immunologically, PTCs exhibit distinct T-cell dynamics, with reduced CD4+ and CD8+ T-cell activation and exhaustion, enhanced natural killer (NK) cell activity, and enhanced proliferative responses of HIV-specific CD8+ T cells post-ART interruption. Additionally, humoral immunity, particularly the development of autologous neutralizing antibodies (aNAbs), plays a role in viral control, though broadly neutralizing antibodies (bnAbs) are rare. SUMMARY The mechanisms behind posttreatment control are multifactorial, involving virological and immunological factors. Early ART initiation, a smaller and less transcriptionally active HIV reservoir, and immune responses including proliferative T-cell activity and NK cell function are key contributors to achieving ART-free HIV remission.
Collapse
Affiliation(s)
- Flavio S Mesquita
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yijia Li
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jonathan Z Li
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
13
|
Yang QW, Yue CL, Chen M, Ling YY, Dong Q, Zhou YX, Cao Y, Ding YX, Zhao X, Huang H, Zhang ZH, Hu L, Xu XH. Daphnetin may protect from SARS-CoV-2 infection by reducing ACE2. Sci Rep 2024; 14:30682. [PMID: 39730426 DOI: 10.1038/s41598-024-79734-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/12/2024] [Indexed: 12/29/2024] Open
Abstract
To combat the SARS-CoV-2 pandemic, innovative prevention strategies are needed, including reducing ACE2 expression on respiratory cells. This study screened approved drugs in China for their ability to downregulate ACE2. Daphnetin (DAP) was found to significantly reduce ACE2 mRNA and protein levels in PC9 cells. DAP exerts its inhibitory effects on ACE2 expression by targeting HIF-1α and JAK2, thereby impeding the transcription of the ACE2 gene. The SARS-CoV-2 pseudovirus infection assay confirmed that DAP-treated PC9 cells exhibited decreased susceptibility to viral infection. At therapeutic doses, DAP effectively lowers ACE2 expression in the respiratory systems of mice and humans. This suggests that DAP, already approved for other conditions, could be a new preventive measure against SARS-CoV-2, offering a cost-effective and accessible way to reduce SARS-CoV-2 spread.
Collapse
Affiliation(s)
- Qian-Wen Yang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Chang-Ling Yue
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Meng Chen
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yun-Yun Ling
- Department of Chemistry, Wannan Medical College, Wuhu, 241002, China
| | - Qi Dong
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Ying-Xin Zhou
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yin Cao
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yan-Xia Ding
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Xu Zhao
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Hai Huang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Zhao-Huan Zhang
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
| | - Lei Hu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China.
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China.
| | - Xiao-Hui Xu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China.
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
14
|
Sáez-Cirión A, Mamez AC, Avettand-Fenoel V, Nabergoj M, Passaes C, Thoueille P, Decosterd L, Hentzien M, Perdomo-Celis F, Salgado M, Nijhuis M, Mélard A, Gardiennet E, Lorin V, Monceaux V, Chapel A, Gourvès M, Lechartier M, Mouquet H, Wensing A, Martinez-Picado J, Yerly S, Rougemont M, Calmy A. Sustained HIV remission after allogeneic hematopoietic stem cell transplantation with wild-type CCR5 donor cells. Nat Med 2024; 30:3544-3554. [PMID: 39222660 PMCID: PMC11645271 DOI: 10.1038/s41591-024-03277-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
HIV cure has been reported for five individuals who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT) with cells from CCR5Δ32 homozygous donors. By contrast, viral rebound has occurred in other people living with HIV who interrupted antiretroviral treatment after undergoing allo-HSCT, with cells mostly from wild-type CCR5 donors. Here we report the case of a male individual who has achieved durable HIV remission following allo-HSCT with cells from an unrelated HLA-matched (9 of 10 matching for HLA-A, HLA-B, HLA-C, HLA-DRB1 and HLA-DQB1 alleles) wild-type CCR5 donor to treat an extramedullary myeloid tumor. To date, plasma viral load has remained undetectable for 32 months after the interruption of antiretroviral treatment. Treatment with ruxolitinib has been maintained during this period to treat chronic graft-versus-host disease. Low levels of proviral DNA were detected sporadically after allo-HSCT, including defective but not intact HIV DNA. No virus could be amplified in cultures of CD4+ T cells obtained after antiretroviral treatment interruption, while CD4+ T cells remained susceptible to HIV-1 infection in vitro. Declines in HIV antibodies and undetectable HIV-specific T cell responses further corroborate the absence of viral rebound after antiretroviral treatment interruption. These results suggest that HIV remission could be achieved in the context of allo-HSCT with wild-type CCR5.
Collapse
Affiliation(s)
- Asier Sáez-Cirión
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France.
- HIV Inflammation and Persistence Unit, Université Paris Cité, Institut Pasteur, Paris, France.
| | - Anne-Claire Mamez
- Division of Hematology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
| | - Véronique Avettand-Fenoel
- Institut Cochin-CNRS 8104/INSERM U1016/Université de Paris, Paris, France
- LI2RSO, Université d'Orléans, Orléans, France
- Virologie, CHU d'Orléans, Orléans, France
| | | | - Caroline Passaes
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France
- HIV Inflammation and Persistence Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Paul Thoueille
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Clinical Pharmacology, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laurent Decosterd
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Maxime Hentzien
- HIV/AIDS Unit, Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Federico Perdomo-Celis
- HIV Inflammation and Persistence Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Maria Salgado
- IrsiCaixa, Badalona, Spain
- Germans Trias i Pujol Research Institute, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Monique Nijhuis
- Translational Virology Research Group, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
- HIV Pathogenesis Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Adeline Mélard
- Institut Cochin-CNRS 8104/INSERM U1016/Université de Paris, Paris, France
| | - Elise Gardiennet
- Institut Cochin-CNRS 8104/INSERM U1016/Université de Paris, Paris, France
| | - Valérie Lorin
- Humoral Immunology Unit, Inserm U1222, Université Paris Cité, Institut Pasteur, Paris, France
| | - Valérie Monceaux
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France
- HIV Inflammation and Persistence Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Anaïs Chapel
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France
- HIV Inflammation and Persistence Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Maël Gourvès
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Marine Lechartier
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Hugo Mouquet
- Humoral Immunology Unit, Inserm U1222, Université Paris Cité, Institut Pasteur, Paris, France
| | - Annemarie Wensing
- Translational Virology Research Group, Department of Global Public Health & Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Ezintsha, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Javier Martinez-Picado
- IrsiCaixa, Badalona, Spain
- Germans Trias i Pujol Research Institute, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
- UVic-UCC, Vic, Spain
- ICREA, Barcelona, Spain
| | - Sabine Yerly
- Laboratory of Virology, Geneva University Hospitals, Geneva, Switzerland
| | | | - Alexandra Calmy
- HIV/AIDS Unit, Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|
15
|
Kelly K, Bekka S, Persaud D. Research Toward a Cure for Perinatal HIV. Clin Perinatol 2024; 51:895-910. [PMID: 39487027 PMCID: PMC11939119 DOI: 10.1016/j.clp.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2024]
Abstract
In virtually all people living with HIV-1 (PLWH), including children, HIV-1 integrates and becomes latent in CD4+ T cells, forming a latent HIV-1 reservoir that current antiretroviral drugs and immune surveillance mechanisms cannot target. This latent infection in CD4+ T cells renders HIV-1 infection lifelong and incurable. Consequently, there is intense research focused on identifying therapeutic strategies to reduce and control the latent reservoir, aiming to avert a lifetime of antiretroviral therapy for PLWH. This review discusses the global efforts for children and adolescents living with HIV-1.
Collapse
Affiliation(s)
- Kristen Kelly
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Ross 1133, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Soumia Bekka
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Ross 1133, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Deborah Persaud
- Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins University School of Medicine, 1170, 720 Rutland Avenue, Baltimore, MD 21205, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
16
|
Guo Q, Parikh K, Zhang J, Brinkley A, Chen G, Jakramonpreeya N, Zhen A, An DS. Anti-HIV-1 HSPC-based gene therapy with safety kill switch to defend against and attack HIV-1 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623476. [PMID: 39605384 PMCID: PMC11601352 DOI: 10.1101/2024.11.13.623476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Hematopoietic stem/progenitor cell (HSPC)-based anti-HIV-1 gene therapy holds promise to provide life-long remission following a single treatment. Here we report a multi-pronged anti-HIV-1 HSPC-based gene therapy designed to defend against and attack HIV-1 infection. We developed a lentiviral vector capable of co-expressing three anti-HIV-1 genes. Two are designed to prevent infection, including a short-hairpin RNA (CCR5sh1005) to knock down HIV-1 co-receptor CCR5 and a membrane anchored HIV-1 fusion inhibitor (C46). The third gene is a CD4-based chimeric antigen receptor (CAR) designed to attack HIV-1 infected cells. Our vector also includes a non-signaling truncated human epidermal growth factor receptor (huEGFRt) which acts as a negative selection-based safety kill switch against transduced cells. Anti-HIV-1 vector-transduced human CD34+ HSPC efficiently reconstituted multi-lineage human hematopoietic cells in humanized bone marrow/liver/thymus (huBLT) mice. HIV-1 viral load was significantly reduced (1-log fold reduction, p <0.001) in transplanted huBLT mice. Anti-huEGFR monoclonal antibody Cetuximab (CTX) administration significantly reduced huEGFRt+ vector-modified cells (>4-fold reduction, p <0.01) in huBLT mice. These results demonstrate that our strategy is highly effective for HIV-1 inhibition, and that CTX-mediated negative selection can deplete anti-HIV-1 vector-modified cells in the event of unwanted adverse effects in huBLT mice.
Collapse
Affiliation(s)
- Qi Guo
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
- Shanghai Key Laboratory of Tumor System Regulation and Clinical Translation, Jiading Branch, Renji Hospital, Shanghai Cancer Institute, Shanghai, China, 201800
| | - Keval Parikh
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
| | - Jian Zhang
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
| | - Alexander Brinkley
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
| | - Grace Chen
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, USA, 90095
| | - Natnicha Jakramonpreeya
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Anjie Zhen
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA, 90095
| | - Dong Sung An
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
| |
Collapse
|
17
|
Long X, Liu G, Liu X, Zhang C, Shi L, Zhu Z. Identifying the HIV-Resistance-Related Factors and Regulatory Network via Multi-Omics Analyses. Int J Mol Sci 2024; 25:11757. [PMID: 39519306 PMCID: PMC11546959 DOI: 10.3390/ijms252111757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/04/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
For research on HIV/AIDS, it is important to elucidate the complex viral-host interaction, host dependency factors (HDFs), and restriction factors. However, the regulatory network of HIV-resistance-related factors remains not well understood. Therefore, we integrated four publicly available HIV-related transcriptome datasets, along with three datasets on HIV-infection-related DNA methylation, miRNA, and ChIP-seq, to predict the factors influencing HIV resistance and infection. Our approach involved differential analysis, functional annotation, and protein-protein interaction network analysis. Through comprehensive analyses, we identified 25 potential HIV-resistance-related genes (including shared EGF) and 24 HIV-infection-related hub genes (including shared JUN). Additionally, we pinpointed five key differentially methylated genes, five crucial differentially expressed microRNAs, and five significant pathways associated with HIV resistance. We mapped the potential regulatory pathways involving these HIV-resistance-related factors. Among the predicted factors, RHOA, RAD51, GATA1, IRF4, and CXCL8 have been validated as HDFs or restriction factors. The identified factors, such as JUN, EGF, and PLEK, are potential HDFs or restriction factors. This study uncovers the gene signatures and regulatory networks associated with HIV-1 resistance, suggesting potential targets for the development of new therapies against HIV/AIDS.
Collapse
Affiliation(s)
| | | | | | | | - Lei Shi
- School of Life Sciences, Chongqing University, No. 55 Daxuecheng South Road, Shapingba, Chongqing 401331, China; (X.L.); (G.L.); (X.L.); (C.Z.)
| | - Zhenglin Zhu
- School of Life Sciences, Chongqing University, No. 55 Daxuecheng South Road, Shapingba, Chongqing 401331, China; (X.L.); (G.L.); (X.L.); (C.Z.)
| |
Collapse
|
18
|
Toner K, McCann CD, Bollard CM. Applications of cell therapy in the treatment of virus-associated cancers. Nat Rev Clin Oncol 2024; 21:709-724. [PMID: 39160243 DOI: 10.1038/s41571-024-00930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 08/21/2024]
Abstract
A diverse range of viruses have well-established roles as the primary driver of oncogenesis in various haematological malignancies and solid tumours. Indeed, estimates suggest that approximately 1.5 million patients annually are diagnosed with virus-related cancers. The predominant human oncoviruses include Epstein-Barr virus (EBV), Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis B and C viruses (HBV and HCV), human papillomavirus (HPV), human T-lymphotropic virus type 1 (HTLV1), and Merkel cell polyomavirus (MCPyV). In addition, although not inherently oncogenic, human immunodeficiency virus (HIV) is associated with immunosuppression that contributes to the development of AIDS-defining cancers (specifically, Kaposi sarcoma, aggressive B cell non-Hodgkin lymphoma and cervical cancer). Given that an adaptive T cell-mediated immune response is crucial for the control of viral infections, increasing research is being focused on evaluating virus-specific T cell therapies for the treatment of virus-associated cancers. In this Review, we briefly outline the roles of viruses in the pathogenesis of these malignancies before describing progress to date in the field of virus-specific T cell therapy and evaluating the potential utility of these therapies to treat or possibly even prevent virus-related malignancies.
Collapse
Affiliation(s)
- Keri Toner
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Chase D McCann
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA.
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
19
|
Manzanares M, Ramos-Martín F, Rodríguez-Mora S, Casado-Fernández G, Sánchez-Menéndez C, Simón-Rueda A, Mateos E, Cervero M, Spivak AM, Planelles V, Torres M, García-Gutiérrez V, Coiras M. Sustained antiviral response against in vitro HIV-1 infection in peripheral blood mononuclear cells from people with chronic myeloid leukemia treated with ponatinib. Front Pharmacol 2024; 15:1426974. [PMID: 39380908 PMCID: PMC11460598 DOI: 10.3389/fphar.2024.1426974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
HIV-1 infection cannot be cured due to long-lived viral reservoirs formed by latently infected CD4+ T cells. "Shock and Kill" strategy has been considered to eliminate the viral reservoir and achieve a functional cure but the stimulation of cytotoxic immunity is necessary. Ponatinib is a tyrosine kinase inhibitor (TKI) clinically used against chronic myeloid leukemia (CML) that has demonstrated to be effective against HIV-1 infection in vitro. Several TKIs may induce a potent cytotoxic response against cancer cells that makes possible to discontinue treatment in people with CML who present long-term deep molecular response. In this longitudinal study, we analyzed the capacity of ponatinib to induce an antiviral response against HIV-1 infection in peripheral blood mononuclear cells (PBMCs) obtained from people with CML previously treated with imatinib for a median of 10 years who changed to ponatinib for 12 months to boost the anticancer response before discontinuing any TKI as part of the clinical trial NCT04043676. Participants were followed-up for an additional 12 months in the absence of treatment. PBMCs were obtained at different time points and then infected in vitro with HIV-1. The rate of infection was determined by quantifying the intracellular levels of p24-gag in CD4+ T cells. The levels of p24-gag+ CD4+ T-cells were lower when these cells were obtained during and after treatment with ponatinib in comparison with those obtained during treatment with imatinib. Cytotoxicity of PBMCs against HIV-infected target cells was significantly higher during treatment with ponatinib than during treatment with imatinib, and it was maintained at least 12 months after discontinuation. There was a significant negative correlation between the lower levels of p24-gag+ CD4+ T-cells and the higher cytotoxicity induced by PBMCs when cells were obtained during and after treatment with ponatinib. This cytotoxic immunity was mostly based on higher levels of Natural Killer and Tγδ cells seemingly boosted by ponatinib. In conclusion, transient treatment with immunomodulators like ponatinib along with ART could be explored to boost the antiviral activity of cytotoxic cells and contribute to the elimination of HIV-1 reservoir.
Collapse
Affiliation(s)
- Mario Manzanares
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- PhD Program in Biomedical Sciences and Public Health, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Fernando Ramos-Martín
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Rodríguez-Mora
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Guiomar Casado-Fernández
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Sciences, Universidad de Alcalá, Madrid, Spain
| | - Clara Sánchez-Menéndez
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- PhD Program in Biomedical Sciences and Public Health, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alicia Simón-Rueda
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- PhD Program in Biomedical Sciences and Public Health, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Elena Mateos
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel Cervero
- Internal Medicine Service, Hospital Universitario Severo Ochoa, Madrid, Spain
- School of Medicine, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Adam M. Spivak
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt LakeCity, UT, United States
| | - Vicente Planelles
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt LakeCity, UT, United States
| | - Montserrat Torres
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Valentín García-Gutiérrez
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Mayte Coiras
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
20
|
Salgado M, Migueles SA, Yu XG, Martinez-Picado J. Exceptional, naturally occurring HIV-1 control: Insight into a functional cure. MED 2024; 5:1071-1082. [PMID: 39013460 PMCID: PMC11411266 DOI: 10.1016/j.medj.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/30/2024] [Accepted: 06/19/2024] [Indexed: 07/18/2024]
Abstract
Exceptional elite controllers represent an extremely rare group of people with HIV-1 (PWH) who exhibit spontaneous, high-level control of viral replication below the limits of detection in sensitive clinical monitoring assays and without disease progression in the absence of antiretroviral therapy for prolonged periods, frequently exceeding 25 years. Here, we discuss the different cases that have been reported in the scientific literature, their unique genetic, virological, and immunological characteristics, and their relevance as the best model for the functional cure of HIV-1.
Collapse
Affiliation(s)
- María Salgado
- IrsiCaixa Immunopathology Research Institute, 08916 Badalona, Spain; CIBERINFEC, 28029 Madrid, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Stephen A Migueles
- Laboratory of Immunoregulation, Division of Intramural Research, and Collaborative Clinical Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Xu G Yu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Javier Martinez-Picado
- IrsiCaixa Immunopathology Research Institute, 08916 Badalona, Spain; CIBERINFEC, 28029 Madrid, Spain; University of Vic - Central University of Catalonia (UVic-UCC), 08500 Vic, Spain; Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
21
|
Yokota T, Kurosawa S, Yoshimura Y, Bingo M, Yamaguchi T, Takada Y, So Y, Miyata N, Nakayama H, Sakurai A, Sato K, Ito C, Aisa Y, Nakazato T. Pioneering cord blood transplantation in relapsed/refractory HIV-related lymphoma: a case study with concurrent intramuscular antiretroviral therapy. Int J Infect Dis 2024; 146:107124. [PMID: 38838848 DOI: 10.1016/j.ijid.2024.107124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024] Open
Abstract
A 44-year-old HIV-positive man diagnosed with diffuse large B-cell lymphoma in 2021 achieved complete remission with six cycles of R-CHOP therapy but had a relapse in November 2022. ESHAP therapy failed to induce remission, leading to complete remission with four cycles of Pola-BR therapy. Post-failure of autologous stem cell harvest, cord blood transplantation (CBT) was performed in June 2023. Notably, this case used recently approved intramuscular antiretroviral therapy (ART) with cabotegravir and rilpivirine, addressing gastrointestinal complications during CBT. This innovative use of intramuscular ART in the treatment of malignancy represents a first in the field, offering a pioneering approach to HIV-related lymphoma.
Collapse
Affiliation(s)
- Takako Yokota
- Department of Hematology, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| | - Shuhei Kurosawa
- Department of Hematology, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan.
| | - Yukihiro Yoshimura
- Division of Infectious Disease, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| | - Masato Bingo
- Department of Laboratory Medicine, Tokyo Medical University Hospital, Shinjuku, Tokyo, Japan
| | - Tomoko Yamaguchi
- Department of Laboratory Medicine, Tokyo Medical University Hospital, Shinjuku, Tokyo, Japan
| | - Yusuke Takada
- Department of Hematology, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| | - Yuna So
- Division of Infectious Disease, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| | - Nobuyuki Miyata
- Division of Infectious Disease, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| | - Hitomi Nakayama
- Department of Hematology, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| | - Aki Sakurai
- Department of Hematology, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| | - Kosuke Sato
- Division of Infectious Disease, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| | - Chisako Ito
- Department of Hematology, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| | - Yoshinobu Aisa
- Department of Hematology, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| | - Tomonori Nakazato
- Department of Hematology, Yokohama Municipal Citizen's Hospital, Yokohama, Kanagawa, Japan
| |
Collapse
|
22
|
Zhou M, Yang T, Yuan M, Li X, Deng J, Wu S, Zhong Z, Lin Y, Zhang W, Xia B, Wu Y, Wang L, Chen T, Liu R, Pan T, Ma X, Li L, Liu B, Zhang H. ORC1 enhances repressive epigenetic modifications on HIV-1 LTR to promote HIV-1 latency. J Virol 2024; 98:e0003524. [PMID: 39082875 PMCID: PMC11334468 DOI: 10.1128/jvi.00035-24] [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: 02/21/2024] [Accepted: 06/21/2024] [Indexed: 08/21/2024] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) reservoir consists of latently infected cells which present a major obstacle to achieving a functional cure for HIV-1. The formation and maintenance of HIV-1 latency have been extensively studied, and latency-reversing agents (LRAs) that can reactivate latent HIV-1 by targeting the involved host factors are developed; however, their clinical efficacies remain unsatisfactory. Therefore, it is imperative to identify novel targets for more potential candidates or better combinations for LRAs. In this study, we utilized CRISPR affinity purification in situ of regulatory elements system to screen for host factors associated with the HIV-1 long terminal repeat region that could potentially be involved in HIV-1 latency. We successfully identified that origin recognition complex 1 (ORC1), the largest subunit of the origin recognition complex, contributes to HIV-1 latency in addition to its function in DNA replication initiation. Notably, ORC1 is enriched on the HIV-1 promoter and recruits a series of repressive epigenetic elements, including DNMT1 and HDAC1/2, and histone modifiers, such as H3K9me3 and H3K27me3, thereby facilitating the establishment and maintenance of HIV-1 latency. Moreover, the reactivation of latent HIV-1 through ORC1 depletion has been confirmed across various latency cell models and primary CD4+ T cells from people living with HIV-1. Additionally, we comprehensively validated the properties of liquid-liquid phase separation (LLPS) of ORC1 from multiple perspectives and identified the key regions that promote the formation of LLPS. This property is important for the recruitment of ORC1 to the HIV-1 promoter. Collectively, these findings highlight ORC1 as a potential novel target implicated in HIV-1 latency and position it as a promising candidate for the development of novel LRAs. IMPORTANCE Identifying host factors involved in maintaining human immunodeficiency virus type 1 (HIV-1) latency and understanding their mechanisms prepares the groundwork to discover novel targets for HIV-1 latent infection and provides further options for the selection of latency-reversing agents in the "shock" strategy. In this study, we identified a novel role of the DNA replication factor origin recognition complex 1 (ORC1) in maintaining repressive chromatin structures surrounding the HIV-1 promoter region, thereby contributing to HIV-1 latency. This discovery expands our understanding of the non-replicative functions of the ORC complex and provides a potential therapeutic strategy for HIV-1 cure.
Collapse
Affiliation(s)
- Mo Zhou
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Center for Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Tao Yang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ming Yuan
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xinyu Li
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jieyi Deng
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shiyu Wu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhihan Zhong
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingtong Lin
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wanying Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Baijin Xia
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Science), Guangzhou, China
| | - Yating Wu
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Science), Guangzhou, China
| | - Lilin Wang
- Shenzhen Blood Center, Shenzhen, Guangdong, China
| | - Tao Chen
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Ruxin Liu
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ting Pan
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiancai Ma
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Linghua Li
- Center for Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Bingfeng Liu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
23
|
Rubinstein PG, Galvez C, Ambinder RF. Hematopoietic stem cell transplantation and cellular therapy in persons living with HIV. Curr Opin Infect Dis 2024; 37:254-263. [PMID: 38820072 DOI: 10.1097/qco.0000000000001022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
PURPOSE OF REVIEW Summarize the latest research of both stem cell transplantation and cellular therapy and present the implications with respect to persons with HIV (PWH), hematologic malignancies, and HIV-1 cure. RECENT FINDINGS Allogeneic (alloSCT) and autologous (autoSCT) stem cell transplantation have been shown to be well tolerated and effective regardless of HIV-1 status. AlloSCT leads to a decrease in the HIV-1 latently infected reservoir orders of magnitude below that achieved with antiretroviral therapy (ART) alone. Utilization of CCR5Δ2/Δ32 donors in an alloSCT has resulted in HIV-1 cures. In the last 12 months, three cases of cure have been published, giving further insight into the conditions required for HIV-1 control. Other advances in the treatment of hematological cancers include chimeric antigen receptor T-cell (CART) therapy, which are active in PWH with lymphoma. SUMMARY Here we discuss the advances in SCT and cellular therapy in PWH and cancer. Additionally, we discuss how these technologies are being utilized to achieve HIV-1 cure.
Collapse
Affiliation(s)
- Paul G Rubinstein
- Section of Hematology/Oncology, Department of Medicine, University of Illinois
- Ruth M. Rothstein CORE Center
- Section of Hematology/Oncology, Department of Medicine, Cook County Health and Hospital Systems (Cook County Hospital), Chicago, Illinois
| | - Carlos Galvez
- Section of Hematology/Oncology, Department of Medicine, University of Illinois
| | - Richard F Ambinder
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
24
|
De Nicolò A, Palermiti A, Dispinseri S, Marchetti G, Trunfio M, De Vivo E, D'Avolio A, Muscatello A, Gori A, Rusconi S, Bruzzesi E, Gabrieli A, Bernasconi DP, Bandera A, Nozza S, Calcagno A. Plasma, intracellular and lymph node antiretroviral concentrations and HIV DNA change during primary HIV infection: Results from the INACTION P25 study. Int J Antimicrob Agents 2024; 64:107200. [PMID: 38768738 DOI: 10.1016/j.ijantimicag.2024.107200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Despite its effectiveness, combination antiretroviral treatment (cART) has a limited effect on HIV DNA reservoir, which establishes early during primary HIV infection (PHI) and is maintained by latency, homeostatic T-cells proliferation, and residual replication. This limited effect can be associated with low drug exposure in lymphoid tissues and/or suboptimal adherence to antiretroviral drugs (ARVs). The aim of this study was to assess ARV concentrations in plasma, peripheral blood mononuclear cells (PBMCs) and lymph nodes (LNs), and their association to HIV RNA and HIV DNA decay during PHI. Participants were randomised to receive standard doses of darunavir/cobicistat (Arm I), dolutegravir (Arm II) or both (Arm III), with a backbone of tenofovir alafenamide and emtricitabine. Total HIV DNA was measured using digital-droplet PCR in PBMCs at baseline, 12 and 48 weeks. Drug concentrations in plasma and PBMCs were determined at 2, 12 and 48 weeks (LNs at 12 weeks) by UHPLC-MS/MS. Seventy-two participants were enrolled, mostly male (n=68), with a median age of 34 years and variable Fiebig stages (V-VI 57.7%, I-II 23.9%, and III-IV 18.3%). Twenty-six patients were assigned to Arm I, 27 to Arm II and 19 to Arm III. After 48 weeks, most patients had undetectable viremia, with minor differences in HIV RNA decay between arms. Patients with Fiebig I-II showed faster HIV RNA and HIV DNA decay. Intracellular tissue penetration was high for nucleoside analogues and low-moderate for darunavir and dolutegravir. Only tenofovir diphosphate concentrations in PBMCs showed correlation with HIV DNA decay. Overall, these results indicate that the timing of treatment initiation and intracellular tenofovir penetration are primary and secondary factors, respectively, affecting HIV reservoir.
Collapse
Affiliation(s)
- Amedeo De Nicolò
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Turin.
| | - Alice Palermiti
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Turin
| | | | - Giulia Marchetti
- Clinic of Infectious Diseases, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan
| | - Mattia Trunfio
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Turin
| | - Elisa De Vivo
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Turin
| | - Antonio D'Avolio
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Turin
| | - Antonio Muscatello
- Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan
| | - Andrea Gori
- Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan
| | - Stefano Rusconi
- UOC Malattie Infettive, Ospedale Civile di Legnano, ASST Ovest Milanese, Legnano; University of Milan, Milan
| | | | - Arianna Gabrieli
- Dipartimento di Scienze Biomediche e Cliniche, Ospedale L Sacco, Milan
| | - Davide Paolo Bernasconi
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre - B4 School of Medicine and Surgery, University of Milano-Bicocca, Monza
| | | | - Silvia Nozza
- Department of Infectious Diseases, IRCCS Ospedale san Raffaele, Milan, Italy
| | - Andrea Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Turin
| |
Collapse
|
25
|
Rojekar S, Gholap AD, Togre N, Bhoj P, Haeck C, Hatvate N, Singh N, Vitore J, Dhoble S, Kashid S, Patravale V. Current status of mannose receptor-targeted drug delivery for improved anti-HIV therapy. J Control Release 2024; 372:494-521. [PMID: 38849091 DOI: 10.1016/j.jconrel.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
In the pursuit of achieving better therapeutic outcomes in the treatment of HIV, innovative drug delivery strategies have been extensively explored. Mannose receptors, which are primarily found on macrophages and dendritic cells, offer promising targets for drug delivery due to their involvement in HIV pathogenesis. This review article comprehensively evaluates recent drug delivery system advancements targeting the mannose receptor. We have systematically described recent developments in creating and utilizing drug delivery platforms, including nanoparticles, liposomes, micelles, noisomes, dendrimers, and other nanocarrier systems targeted at the mannose receptor. These strategies aim to enhance drug delivery specificity, bioavailability, and therapeutic efficacy while decreasing off-target effects and systemic toxicity. Furthermore, the article delves into how mannose receptors and HIV interact, highlighting the potential for exploiting this interaction to enhance drug delivery to infected cells. The review covers essential topics, such as the rational design of nanocarriers for mannose receptor recognition, the impact of physicochemical properties on drug delivery performance, and how targeted delivery affects the pharmacokinetics and pharmacodynamics of anti-HIV agents. The challenges of these novel strategies, including immunogenicity, stability, and scalability, and future research directions in this rapidly growing area are discussed. The knowledge synthesis presented in this review underscores the potential of mannose receptor-based targeted drug delivery as a promising avenue for advancing HIV treatment. By leveraging the unique properties of mannose receptors, researchers can design drug delivery systems that cater to individual needs, overcome existing limitations, and create more effective and patient-friendly treatments in the ongoing fight against HIV/AIDS.
Collapse
Affiliation(s)
- Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Namdev Togre
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Priyanka Bhoj
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Clement Haeck
- Population Council, , Center for Biomedical Research, 1230 York Avenue, New York, NY 10065, USA
| | - Navnath Hatvate
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Nidhi Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata 700054, India
| | - Jyotsna Vitore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Sagar Dhoble
- Department of Pharmacology and Toxicology, R. K. Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Snehal Kashid
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
26
|
Ni F, Hu K, Li M, Yang M, Xiao Y, Fu M, Zhu Z, Liu Y, Hu Q. Tat-dependent conditionally replicating adenoviruses expressing diphtheria toxin A for specifically killing HIV-1-infected cells. Mol Ther 2024; 32:2316-2327. [PMID: 38734901 PMCID: PMC11286811 DOI: 10.1016/j.ymthe.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/19/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024] Open
Abstract
HIV-1 infection remains a public health problem with no cure. Although antiretroviral therapy (ART) is effective for suppressing HIV-1 replication, it requires lifelong drug administration due to a stable reservoir of latent proviruses and may cause serious side effects and drive the emergence of drug-resistant HIV-1 variants. Gene therapy represents an alternative approach to overcome the limitations of conventional treatments against HIV-1 infection. In this study, we constructed and investigated the antiviral effects of an HIV-1 Tat-dependent conditionally replicating adenovirus, which selectively replicates and expresses the diphtheria toxin A chain (Tat-CRAds-DTA) in HIV-1-infected cells both in vitro and in vivo. We found that Tat-CRAds-DTA could specifically induce cell death and inhibit virus replication in HIV-1-infected cells mediated by adenovirus proliferation and DTA expression. A low titer of progeny Tat-CRAds-DTA was also detected in HIV-1-infected cells. In addition, Tat-CRAds-DTA showed no apparent cytotoxicity to HIV-1-negative cells and demonstrated significant therapeutic efficacy against HIV-1 infection in a humanized mouse model. The findings in this study highlight the potential of Tat-CRAds-DTA as a new gene therapy for the treatment of HIV-1 infection.
Collapse
Affiliation(s)
- Fengfeng Ni
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China; Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Kai Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China
| | - Miaomiao Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China; Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Mengshi Yang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China
| | - Yingying Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China
| | - Ming Fu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China
| | - Zhiyuan Zhu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China; Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Yalan Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China; Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China; Hubei Jiangxia Laboratory, Wuhan 430200, P.R. China.
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China; Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P.R. China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, P.R. China.
| |
Collapse
|
27
|
Ropa J, Van't Hof W. The fulfilled promise and unmet potential of umbilical cord blood. Curr Opin Hematol 2024; 31:168-174. [PMID: 38602152 DOI: 10.1097/moh.0000000000000817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
PURPOSE OF REVIEW Here, we review classic and emerging uses of umbilical cord blood and highlight strategies to improve its utility, focusing on selection of the appropriate units and cell types for the intended applications. RECENT LITERATURE Recent studies have shown advancements in cord blood cell utility in a variety of cellular therapies and have made strides in elucidating manners to select the best units for therapy and target new ways to improve the various cell subpopulations for their respective applications. SUMMARY Umbilical cord blood is a proven source of cells for hematopoietic cell transplantation and research and is an important potential source for additional cellular therapies. However, cord blood utility is limited by low "doses" of potent cells that can be obtained from individual units, a limitation that is specific to cord blood as a donor source. In addition to traditional CD34 + progenitor cells, cord blood lymphocytes are being pursued as therapeutic entities with their own unique properties and characteristics. Thus, selection of ideal units depends on the intended therapeutic entity and target, and identification of differential potency parameters is critical to drive effective banking strategies accommodating successful clinical use of cord blood in broader cell therapy settings.
Collapse
Affiliation(s)
- James Ropa
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | | |
Collapse
|
28
|
Su H, Mueller A, Goldstein H. Recent advances on anti-HIV chimeric antigen receptor-T-cell treatment to provide sustained HIV remission. Curr Opin HIV AIDS 2024; 19:169-178. [PMID: 38695148 PMCID: PMC11981014 DOI: 10.1097/coh.0000000000000858] [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: 06/07/2024]
Abstract
PURPOSE OF REVIEW Successful sustained remission of HIV infection has been achieved after CCR5Δ32/Δ32 allogeneic hematopoietic stem cell transplantation for treatment of leukemia in a small cohort of people living with HIV (PLWH). This breakthrough demonstrated that the goal of curing HIV was achievable. However, the high morbidity and mortality associated with bone marrow transplantation limits the routine application of this approach and provides a strong rationale for pursuing alternative strategies for sustained long-term antiretroviral therapy (ART)-free HIV remission. Notably, long-term immune-mediated control of HIV replication observed in elite controllers and posttreatment controllers suggests that potent HIV-specific immune responses could provide sustained ART-free remission in PLWH. The capacity of chimeric antigen receptor (CAR)-T cells engineered to target malignant cells to induce remission and cure in cancer patients made this an attractive approach to provide PLWH with a potent HIV-specific immune response. Here, we review the recent advances in the design and application of anti-HIV CAR-T-cell therapy to provide a functional HIV cure. RECENT FINDINGS HIV reservoirs are established days after infection and persist through clonal expansion of infected cells. The continuous interaction between latently infected cells and the immune system shapes the landscape of HIV latency and likely contributes to ART-free viral control in elite controllers. CAR-T cells can exhibit superior antiviral activity as compared with native HIV-specific T cells, particularly because they can be engineered to have multiple HIV specificities, resistance to HIV infection, dual costimulatory signaling, immune checkpoint inhibitors, stem cell derivation, CMV TCR coexpression, and tissue homing ligands. These modifications can significantly improve the capacities of anti-HIV CAR-T cells to prevent viral escape, resist HIV infection, and enhance cytotoxicity, persistence, and tissue penetration. Collectively, these novel modifications of anti-HIV CAR-T cell design have increased their capacity to control HIV infection. SUMMARY Anti-HIV CAR-T cells can be engineered to provide potent and sustained in-vitro and in-vivo antiviral function. The combination of anti-HIV CAR-T cells with other immunotherapeutics may contribute to long-term HIV remission in PLWH.
Collapse
Affiliation(s)
- Hang Su
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, U.S.A
| | - April Mueller
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, U.S.A
| | - Harris Goldstein
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, U.S.A
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, 10461, U.S.A
| |
Collapse
|
29
|
Buck AM, LaFranchi BH, Henrich TJ. Gaining momentum: stem cell therapies for HIV cure. Curr Opin HIV AIDS 2024; 19:194-200. [PMID: 38686850 PMCID: PMC11155292 DOI: 10.1097/coh.0000000000000859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
PURPOSE OF REVIEW Durable HIV-1 remission has been reported in a person who received allogeneic stem cell transplants (SCTs) involving CCR5 Δ32/Δ32 donor cells. Much of the reduction in HIV-1 burden following allogeneic SCT with or without donor cells inherently resistant to HIV-1 infection is likely due to cytotoxic graft-versus-host effects on residual recipient immune cells. Nonetheless, there has been growing momentum to develop and implement stem cell therapies that lead to durable long-term antiretroviral therapy (ART)-free remission without the need for SCT. RECENT FINDINGS Most current research leverages gene editing techniques to modify hematopoietic stem cells which differentiate into immune cells capable of harboring HIV-1. Approaches include targeting genes that encode HIV-1 co-receptors using Zinc Finger Nucleases (ZFN) or CRISPR-Cas-9 to render a pool of adult or progenitor cells resistant to de-novo infection. Other strategies involve harnessing multipotent mesenchymal stromal cells to foster immune environments that can more efficiently recognize and target HIV-1 while promoting tissue homeostasis. SUMMARY Many of these strategies are currently in a state of infancy or adolescence; nonetheless, promising preclinical and first-in-human studies have been performed, providing further rationale to focus resources on stem cell therapies.
Collapse
Affiliation(s)
- Amanda M Buck
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, USA
| | | | | |
Collapse
|
30
|
Shalekoff S, Dias BDC, Loubser S, Strehlau R, Kuhn L, Tiemessen CT. Higher CCR5 density on CD4 + T-cells in mothers and infants is associated with increased risk of in-utero HIV-1 transmission. AIDS 2024; 38:945-954. [PMID: 38329228 PMCID: PMC11064911 DOI: 10.1097/qad.0000000000003857] [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: 10/19/2023] [Revised: 01/17/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVE CCR5-tropic viruses are preferentially transmitted during perinatal HIV-1 infection. CCR5 density on CD4 + T-cells likely impacts susceptibility to HIV-1 infection. DESIGN Fifty-two mother-infant dyads were enrolled. All mothers were living with HIV-1, 27 of the infants acquired HIV-1 in utero and 25 infants remained uninfected. METHODS CCR5 density, together with frequencies of CD4 + and CD8 + T-cells expressing immune activation (CCR5, ICOS and HLA-DR) and immune checkpoint (TIGIT and PD-1) markers, were measured in whole blood from the dyads close to delivery. RESULTS Compared with mothers who did not transmit, mothers who transmitted HIV-1 had less exposure to ART during pregnancy ( P = 0.015) and higher plasma viral load close to delivery ( P = 0.0005). These mothers, additionally, had higher CCR5 density on CD4 + and CD8 + T-cells and higher frequencies of CCR5, ICOS and TIGIT-expressing CD8 + T-cells. Similarly, compared with infants without HIV-1, infants with HIV-1 had higher CCR5 density on CD4 + and CD8 + T-cells and higher frequencies of CCR5, TIGIT, and PD-1-expressing CD4 + and CD8 + T-cells as well as higher frequencies of HLA-DR-expressing CD8 + T-cells. CCR5 density on maternal CD4 + T-cells remained significantly associated with transmission after adjusting for maternal viral load and CD4 + T cell counts. Mother-infant dyads with shared high CCR5 density phenotypes had the highest risk of transmission/acquisition of infection compared with dyads with shared low-CCR5 density phenotypes. CONCLUSION This study provides strong evidence of a protective role for a combined mother-infant low CD4 + T-cell CCR5 density phenotype in in-utero transmission/acquisition of HIV-1.
Collapse
Affiliation(s)
- Sharon Shalekoff
- Centre for HIV and STIs, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bianca Da Costa Dias
- Centre for HIV and STIs, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shayne Loubser
- Centre for HIV and STIs, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Renate Strehlau
- VIDA Nkanyezi Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Louise Kuhn
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Caroline T. Tiemessen
- Centre for HIV and STIs, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
31
|
Armani-Tourret M, Bone B, Tan TS, Sun W, Bellefroid M, Struyve T, Louella M, Yu XG, Lichterfeld M. Immune targeting of HIV-1 reservoir cells: a path to elimination strategies and cure. Nat Rev Microbiol 2024; 22:328-344. [PMID: 38337034 PMCID: PMC11131351 DOI: 10.1038/s41579-024-01010-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
Successful approaches for eradication or cure of HIV-1 infection are likely to include immunological mechanisms, but remarkably little is known about how human immune responses can recognize and interact with the few HIV-1-infected cells that harbour genome-intact viral DNA, persist long term despite antiretroviral therapy and represent the main barrier to a cure. For a long time regarded as being completely shielded from host immune responses due to viral latency, these cells do, on closer examination with single-cell analytic techniques, display discrete footprints of immune selection, implying that human immune responses may be able to effectively engage and target at least some of these cells. The failure to eliminate rebound-competent virally infected cells in the majority of persons likely reflects the evolution of a highly selected pool of reservoir cells that are effectively camouflaged from immune recognition or rely on sophisticated approaches for resisting immune-mediated killing. Understanding the fine-tuned interplay between host immune responses and viral reservoir cells will help to design improved interventions that exploit the immunological vulnerabilities of HIV-1 reservoir cells.
Collapse
Affiliation(s)
- Marie Armani-Tourret
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Benjamin Bone
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Toong Seng Tan
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Weiwei Sun
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Maxime Bellefroid
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Tine Struyve
- HIV Cure Research Center, Ghent University, Ghent, Belgium
| | - Michael Louella
- Community Advisory Board, Delaney AIDS Research Enterprise (DARE), San Francisco, CA, USA
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Xu G Yu
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Mathias Lichterfeld
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
32
|
Salgado M, Gálvez C, Nijhuis M, Kwon M, Cardozo-Ojeda EF, Badiola J, Gorman MJ, Huyveneers LEP, Urrea V, Bandera A, Jensen BEO, Vandekerckhove L, Jurado M, Raj K, Schulze Zur Wiesch J, Bailén R, Eberhard JM, Nabergoj M, Hütter G, Saldaña-Moreno R, Oldford S, Barrett L, Ramirez MLM, Garba S, Gupta RK, Revollo B, Ferra-Coll C, Kuball J, Alter G, Sáez-Cirión A, Diez-Martin JL, Duke ER, Schiffer JT, Wensing A, Martinez-Picado J. Dynamics of virological and immunological markers of HIV persistence after allogeneic haematopoietic stem-cell transplantation in the IciStem cohort: a prospective observational cohort study. Lancet HIV 2024; 11:e389-e405. [PMID: 38816141 PMCID: PMC11417461 DOI: 10.1016/s2352-3018(24)00090-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Allogeneic haematopoietic stem-cell transplantation (allo-HSCT) markedly reduces HIV reservoirs, but the mechanisms by which this occurs are only partly understood. In this study, we aimed to describe the dynamics of virological and immunological markers of HIV persistence after allo-HSCT. METHODS In this prospective observational cohort study, we analysed the viral reservoir and serological dynamics in IciStem cohort participants with HIV who had undergone allo-HSCT and were receiving antiretroviral therapy, ten of whom had received cells from donors with the CCR5Δ32 mutation. Participants from Belgium, Canada, Germany, Italy, the Netherlands, Spain, Switzerland, and the UK were included in the cohort both prospectively and retrospectively between June 1, 2014 and April 30, 2019. In the first 6 months after allo-HSCT, participants had monthly assessments, with annual assessments thereafter, with the protocol tailored to accommodate for the individual health status of each participant. HIV reservoirs were measured in blood and tissues and HIV-specific antibodies were measured in plasma. We used the Wilcoxon signed-rank test to compare data collected before and after allo-HSCT in participants for whom longitudinal data were available. When the paired test was not possible, we used the Mann-Whitney U test. We developed a mathematical model to study the factors influencing HIV reservoir reduction in people with HIV after allo-HSCT. FINDINGS We included 30 people with HIV with haematological malignancies who received a transplant between Sept 1, 2009 and April 30, 2019 and were enrolled within the IciStem cohort and included in this analysis. HIV reservoirs in peripheral blood were reduced immediately after full donor chimerism was achieved, generally accompanied by undetectable HIV-DNA in bone marrow, ileum, lymph nodes, and cerebrospinal fluid, regardless of donor CCR5 genotype. HIV-specific antibody levels and functionality values declined more slowly than direct HIV reservoir values, decaying significantly only months after full donor chimerism. Mathematical modelling suggests that allogeneic immunity mediated by donor cells is the main viral reservoir depletion mechanism after massive reservoir reduction during conditioning chemotherapy before allo-HSCT (half-life of latently infected replication-competent cells decreased from 44 months to 1·5 months). INTERPRETATION Our work provides, for the first time, data on the effects of allo-HSCT in the context of HIV infection. Additionally, we raise the question of which marker can serve as the last reporter of the residual viraemia, postulating that the absence of T-cell immune responses might be a more reliable marker than antibody decline after allo-HSCT. FUNDING amfAR (American Foundation for AIDS Research; ARCHE Program), National Institutes of Health, National Institute of Allergy and Infectious Diseases, and Dutch Aidsfonds.
Collapse
Affiliation(s)
- Maria Salgado
- IrsiCaixa, Badalona, Spain; Germans Trias i Pujol Research Institute, Badalona, Spain; Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain.
| | | | - Monique Nijhuis
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands; HIV Pathogenesis Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Mi Kwon
- Department of Hematology, Hospital Universitario Gregorio Marañón, Institute of Health Research Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - E Fabian Cardozo-Ojeda
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Xencor, Pasadena, CA, USA
| | - Jon Badiola
- University Hospital Virgen de las Nieves, Granada, Spain
| | - Matthew J Gorman
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA; Moderna Therapeutics, Cambridge, MA, USA
| | - Laura E P Huyveneers
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Alessandra Bandera
- Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Björn-Erik Ole Jensen
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Manuel Jurado
- University Hospital Virgen de las Nieves, Granada, Spain
| | | | - Julian Schulze Zur Wiesch
- Infectious Diseases Unit, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Rebeca Bailén
- Department of Hematology, Hospital Universitario Gregorio Marañón, Institute of Health Research Gregorio Marañón, Madrid, Spain
| | - Johanna M Eberhard
- Infectious Diseases Unit, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany; Helmholtz Institute for One Health, Greifswald, Germany
| | - Mitja Nabergoj
- Division of Hematology, Hôpitaux Universitaires de Genève, Geneva, Switzerland; Hematology Service, Institut Central des Hôpitaux, Sion, Switzerland
| | | | | | - Sharon Oldford
- Nova Scotia Health, Dalhousie University, Halifax, NS, Canada
| | - Lisa Barrett
- Nova Scotia Health, Dalhousie University, Halifax, NS, Canada
| | - Maria Luisa Montes Ramirez
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain; University Hospital La Paz, IdiPAZ, Madrid, Spain
| | - Salisu Garba
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Merck, Rahway, NJ, USA
| | | | - Boris Revollo
- Department of Infectious Diseases, University Hospital Germans Trias i Pujol, Institut Català d'Oncologia, Badalona, Spain
| | - Christelle Ferra-Coll
- Department of Hematology, University Hospital Germans Trias i Pujol, Institut Català d'Oncologia, Badalona, Spain; University of Vic-Central University of Catalonia, Vic, Spain
| | - Jurgen Kuball
- Department of Hematology and Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Galit Alter
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA; Moderna Therapeutics, Cambridge, MA, USA
| | - Asier Sáez-Cirión
- Viral Reservoirs and Immune Control Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Jose Luis Diez-Martin
- Department of Hematology, Hospital Universitario Gregorio Marañón, Institute of Health Research Gregorio Marañón, Madrid, Spain
| | - Elizabeth R Duke
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Medicine, Allergy and Infectious Diseases Division, University of Washington, WA, Seattle, USA
| | - Joshua T Schiffer
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Medicine, Allergy and Infectious Diseases Division, University of Washington, WA, Seattle, USA
| | - Annemarie Wensing
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands; Ezintsha, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Javier Martinez-Picado
- IrsiCaixa, Badalona, Spain; Germans Trias i Pujol Research Institute, Badalona, Spain; Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain; University of Vic-Central University of Catalonia, Vic, Spain; Catalan Institution for Research and Advanced Studies, Barcelona, Spain.
| |
Collapse
|
33
|
Schulze WJ, Gregory DA, Johnson MC, Lange MJ. Genome-wide CRISPR/Cas9 screen reveals JunB downmodulation of HIV co-receptor CXCR4. Front Microbiol 2024; 15:1342444. [PMID: 38835488 PMCID: PMC11149427 DOI: 10.3389/fmicb.2024.1342444] [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: 11/21/2023] [Accepted: 04/24/2024] [Indexed: 06/06/2024] Open
Abstract
HIV-1 relies extensively on host cell machinery for replication. Identification and characterization of these host-virus interactions is vital to our understanding of viral replication and the consequences of infection in cells. Several prior screens have identified host factors important for HIV replication but with limited replication of findings, likely due to differences in experimental design and conditions. Thus, unidentified factors likely exist. To identify novel host factors required for HIV-1 infection, we performed a genome-wide CRISPR/Cas9 screen using HIV-induced cell death as a partitioning method. We created a gene knockout library in TZM-GFP reporter cells using GeCKOv2, which targets 19,050 genes, and infected the library with a lethal dose of HIV-1NL4-3. We hypothesized that cells with a knockout of a gene critical for HIV infection would survive while cells with a knockout of a non-consequential gene would undergo HIV-induced death and be lost from the population. Surviving cells were analyzed by high throughput sequencing of the integrated CRISPR/Cas9 cassette to identify the gene knockout. Of the gene targets, an overwhelming majority of the surviving cells harbored the guide sequence for the AP-1 transcription factor family protein, JunB. Upon the generation of a clonal JunB knockout cell line, we found that HIV-1NL4-3 infection was blocked in the absence of JunB. The phenotype resulted from downregulation of CXCR4, as infection levels were recovered by reintroduction of CXCR4 in JunB KO cells. Thus, JunB downmodulates CXCR4 expression in TZM-GFP cells, reducing CXCR4-tropic HIV infection.
Collapse
Affiliation(s)
| | | | | | - Margaret J. Lange
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
34
|
Yin DE, Palin AC, Lombo TB, Mahon RN, Poon B, Wu DY, Atala A, Brooks KM, Chen S, Coyne CB, D’Souza MP, Fackler OT, Furler O’Brien RL, Garcia-de-Alba C, Jean-Philippe P, Karn J, Majji S, Muotri AR, Ozulumba T, Sakatis MZ, Schlesinger LS, Singh A, Spiegel HM, Struble E, Sung K, Tagle DA, Thacker VV, Tidball AM, Varthakavi V, Vunjak-Novakovic G, Wagar LE, Yeung CK, Ndhlovu LC, Ott M. 3D human tissue models and microphysiological systems for HIV and related comorbidities. Trends Biotechnol 2024; 42:526-543. [PMID: 38071144 PMCID: PMC11065605 DOI: 10.1016/j.tibtech.2023.10.008] [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: 07/03/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 03/03/2024]
Abstract
Three-dimensional (3D) human tissue models/microphysiological systems (e.g., organs-on-chips, organoids, and tissue explants) model HIV and related comorbidities and have potential to address critical questions, including characterization of viral reservoirs, insufficient innate and adaptive immune responses, biomarker discovery and evaluation, medical complexity with comorbidities (e.g., tuberculosis and SARS-CoV-2), and protection and transmission during pregnancy and birth. Composed of multiple primary or stem cell-derived cell types organized in a dedicated 3D space, these systems hold unique promise for better reproducing human physiology, advancing therapeutic development, and bridging the human-animal model translational gap. Here, we discuss the promises and achievements with 3D human tissue models in HIV and comorbidity research, along with remaining barriers with respect to cell biology, virology, immunology, and regulatory issues.
Collapse
|
35
|
Bone B, Lichterfeld M. "Block and lock" viral integration sites in persons with drug-free control of HIV-1 infection. Curr Opin HIV AIDS 2024; 19:110-115. [PMID: 38457193 DOI: 10.1097/coh.0000000000000845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
PURPOSE OF REVIEW Elite controllers (ECs) and Posttreatment controllers (PTCs) represent a small subset of individuals who are capable of maintaining drug-free control of HIV plasma viral loads despite the persistence of a replication-competent viral reservoir. This review aims to curate recent experimental studies evaluating viral reservoirs that distinguish EC/PTC and may contribute to their ability to maintain undetectable viral loads in the absence of antiretroviral therapy. RECENT FINDINGS Recent studies on ECs have demonstrated that integration sites of intact proviruses in EC/PTC are markedly biased towards heterochromatin regions; in contrast, intact proviruses in accessible and permissive chromatin were profoundly underrepresented. Of note, no such biases were noted when CD4 + T cells from EC were infected directly ex vivo, suggesting that the viral reservoir profile in EC is not related to altered integration site preferences during acute infection, but instead represents the result of immune-mediated selection mechanisms that can eliminate proviruses in transcriptionally-active euchromatin regions while promoting preferential persistence of intact proviruses in nonpermissive genome regions. Proviral transcription in such "blocked and locked" regions may be restricted through epigenetic mechanisms, protecting them from immune-recognition but presumably limiting their ability to drive viral rebound. While the exact immune mechanisms driving this selection process remain undefined, recent single-cell analytic approaches support the hypothesis that HIV reservoir cells are subject to immune selection pressure by host factors. SUMMARY A "blocked and locked" viral reservoir profile may constitute a structural virological correlate of a functional cure of HIV-1 infection. Further research into the immunological mechanism promoting HIV-1 reservoir selection and evolution in EC/PTC is warranted and could inform foreseeable cure strategies.
Collapse
Affiliation(s)
- Benjamin Bone
- Infectious Disease Division, Brigham Women's Hospital, Boston
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Mathias Lichterfeld
- Infectious Disease Division, Brigham Women's Hospital, Boston
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
36
|
Pellaers E, Debyser Z. EDITORIAL: Putting HIV in the big sleep with the block-and-lock strategy. Curr Opin HIV AIDS 2024; 19:93-94. [PMID: 38568102 DOI: 10.1097/coh.0000000000000853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Affiliation(s)
- Eline Pellaers
- Molecular Virology and Gene Therapy, KU Leuven, Leuven, Flanders, Belgium
| | | |
Collapse
|
37
|
Wu L, Feng Y, Huang Y, Feng J, Hu Y, Huang H. CAR-T Cell Therapy: Advances in Kidney-Related Diseases. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:143-152. [PMID: 38751795 PMCID: PMC11095583 DOI: 10.1159/000536194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/15/2023] [Indexed: 05/18/2024]
Abstract
Background Chimeric antigen receptor (CAR)-T cell therapy represents a significant advancement in the field of immunotherapy, providing targeted eradication of abnormal cells through the recognition between CAR and target antigens. This approach has garnered considerable attention due to its promising results in the clinical treatment of hematological malignancies and autoimmune diseases. As the focus shifts toward exploring novel targets and expanding the application of CAR-T cell therapy to solid tumors, including renal malignancies, researchers are pushing the boundaries of this innovative treatment. However, it is crucial to address the observed comorbidities associated with CAR-T cell therapy, particularly nephrotoxicity, due to the superseding release of cytokines and impairment of normal tissue. Summary Our review discusses the research strategies and nephrotoxicity related to CAR-T cell therapy in various kidney-related diseases and provides insights into enhancing investigation and optimization. Key Messages CAR-T cell therapy has captured the attention of researchers and clinicians in the treatment of renal malignancies, multiple myeloma, systemic lupus erythematosus, and acquired immunodeficiency syndrome, which may lead to potential nephrotoxicity as they involve primary or secondary kidney complications. Understanding and summarizing the current research progress of CAR-T cell therapies can provide valuable insights into novel targets and combinations to optimize research models and enhance their clinical value.
Collapse
Affiliation(s)
- Longyuan Wu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Youqin Feng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yue Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Jingjing Feng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| |
Collapse
|
38
|
Luo F, Wang Q, Chen S. Editorial: Prevention of viral diseases by gene targeting. Front Genome Ed 2024; 6:1395468. [PMID: 38572375 PMCID: PMC10987956 DOI: 10.3389/fgeed.2024.1395468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Affiliation(s)
- Fan Luo
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qiankun Wang
- Shenzhen Bay Laboratory, Institute of Infectious Diseases, Shenzhen, China
| | - Shuliang Chen
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
39
|
OBrien SJ. Legacy of a magic gene- CCR5-∆32: From discovery to clinical benefit in a generation. Proc Natl Acad Sci U S A 2024; 121:e2321907121. [PMID: 38457490 PMCID: PMC10962972 DOI: 10.1073/pnas.2321907121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/22/2024] [Indexed: 03/10/2024] Open
Abstract
The discovery of the 32-bp deletion allele of the chemokine receptor gene CCR5 showed that homozygous carriers display near-complete resistance to HIV infection, irrespective of exposure. Algorithms of molecular evolutionary theory suggested that the CCR5-∆32 mutation occurred but once in the last millennium and rose by strong selective pressure relatively recently to a ~10% allele frequency in Europeans. Several lines of evidence support the hypothesis that CCR5-∆32 was selected due to its protective influence to resist Yersinia pestis, the agent of the Black Death/bubonic plague of the 14th century. Powerful anti-AIDS entry inhibitors targeting CCR5 were developed as a treatment for HIV patients, particularly those whose systems had developed resistance to powerful anti-retroviral therapies. Homozygous CCR5-∆32/∆32 stem cell transplant donors were used to produce HIV-cleared AIDS patients in at least five "cures" of HIV infection. CCR5 has also been implicated in regulating infection with Staphylococcus aureus, in recovery from stroke, and in ablation of the fatal graft versus host disease (GVHD) in cancer transplant patients. While homozygous CCR5-∆32/32 carriers block HIV infection, alternatively they display an increased risk for encephalomyelitis and death when infected with the West Nile virus.
Collapse
Affiliation(s)
- Stephen J. OBrien
- Guy Harvey Oceanographic Center, Halmos College of Arts and Sciences, Nova Southeastern University, Ft Lauderdale, FL33004
- Indiana University School of Public Health, Bloomington, IN47405
| |
Collapse
|
40
|
Roux H, Chomont N. Measuring Human Immunodeficiency Virus Reservoirs: Do We Need to Choose Between Quantity and Quality? J Infect Dis 2024; 229:635-643. [PMID: 37665978 PMCID: PMC10938203 DOI: 10.1093/infdis/jiad381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/17/2023] [Accepted: 08/31/2023] [Indexed: 09/06/2023] Open
Abstract
The persistence of latent viral genomes in people receiving antiretroviral therapy (ART) is the main obstacle to a cure for human immunodeficiency virus (HIV) infection. Viral reservoirs can be defined as cells harboring HIV genomes that have the ability to produce infectious virions. Precise quantification of the cellular reservoirs of HIV is challenging because these cells are rare, heterogeneous, and outnumbered by a larger number of cells carrying defective genomes. In addition, measuring the inducibility of these proviruses requires functional assays and remains technically difficult. The recent development of single-cell and single-viral genome approaches revealed additional layers of complexity: the cell subsets that harbor proviruses are heterogeneous and their ability to be induced is variable. A substantial fraction of intact HIV genomes may be permanently silenced after years of ART, revealing the underappreciated importance of induction assays. As such, a simple approach that would assess simultaneously the genetic intactness and the inducibility of the reservoir is still lacking. In this study, we review recent advances in the development of methods to quantify and characterize persistently infected cells, and we discuss how these findings can inform the design of future assays aimed at measuring the size of the intact and inducible HIV reservoir.
Collapse
Affiliation(s)
- Hélène Roux
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada
- Centre de Recherche du CHUM, Montreal, Quebec, Canada
| | - Nicolas Chomont
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada
- Centre de Recherche du CHUM, Montreal, Quebec, Canada
| |
Collapse
|
41
|
Harper J, Betts MR, Lichterfeld M, Müller-Trutwin M, Margolis D, Bar KJ, Li JZ, McCune JM, Lewin SR, Kulpa D, Ávila-Ríos S, Diallo DD, Lederman MM, Paiardini M. Erratum to: Progress Note 2024: Curing HIV; Not in My Lifetime or Just Around the Corner? Pathog Immun 2024; 8:179-222. [PMID: 38505662 PMCID: PMC10949969 DOI: 10.20411/pai.v8i2.696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024] Open
Abstract
[This corrects the article DOI: 10.20411/pai.v8i2.665.].
Collapse
Affiliation(s)
- Justin Harper
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Michael R. Betts
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for AIDS Research, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts
| | - Michaela Müller-Trutwin
- HIV Inflammation and Persistence Unit, Institut Pasteur, Université Paris-Cité, Paris, France
| | - David Margolis
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina
| | - Katharine J. Bar
- Center for AIDS Research, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jonathan Z. Li
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Joseph M. McCune
- HIV Frontiers, Global Health Accelerator, Bill & Melinda Gates Foundation
| | - Sharon R. Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Deanna Kulpa
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Santiago Ávila-Ríos
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | | | - Michael M. Lederman
- Division of Infectious Diseases and HIV Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
42
|
Beschorner N, Künzle P, Voges M, Hauber I, Indenbirken D, Nakel J, Virdi S, Bradtke P, Lory NC, Rothe M, Paszkowski-Rogacz M, Buchholz F, Grundhoff A, Schambach A, Thirion C, Mittrücker HW, Schulze zur Wiesch J, Hauber J, Chemnitz J. Preclinical toxicity analyses of lentiviral vectors expressing the HIV-1 LTR-specific designer-recombinase Brec1. PLoS One 2024; 19:e0298542. [PMID: 38457474 PMCID: PMC10923487 DOI: 10.1371/journal.pone.0298542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/25/2024] [Indexed: 03/10/2024] Open
Abstract
Drug-based antiretroviral therapies (ART) efficiently suppress HIV replication in humans, but the virus persists as integrated proviral reservoirs in small numbers of cells. Importantly, ART cannot eliminate HIV from an infected individual, since it does not target the integrated provirus. Therefore, genome editing-based strategies that can inactivate or excise HIV genomes would provide the technology for novel curative therapies. In fact, the HIV-1 LTR-specific designer-recombinase Brec1 has been shown to remove integrated proviruses from infected cells and is highly efficacious on clinical HIV-1 isolates in vitro and in vivo, suggesting that Brec1 has the potential for clinical development of advanced HIV-1 eradication strategies in people living with HIV. In line with the preparation of a first-in-human advanced therapy medicinal product gene therapy trial, we here present an extensive preclinical evaluation of Brec1 and lentiviral vectors expressing the Brec1 transgene. This included detailed functional analysis of potential genomic off-target sites, assessing vector safety by investigating vector copy number (VCN) and the risk for potential vector-related insertional mutagenesis, as well as analyzing the potential of Brec1 to trigger an undesired strong T cell immune response. In conclusion, the antiviral designer-recombinase Brec1 is shown to lack any detectable cytopathic, genotoxic or T cell-related immunogenic effects, thereby meeting an important precondition for clinical application of the therapeutic lentiviral vector LV-Brec1 in novel HIV-1 curative strategies.
Collapse
Affiliation(s)
- Niklas Beschorner
- Leibniz-Institute of Virology (LIV), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg–Lübeck–Borstel–Riems, Germany
- PROVIREX Genome Editing Therapies GmbH, Hamburg, Germany
| | - Paul Künzle
- Leibniz-Institute of Virology (LIV), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg–Lübeck–Borstel–Riems, Germany
| | - Maike Voges
- Leibniz-Institute of Virology (LIV), Hamburg, Germany
- PROVIREX Genome Editing Therapies GmbH, Hamburg, Germany
| | - Ilona Hauber
- Leibniz-Institute of Virology (LIV), Hamburg, Germany
- PROVIREX Genome Editing Therapies GmbH, Hamburg, Germany
| | - Daniela Indenbirken
- Leibniz-Institute of Virology (LIV), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg–Lübeck–Borstel–Riems, Germany
| | - Jacqueline Nakel
- Leibniz-Institute of Virology (LIV), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg–Lübeck–Borstel–Riems, Germany
| | - Sanamjeet Virdi
- Leibniz-Institute of Virology (LIV), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg–Lübeck–Borstel–Riems, Germany
| | - Peter Bradtke
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Niels Christian Lory
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | - Frank Buchholz
- PROVIREX Genome Editing Therapies GmbH, Hamburg, Germany
- Medical Systems Biology, UCC, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Adam Grundhoff
- Leibniz-Institute of Virology (LIV), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg–Lübeck–Borstel–Riems, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | - Hans-Willi Mittrücker
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Schulze zur Wiesch
- German Center for Infection Research (DZIF), Partner Site Hamburg–Lübeck–Borstel–Riems, Germany
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joachim Hauber
- Leibniz-Institute of Virology (LIV), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg–Lübeck–Borstel–Riems, Germany
- PROVIREX Genome Editing Therapies GmbH, Hamburg, Germany
| | - Jan Chemnitz
- Leibniz-Institute of Virology (LIV), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg–Lübeck–Borstel–Riems, Germany
- PROVIREX Genome Editing Therapies GmbH, Hamburg, Germany
| |
Collapse
|
43
|
Payra S, Manjhi PK, Singh S, Kumar R, Singh SK, Kumar A, Maharshi V. HIV cure: Are we going to make history? HIV Med 2024; 25:322-331. [PMID: 37821095 DOI: 10.1111/hiv.13557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND At present, combination antiretroviral therapy (cART) is the mainstay for the treatment of people living with HIV/AIDS. cART can suppress the viral load to a minimal level; however, the possibility of the emergence of full-blown AIDS is always there. In the latter part of the first decade of the 21st century, an HIV-positive person received stem cell transplantation (SCT) for treatment of his haematological malignancy. The patient was able to achieve remission of the haematological condition as well as of HIV following SCT. Thorough investigations of various samples including blood and biopsy could not detect the virus in the person's body. The person was declared to be the first cured case of HIV. LITERATURE SEARCH Over the next decade, a few more similar cases were observed and have recently been declared cured of the infection. A comprehensive search was performed in PubMed, Cochrane library and Google Scholar. Four such additional cases were found in literature. DESCRIPTION & DISCUSSION These cases all share a common proposed mechanism for the HIV cure, that is, transplantation of stem cells from donors carrying a homozygous mutation in a gene encoding for CCR5 (receptor utilized by HIV for entry into the host cell), denoted as CCR5△32. This mutation makes the host immune cells devoid of CCR5, causing the host to acquire resistance against HIV. To the best of our knowledge, this is the first review to look at relevant and updated information of all cured cases of HIV as well as the related landmarks in history and discusses the underlying mechanism(s).
Collapse
Affiliation(s)
- Shuvasree Payra
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Pramod Kumar Manjhi
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Shruti Singh
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Rajesh Kumar
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Sunil Kumar Singh
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Alok Kumar
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Vikas Maharshi
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| |
Collapse
|
44
|
Harper J, Betts MR, Lichterfeld M, Müller-Trutwin M, Margolis D, Bar KJ, Li JZ, McCune JM, Lewin SR, Kulpa D, Ávila-Ríos S, Diallo DD, Lederman MM, Paiardini M. Progress Note 2024: Curing HIV; Not in My Lifetime or Just Around the Corner? Pathog Immun 2024; 8:115-157. [PMID: 38455668 PMCID: PMC10919397 DOI: 10.20411/pai.v8i2.665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/14/2024] [Indexed: 03/09/2024] Open
Abstract
Once a death sentence, HIV is now considered a manageable chronic disease due to the development of antiretroviral therapy (ART) regimens with minimal toxicity and a high barrier for genetic resistance. While highly effective in arresting AIDS progression and rendering the virus untransmissible in people living with HIV (PLWH) with undetectable viremia (U=U) [1, 2]), ART alone is incapable of eradicating the "reservoir" of resting, latently infected CD4+ T cells from which virus recrudesces upon treatment cessation. As of 2022 estimates, there are 39 million PLWH, of whom 86% are aware of their status and 76% are receiving ART [3]. As of 2017, ART-treated PLWH exhibit near normalized life expectancies without adjustment for socioeconomic differences [4]. Furthermore, there is a global deceleration in the rate of new infections [3] driven by expanded access to pre-exposure prophylaxis (PrEP), HIV testing in vulnerable populations, and by ART treatment [5]. Therefore, despite outstanding issues pertaining to cost and access in developing countries, there is strong enthusiasm that aggressive testing, treatment, and effective viral suppression may be able to halt the ongoing HIV epidemic (ie, UNAIDS' 95-95-95 targets) [6-8]; especially as evidenced by recent encouraging observations in Sydney [9]. Despite these promising efforts to limit further viral transmission, for PLWH, a "cure" remains elusive; whether it be to completely eradicate the viral reservoir (ie, cure) or to induce long-term viral remission in the absence of ART (ie, control; Figure 1). In a previous salon hosted by Pathogens and Immunity in 2016 [10], some researchers were optimistic that a cure was a feasible, scalable goal, albeit with no clear consensus on the best route. So, how are these cure strategies panning out? In this commentary, 8 years later, we will provide a brief overview on recent advances and failures towards identifying determinants of viral persistence and developing a scalable cure for HIV. Based on these observations, and as in the earlier salon, we have asked several prominent HIV cure researchers for their perspectives.
Collapse
Affiliation(s)
- Justin Harper
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Michael R. Betts
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for AIDS Research, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts
| | - Michaela Müller-Trutwin
- HIV Inflammation and Persistence Unit, Institut Pasteur, Université Paris-Cité, Paris, France
| | - David Margolis
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina
| | - Katharine J. Bar
- Center for AIDS Research, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jonathan Z. Li
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Joseph M. McCune
- HIV Frontiers, Global Health Accelerator, Bill & Melinda Gates Foundation
| | - Sharon R. Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Deanna Kulpa
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Santiago Ávila-Ríos
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | | | - Michael M. Lederman
- Division of Infectious Diseases and HIV Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
45
|
Li K, Zhang Q. Eliminating the HIV tissue reservoir: current strategies and challenges. Infect Dis (Lond) 2024; 56:165-182. [PMID: 38149977 DOI: 10.1080/23744235.2023.2298450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/16/2023] [Indexed: 12/28/2023] Open
Abstract
BACKGROUND Acquired immunodeficiency syndrome (AIDS) is still one of the most widespread and harmful infectious diseases in the world. The presence of reservoirs housing the human immunodeficiency virus (HIV) represents a significant impediment to the development of clinically applicable treatments on a large scale. The viral load in the blood can be effectively reduced to undetectable levels through antiretroviral therapy (ART), and a higher concentration of HIV is sequestered in various tissues throughout the body, forming the tissue reservoir - the source of viremia after interruption treatment. METHODS We take the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) as a guideline for this review. In June 2023, we used the Pubmed, Embase, and Scopus databases to search the relevant literature published in the last decade. RESULTS Here we review the current strategies and treatments for eliminating the HIV tissue reservoirs: early and intensive therapy, gene therapy (including ribozyme, RNA interference, RNA aptamer, zinc finger enzyme, transcriptional activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats/associated nuclease 9 (CRISPR/Cas9)), 'Shock and Kill', 'Block and lock', immunotherapy (including therapeutic vaccines, broadly neutralising antibodies (bNAbs), chimeric antigen receptor T-cell immunotherapy (CAR-T)), and haematopoietic stem cell transplantation (HSCT). CONCLUSION The existence of an HIV reservoir is the main obstacle to the complete cure of AIDS. Choosing the appropriate strategy to deplete the HIV reservoir and achieve a functional cure for AIDS is the focus and difficulty of current research. So far, there has been a lot of research and progress in reducing the HIV reservoir, but in general, the current research is still very preliminary. Much research is still needed to properly assess the reliability, effectiveness, and necessity of these strategies.
Collapse
Affiliation(s)
- Kangpeng Li
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Qiang Zhang
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
46
|
Armani-Tourret M, Gao C, Hartana CA, Sun W, Carrere L, Vela L, Hochroth A, Bellefroid M, Sbrolla A, Shea K, Flynn T, Roseto I, Rassadkina Y, Lee C, Giguel F, Malhotra R, Bushman FD, Gandhi RT, Yu XG, Kuritzkes DR, Lichterfeld M. Selection of epigenetically privileged HIV-1 proviruses during treatment with panobinostat and interferon-α2a. Cell 2024; 187:1238-1254.e14. [PMID: 38367616 PMCID: PMC10903630 DOI: 10.1016/j.cell.2024.01.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/26/2023] [Accepted: 01/24/2024] [Indexed: 02/19/2024]
Abstract
CD4+ T cells with latent HIV-1 infection persist despite treatment with antiretroviral agents and represent the main barrier to a cure of HIV-1 infection. Pharmacological disruption of viral latency may expose HIV-1-infected cells to host immune activity, but the clinical efficacy of latency-reversing agents for reducing HIV-1 persistence remains to be proven. Here, we show in a randomized-controlled human clinical trial that the histone deacetylase inhibitor panobinostat, when administered in combination with pegylated interferon-α2a, induces a structural transformation of the HIV-1 reservoir cell pool, characterized by a disproportionate overrepresentation of HIV-1 proviruses integrated in ZNF genes and in chromatin regions with reduced H3K27ac marks, the molecular target sites for panobinostat. By contrast, proviruses near H3K27ac marks were actively selected against, likely due to increased susceptibility to panobinostat. These data suggest that latency-reversing treatment can increase the immunological vulnerability of HIV-1 reservoir cells and accelerate the selection of epigenetically privileged HIV-1 proviruses.
Collapse
Affiliation(s)
| | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Ciputra Adijaya Hartana
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - WeiWei Sun
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Leah Carrere
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Liliana Vela
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | | | - Amy Sbrolla
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Katrina Shea
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Theresa Flynn
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Isabelle Roseto
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Carole Lee
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Francoise Giguel
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rajeev Malhotra
- Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Frederic D Bushman
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajesh T Gandhi
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xu G Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel R Kuritzkes
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
47
|
Tassaneetrithep B, Phuphuakrat A, Pasomsub E, Bhukhai K, Wongkummool W, Priengprom T, Khamaikawin W, Chaisavaneeyakorn S, Anurathapan U, Apiwattanakul N, Hongeng S. HIV-1 proviral DNA in purified peripheral blood CD34 + stem and progenitor cells in individuals with long-term HAART; paving the way to HIV gene therapy. Heliyon 2024; 10:e26613. [PMID: 38434025 PMCID: PMC10906414 DOI: 10.1016/j.heliyon.2024.e26613] [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: 08/04/2023] [Revised: 02/03/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Human immunodeficiency virus (HIV)-1 infection is an important public health problem worldwide. After primary HIV-1 infection, transcribed HIV-1 DNA is integrated into the host genome, serving as a reservoir of the virus and hindering a definite cure. Although highly active antiretroviral therapy suppresses active viral replication, resulting in undetectable levels of HIV RNA in the blood, a viral rebound can be detected after a few weeks of treatment interruption. This supports the concept that there is a stable HIV-1 reservoir in people living with HIV-1. Recently, a few individuals with HIV infection were reported to be probably cured by hematopoietic stem transplantation (HSCT). The underlying mechanism for this success involved transfusion of uninfected hematopoietic stem and progenitor cells (HSPCs) from CCR5-mutated donors who were naturally resistant to HIV infection. Thus, gene editing technology to provide HIV-resistant HSPC has promise in the treatment of HIV infections by HSCT. In this study, we aimed to find HIV-infected individuals likely to achieve a definite cure via gene editing HSCT. We screened for total HIV proviral DNA by Alu PCR in peripheral blood mononuclear cells (PBMCs) of 20 HIV-infected individuals with prolonged viral suppression. We assessed the amount of intact proviral DNA via a modified intact proviral DNA assay (IPDA) in purified peripheral CD34+ HSPCs. PBMCs from all 20 individuals were positive for the gag gene in Alu PCR, and peripheral CD34+ HSPCs were IPDA-negative for six individuals. Our results suggested that these six HIV-infected individuals could be candidates for further studies into the ability of gene editing HSCT to lead to a definite HIV cure.
Collapse
Affiliation(s)
- Boonrat Tassaneetrithep
- Center of Research Excellence in Immunoregulation, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
| | - Angsana Phuphuakrat
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Ekawat Pasomsub
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Kanit Bhukhai
- Department of Physiology, Faculty of Science, Mahidol University, Thailand
| | | | - Thongkoon Priengprom
- Center of Research Excellence in Immunoregulation, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
| | - Wannisa Khamaikawin
- Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, Thailand
| | | | - Usanarat Anurathapan
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Nopporn Apiwattanakul
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| |
Collapse
|
48
|
Kitawi R, Ledger S, Kelleher AD, Ahlenstiel CL. Advances in HIV Gene Therapy. Int J Mol Sci 2024; 25:2771. [PMID: 38474018 DOI: 10.3390/ijms25052771] [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: 11/02/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Early gene therapy studies held great promise for the cure of heritable diseases, but the occurrence of various genotoxic events led to a pause in clinical trials and a more guarded approach to progress. Recent advances in genetic engineering technologies have reignited interest, leading to the approval of the first gene therapy product targeting genetic mutations in 2017. Gene therapy (GT) can be delivered either in vivo or ex vivo. An ex vivo approach to gene therapy is advantageous, as it allows for the characterization of the gene-modified cells and the selection of desired properties before patient administration. Autologous cells can also be used during this process which eliminates the possibility of immune rejection. This review highlights the various stages of ex vivo gene therapy, current research developments that have increased the efficiency and safety of this process, and a comprehensive summary of Human Immunodeficiency Virus (HIV) gene therapy studies, the majority of which have employed the ex vivo approach.
Collapse
Affiliation(s)
- Rose Kitawi
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Scott Ledger
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Anthony D Kelleher
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
- St. Vincent's Hospital, Darlinghurst, NSW 2010, Australia
- UNSW RNA Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Chantelle L Ahlenstiel
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
- UNSW RNA Institute, University of New South Wales, Kensington, NSW 2052, Australia
| |
Collapse
|
49
|
Dickter JK, Aribi A, Cardoso AA, Gianella S, Gendzekhadze K, Li S, Feng Y, Chaillon A, Laird GM, Browning DL, Ross JA, Nanayakkara DD, Puing A, Stan R, Lai LL, Chang S, Kidambi TD, Thomas S, Al Malki MM, Nakamura R, Alvarnas J, Taplitz RA, Dadwal SS, Forman SJ, Zaia JA. HIV-1 Remission after Allogeneic Hematopoietic-Cell Transplantation. N Engl J Med 2024; 390:669-671. [PMID: 38354149 PMCID: PMC10906479 DOI: 10.1056/nejmc2312556] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Affiliation(s)
| | - Ahmed Aribi
- City of Hope National Medical Center, Duarte, CA
| | | | | | | | - Shirley Li
- City of Hope National Medical Center, Duarte, CA
| | - Ye Feng
- City of Hope National Medical Center, Duarte, CA
| | | | | | | | | | | | | | - Rodica Stan
- City of Hope National Medical Center, Duarte, CA
| | - Lily L Lai
- City of Hope National Medical Center, Duarte, CA
| | - Sue Chang
- City of Hope National Medical Center, Duarte, CA
| | | | | | | | - Ryo Nakamura
- City of Hope National Medical Center, Duarte, CA
| | | | | | | | | | - John A Zaia
- City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
50
|
Guo Q, Zhang J, Parikh K, Brinkley A, Lin S, Zakarian C, Pernet O, Shimizu S, Khamaikawin W, Hacke K, Kasahara N, An DS. In vivo selection of anti-HIV-1 gene-modified human hematopoietic stem/progenitor cells to enhance engraftment and HIV-1 inhibition. Mol Ther 2024; 32:384-394. [PMID: 38087779 PMCID: PMC10862071 DOI: 10.1016/j.ymthe.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/17/2023] [Accepted: 12/08/2023] [Indexed: 12/26/2023] Open
Abstract
Hematopoietic stem/progenitor cell (HSPC)-based anti-HIV-1 gene therapy holds great promise to eradicate HIV-1 or to provide long-term remission through a continuous supply of anti-HIV-1 gene-modified cells without ongoing antiretroviral therapy. However, achieving sufficient engraftment levels of anti-HIV gene-modified HSPC to provide therapeutic efficacy has been a major limitation. Here, we report an in vivo selection strategy for anti-HIV-1 gene-modified HSPC by introducing 6-thioguanine (6TG) chemoresistance through knocking down hypoxanthine-guanine phosphoribosyl transferase (HPRT) expression using RNA interference (RNAi). We developed a lentiviral vector capable of co-expressing short hairpin RNA (shRNA) against HPRT alongside two anti-HIV-1 genes: shRNA targeting HIV-1 co-receptor CCR5 and a membrane-anchored HIV-1 fusion inhibitor, C46, for efficient in vivo selection of anti-HIV-1 gene-modified human HSPC. 6TG-mediated preconditioning and in vivo selection significantly enhanced engraftment of HPRT-knockdown anti-HIV-1 gene-modified cells (>2-fold, p < 0.0001) in humanized bone marrow/liver/thymus (huBLT) mice. Viral load was significantly reduced (>1 log fold, p < 0.001) in 6TG-treated HIV-1-infected huBLT mice compared to 6TG-untreated mice. We demonstrated that 6TG-mediated preconditioning and in vivo selection considerably improved engraftment of HPRT-knockdown anti-HIV-1 gene-modified HSPC and repopulation of anti-HIV-1 gene-modified hematopoietic cells in huBLT mice, allowing for efficient HIV-1 inhibition.
Collapse
Affiliation(s)
- Qi Guo
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Jian Zhang
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Keval Parikh
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Alexander Brinkley
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Samantha Lin
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Christina Zakarian
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Olivier Pernet
- Maternal, Child, and Adolescent Center for Infectious Diseases, University of Southern California, Los Angeles, CA 90089, USA
| | - Saki Shimizu
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Wannisa Khamaikawin
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA; Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, Bangkok 10520, Thailand
| | - Katrin Hacke
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Phoenix, AZ 85054, USA
| | - Noriyuki Kasahara
- UCSF, Neurological Surgery, Radiation Oncology, San Francisco, CA 94158, USA
| | - Dong Sung An
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|