51
|
Mangare C, Tischer-Zimmermann S, Bonifacius A, Riese SB, Dragon AC, Blasczyk R, Maecker-Kolhoff B, Eiz-Vesper B. Variances in Antiviral Memory T-Cell Repertoire of CD45RA- and CD62L-Depleted Lymphocyte Products Reflect the Need of Individual T-Cell Selection Strategies to Reduce the Risk of GvHD while Preserving Antiviral Immunity in Adoptive T-Cell Therapy. Transfus Med Hemother 2022; 49:30-43. [PMID: 35221866 PMCID: PMC8832244 DOI: 10.1159/000516284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/01/2021] [Indexed: 04/03/2025] Open
Abstract
INTRODUCTION Viral infections and reactivations still remain a cause of morbidity and mortality after hematopoietic stem cell transplantation due to immunodeficiency and immunosuppression. Transfer of unmanipulated donor-derived lymphocytes (DLI) represents a promising strategy for improving cellular immunity but carries the risk of graft versus host disease (GvHD). Depleting alloreactive naïve T cells (TN) from DLIs was implemented to reduce the risk of GvHD induction while preserving antiviral memory T-cell activity. Here, we compared two TN depletion strategies via CD45RA and CD62L expression and investigated the presence of antiviral memory T cells against human adenovirus (AdV) and Epstein-Barr virus (EBV) in the depleted fractions in relation to their functional and immunophenotypic characteristics. METHODS T-cell responses against ppEBV_EBNA1, ppEBV_Consensus and ppAdV_Hexon within TN-depleted (CD45RA-/CD62L-) and TN-enriched (CD45RA+/CD62L+) fractions were quantified by interferon-gamma (IFN-γ) ELISpot assay after short- and long-term in vitro stimulation. T-cell frequencies and immunophenotypic composition were assessed in all fractions by flow cytometry. Moreover, alloimmune T-cell responses were evaluated by mixed lymphocyte reaction. RESULTS According to differences in the phenotype composition, antigen-specific T-cell responses in CD45RA- fraction were up to 2 times higher than those in the CD62L- fraction, with the highest increase (up to 4-fold) observed after 7 days for ppEBV_EBNA1-specific T cells. The CD4+ effector memory T cells (TEM) were mainly responsible for EBV_EBNA1- and AdV_Hexon-specific T-cell responses, whereas the main functionally active T cells against ppEBV_Consensus were CD8+ central memory T cells (TCM) and TEM. Moreover, comparison of both depletion strategies indicated that alloreactivity in CD45RA- was lower than that in CD62L- fraction. CONCLUSION Taken together, our results indicate that CD45RA depletion is a more suitable strategy for generating TN-depleted products consisting of memory T cells against ppEBV_EBNA1 and ppAdV_Hexon than CD62L in terms of depletion effectiveness, T-cell functionality and alloreactivity. To maximally exploit the beneficial effects mediated by antiviral memory T cells in TN-depleted products, depletion methods should be selected individually according to phenotype composition and CD4/CD8 antigen restriction. TN-depleted DLIs may improve the clinical outcome in terms of infections, GvHD, and disease relapse if selection of pathogen-specific donor T cells is not available.
Collapse
Affiliation(s)
- Caroline Mangare
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sebastian B. Riese
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Anna Christina Dragon
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| |
Collapse
|
52
|
Füchsl F, Krackhardt AM. Adoptive Cellular Therapy for Multiple Myeloma Using CAR- and TCR-Transgenic T Cells: Response and Resistance. Cells 2022; 11:410. [PMID: 35159220 PMCID: PMC8834324 DOI: 10.3390/cells11030410] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 12/15/2022] Open
Abstract
Despite the substantial improvement of therapeutic approaches, multiple myeloma (MM) remains mostly incurable. However, immunotherapeutic and especially T cell-based approaches pioneered the therapeutic landscape for relapsed and refractory disease recently. Targeting B-cell maturation antigen (BCMA) on myeloma cells has been demonstrated to be highly effective not only by antibody-derived constructs but also by adoptive cellular therapies. Chimeric antigen receptor (CAR)-transgenic T cells lead to deep, albeit mostly not durable responses with manageable side-effects in intensively pretreated patients. The spectrum of adoptive T cell-transfer covers synthetic CARs with diverse specificities as well as currently less well-established T cell receptor (TCR)-based personalized strategies. In this review, we want to focus on treatment characteristics including efficacy and safety of CAR- and TCR-transgenic T cells in MM as well as the future potential these novel therapies may have. ACT with transgenic T cells has only entered clinical trials and various engineering strategies for optimization of T cell responses are necessary to overcome therapy resistance mechanisms. We want to outline the current success in engineering CAR- and TCR-T cells, but also discuss challenges including resistance mechanisms of MM for evading T cell therapy and point out possible novel strategies.
Collapse
Affiliation(s)
- Franziska Füchsl
- School of Medicine, Klinik und Poliklinik für Innere Medizin III, Klinikum rechts der Isar, Technische Universität München, Ismaningerstraße 22, 81675 Munich, Germany;
| | - Angela M. Krackhardt
- School of Medicine, Klinik und Poliklinik für Innere Medizin III, Klinikum rechts der Isar, Technische Universität München, Ismaningerstraße 22, 81675 Munich, Germany;
- German Cancer Consortium (DKTK), Partner-Site Munich, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Einsteinstraße 25, 81675 Munich, Germany
| |
Collapse
|
53
|
Adaptive Immune Responses, Immune Escape and Immune-Mediated Pathogenesis during HDV Infection. Viruses 2022; 14:v14020198. [PMID: 35215790 PMCID: PMC8880046 DOI: 10.3390/v14020198] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 12/13/2022] Open
Abstract
The hepatitis delta virus (HDV) is the smallest known human virus, yet it causes great harm to patients co-infected with hepatitis B virus (HBV). As a satellite virus of HBV, HDV requires the surface antigen of HBV (HBsAg) for sufficient viral packaging and spread. The special circumstance of co-infection, albeit only one partner depends on the other, raises many virological, immunological, and pathophysiological questions. In the last years, breakthroughs were made in understanding the adaptive immune response, in particular, virus-specific CD4+ and CD8+ T cells, in self-limited versus persistent HBV/HDV co-infection. Indeed, the mechanisms of CD8+ T cell failure in persistent HBV/HDV co-infection include viral escape and T cell exhaustion, and mimic those in other persistent human viral infections, such as hepatitis C virus (HCV), human immunodeficiency virus (HIV), and HBV mono-infection. However, compared to these larger viruses, the small HDV has perfectly adapted to evade recognition by CD8+ T cells restricted by common human leukocyte antigen (HLA) class I alleles. Furthermore, accelerated progression towards liver cirrhosis in persistent HBV/HDV co-infection was attributed to an increased immune-mediated pathology, either caused by innate pathways initiated by the interferon (IFN) system or triggered by misguided and dysfunctional T cells. These new insights into HDV-specific adaptive immunity will be discussed in this review and put into context with known well-described aspects in HBV, HCV, and HIV infections.
Collapse
|
54
|
Koning D, Quakkelaar ED, Schellens IMM, Spierings E, van Baarle D. Protective HLA Alleles Recruit Biased and Largely Similar Antigen-Specific T Cell Repertoires across Different Outcomes in HIV Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:3-15. [PMID: 34880106 DOI: 10.4049/jimmunol.2001145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/01/2021] [Indexed: 11/19/2022]
Abstract
CD8+ T cells play an important role in the control of untreated HIV infection. Several studies have suggested a decisive role of TCRs involved in anti-HIV immunity. HLA-B*27 and B*57 are often associated with a delayed HIV disease progression, but the exact correlates that provide superior immunity against HIV are not known. To investigate if the T cell repertoire underlies the protective effect in disease outcome in HLA-B*27 and B*57+ individuals, we analyzed Ag-specific TCR profiles from progressors (n = 13) and slow progressors (n = 11) expressing either B*27 or B*57. Our data showed no differences in TCR diversity between progressors and slow progressors. Both alleles recruit biased T cell repertoires (i.e., TCR populations skewed toward specific TRBV families or CDR3 regions). This bias was unrelated to disease progression and was remarkably profound for HLA-B*57, in which TRBV family usage and CDR3 sequences were shared to some extent even between epitopes. Conclusively, these data suggest that the T cell repertoires recruited by protective HLA alleles are highly similar between progressors and slow progressors in terms of TCR diversity, TCR usage, and cross-reactivity.
Collapse
Affiliation(s)
- Dan Koning
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands; and
| | - Esther D Quakkelaar
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands; and
| | - Ingrid M M Schellens
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands; and
| | - Eric Spierings
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands; and
| | - Debbie van Baarle
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands; and .,Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| |
Collapse
|
55
|
Vieira VA, Millar J, Adland E, Muenchhoff M, Roider J, Guash CF, Peluso D, Thomé B, Garcia-Guerrero MC, Puertas MC, Bamford A, Brander C, Carrington M, Martinez-Picado J, Frater J, Tudor-Williams G, Goulder P. Robust HIV-specific CD4+ and CD8+ T-cell responses distinguish elite control in adolescents living with HIV from viremic nonprogressors. AIDS 2022; 36:95-105. [PMID: 34581306 PMCID: PMC8654249 DOI: 10.1097/qad.0000000000003078] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Elite controllers are therapy-naive individuals living with HIV capable of spontaneous control of plasma viraemia for at least a year. Although viremic nonprogressors are more common in vertical HIV-infection than in adults' infection, elite control has been rarely characterized in the pediatric population. DESIGN We analyzed the T-cell immunophenotype and the HIV-specific response by flow cytometry in four pediatric elite controllers (PECs) compared with age-matched nonprogressors (PNPs), progressors and HIV-exposed uninfected (HEUs) adolescents. RESULTS PECs T-cell populations had lower immune activation and exhaustion levels when compared with progressors, reflected by a more sustained and preserved effector function. The HIV-specific T-cell responses among PECs were characterized by high-frequency Gag-specific CD4+ T-cell activity, and markedly more polyfunctional Gag-specific CD8+ activity, compared with PNPs and progressors. These findings were consistently observed even in the absence of protective HLA-I molecules such as HLA-B∗27/57/81. CONCLUSION Pediatric elite control is normally achieved after years of infection, and low immune activation in PNPs precedes the increasing ability of CD8+ T-cell responses to achieve immune control of viraemia over the course of childhood, whereas in adults, high immune activation in acute infection predicts subsequent CD8+ T-cell mediated immune control of viremia, and in adult elite controllers, low immune activation is therefore the consequence of the rapid CD8+ T-cell mediated immune control generated after acute infection. This distinct strategy adopted by PECs may help identify pathways that facilitate remission in posttreatment controllers, in whom protective HLA-I molecules are not the main factor.
Collapse
Affiliation(s)
| | - Jane Millar
- Department of Paediatrics, University of Oxford, Oxford, UK
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Emily Adland
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Maximilian Muenchhoff
- Max von Pettenkofer-Institute, Department of Virology, Ludwig-Maximilians-University
- German Center for Infection Research (DZIF)
| | - Julia Roider
- German Center for Infection Research (DZIF)
- Department of Infectious Diseases, Ludwig-Maximilians-University, Munich, Germany
| | - Claudia Fortuny Guash
- Unidad de Enfermedades Infecciosas, Servicio de Pediatría, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
| | | | - Beatriz Thomé
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Medicina Preventiva, São Paulo, Brazil
| | | | | | - Alasdair Bamford
- Great Ormond Street Hospital for Children NHS Foundation Trust
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Christian Brander
- IrsiCaixa - AIDS Research Institute, Badalona, Spain
- Universitat de Vic-Universitat Central de Catalunya, Vic
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Mary Carrington
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts
- Basic Science Program, Frederick National Laboratory for Cancer Research in the Laboratory of Integrative Cancer Immunology, Bethesda, Maryland, USA
| | - Javier Martinez-Picado
- IrsiCaixa - AIDS Research Institute, Badalona, Spain
- Universitat de Vic-Universitat Central de Catalunya, Vic
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - John Frater
- Nuffield Department of Medicine, University of Oxford
- Oxford NIHR Biomedical Research Centre, Oxford
| | | | - Philip Goulder
- Department of Paediatrics, University of Oxford, Oxford, UK
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa
| |
Collapse
|
56
|
Calvet-Mirabent M, Claiborne DT, Deruaz M, Tanno S, Serra C, Delgado-Arévalo C, Sánchez-Cerrillo I, de Los Santos I, Sanz J, García-Fraile L, Sánchez-Madrid F, Alfranca A, Muñoz-Fernández MÁ, Allen TM, Buzón MJ, Balazs A, Vrbanac V, Martín-Gayo E. Poly I:C and STING agonist-primed DC increase lymphoid tissue polyfunctional HIV-1-specific CD8 + T cells and limit CD4 + T cell loss in BLT mice. Eur J Immunol 2021; 52:447-461. [PMID: 34935145 DOI: 10.1002/eji.202149502] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/19/2021] [Accepted: 12/14/2021] [Indexed: 11/11/2022]
Abstract
Effective function of CD8+ T cells and enhanced innate activation of dendritic cells (DC) in response to HIV-1 is linked to protective antiviral immunity in controllers. Manipulation of DC targeting the master regulator TANK-binding Kinase 1 (TBK1) might be useful to acquire controller-like properties. Here, we evaluated the impact of the combination of 2´3´-c´diAM(PS)2 and Poly I:C as potential adjuvants capable of potentiating DC´s abilities to induce polyfunctional HIV-1 specific CD8+ T cell responses in vitro and in vivo using a humanized BLT mouse model. Adjuvant combination enhanced TBK-1 phosphorylation and IL-12 and IFNβ expression on DC and increased their ability to activate polyfunctional HIV-1-specific CD8+ T cells in vitro. Moreover, higher proportions of hBLT mice vaccinated with ADJ-DC exhibited less severe CD4+ T cell depletion following HIV-1 infection compared to control groups. This was associated with infiltration of CD8+ T cells in the white pulp from the spleen, reduced spread of infected p24+ cells to lymph node and with preserved abilities of CD8+ T cells from the spleen and blood of vaccinated animals to induce specific polyfunctional responses upon antigen stimulation. Therefore, priming of DC with Poly I:C and STING agonists might be useful for future HIV-1 vaccine studies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Marta Calvet-Mirabent
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa.,Universidad Autónoma of Madrid, Medicine Department Spain
| | | | - Maud Deruaz
- Human Immune System Mouse Program from Massachusetts General Hospital, Boston.,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Serah Tanno
- Ragon Institute of MGH, MIT and Harvard.,Human Immune System Mouse Program from Massachusetts General Hospital, Boston
| | - Carla Serra
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona
| | - Cristina Delgado-Arévalo
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa.,Universidad Autónoma of Madrid, Medicine Department Spain
| | - Ildefonso Sánchez-Cerrillo
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa
| | - Ignacio de Los Santos
- Infectious Diseases Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa
| | - Jesús Sanz
- Infectious Diseases Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa
| | - Lucio García-Fraile
- Infectious Diseases Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa
| | - Francisco Sánchez-Madrid
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa.,Universidad Autónoma of Madrid, Medicine Department Spain
| | - Arantzazu Alfranca
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa
| | - María Ángeles Muñoz-Fernández
- Immunology Section, Instituto Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón. Madrid, Spain
| | | | - Maria J Buzón
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona
| | - Alejandro Balazs
- Ragon Institute of MGH, MIT and Harvard.,Human Immune System Mouse Program from Massachusetts General Hospital, Boston
| | - Vladimir Vrbanac
- Ragon Institute of MGH, MIT and Harvard.,Human Immune System Mouse Program from Massachusetts General Hospital, Boston
| | - Enrique Martín-Gayo
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa.,Universidad Autónoma of Madrid, Medicine Department Spain
| |
Collapse
|
57
|
Early Response of CD8+ T Cells in COVID-19 Patients. J Pers Med 2021; 11:jpm11121291. [PMID: 34945761 PMCID: PMC8704882 DOI: 10.3390/jpm11121291] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Healthy and controlled immune response in COVID-19 is crucial for mild forms of the disease. Although CD8+ T cells play important role in this response, there is still a lack of studies showing the gene expression profiles in those cells at the beginning of the disease as potential predictors of more severe forms after the first week. We investigated a proportion of different subpopulations of CD8+ T cells and their gene expression patterns for cytotoxic proteins (perforin-1 (PRF1), granulysin (GNLY), granzyme B (GZMB), granzyme A (GZMA), granzyme K (GZMK)), cytokine interferon-γ (IFN-γ), and apoptotic protein Fas ligand (FASL) in CD8+ T cells from peripheral blood in first weeks of SARS-CoV-2 infection. Sixteen COVID-19 patients and nine healthy controls were included. The absolute counts of total lymphocytes (p = 0.007), CD3+ (p = 0.05), and CD8+ T cells (p = 0.01) in COVID-19 patients were significantly decreased compared to healthy controls. In COVID-19 patients in CD8+ T cell compartment, we observed lower frequency effector memory 1 (EM1) (p = 0.06) and effector memory 4 (EM4) (p < 0.001) CD8+ T cells. Higher mRNA expression of PRF1 (p = 0.05) and lower mRNA expression of FASL (p = 0.05) at the fifth day of the disease were found in COVID-19 patients compared to healthy controls. mRNA expression of PRF1 (p < 0.001) and IFN-γ (p < 0.001) was significantly downregulated in the first week of disease in COVID-19 patients who progressed to moderate and severe forms after the first week, compared to patients with mild symptoms during the entire disease course. GZMK (p < 0.01) and FASL (p < 0.01) mRNA expression was downregulated in all COVID-19 patients compared to healthy controls. Our results can lead to a better understanding of the inappropriate immune response of CD8+ T cells in SARS-CoV2 with the faster progression of the disease.
Collapse
|
58
|
Schreiber S, Honz M, Mamozai W, Kurktschiev P, Schiemann M, Witter K, Moore E, Zielinski C, Sette A, Protzer U, Wisskirchen K. Characterization of a library of 20 HBV-specific MHC class II-restricted T cell receptors. Mol Ther Methods Clin Dev 2021; 23:476-489. [PMID: 34853796 PMCID: PMC8605085 DOI: 10.1016/j.omtm.2021.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/27/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023]
Abstract
CD4+ T cells play an important role in the immune response against cancer and infectious diseases. However, mechanistic details of their helper function in hepatitis B virus (HBV) infection in particular, or their advantage for adoptive T cell therapy remain poorly understood as experimental and therapeutic tools are missing. Therefore, we identified, cloned, and characterized a comprehensive library of 20 MHC class II-restricted HBV-specific T cell receptors (TCRs) from donors with acute or resolved HBV infection. The TCRs were restricted by nine different MHC II molecules and specific for eight different epitopes derived from intracellularly processed HBV envelope, core, and polymerase proteins. Retroviral transduction resulted in a robust expression of all TCRs on primary T cells. A high functional avidity was measured for all TCRs specific for epitopes S17, S21, S36, and P774 (half-maximal effective concentration [EC50] <10 nM), or C61 and preS9 (EC50 <100 nM). Eight TCRs recognized peptide variants of HBV genotypes A to D. Both CD4+ and CD8+ T cells transduced with the MHC II-restricted TCRs were polyfunctional, producing interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-2, and granzyme B (GrzB), and killed peptide-loaded target cells. Our set of MHC class II-restricted TCRs represents an important tool for elucidating CD4+ T cell help in viral infection with potential benefit for T cell therapy.
Collapse
|
59
|
Notsu K, El Daous H, Mitoma S, Norimine J, Sekiguchi S. A pooled testing system to rapidly identify cattle carrying the elite controller BoLA-DRB3*009:02 haplotype against bovine leukemia virus infection. HLA 2021; 99:12-24. [PMID: 34837483 PMCID: PMC9543338 DOI: 10.1111/tan.14502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 11/30/2022]
Abstract
As genetically resistant individuals, the “elite controllers” (ECs) of human immunodeficiency virus infection have been focused on as the keys to developing further functional treatments in medicine. In the livestock production field, identifying the ECs of bovine leukemia virus (BLV) infection in cattle is desired to stop BLV transmission chains on farms. Cattle carrying the bovine leukocyte antigen (BoLA)‐DRB3*009:02 allele (DRB3*009:02) have a strong possibility of being BLV ECs. Most of cattle carrying this allele maintain undetectable BLV proviral loads and do not shed virus even when infected. BLV ECs can act as transmission barriers when placed between uninfected and infected cattle in a barn. To identify cattle carrying DRB3*009:02 in large populations more easily, we developed a pooled testing system. It employs a highly sensitive, specific real‐time PCR assay and TaqMan MGB probes (DRB3*009:02‐TaqMan assay). Using this system, we determined the percentage of DRB3*009:02‐carrying cattle on Kyushu Island, Japan. Our pooled testing system detected cattle carrying the DRB3*009:02 allele from a DNA pool containing one DRB3*009:02‐positive animal and 29 cattle with other alleles. Its capacity is sufficient for herd‐level screening for DRB3*009:02‐carrying cattle. The DRB3*009:02‐TaqMan assay showed high‐discriminative sensitivity and specificity toward DRB3*009:02, making it suitable for identifying DRB3*009:02‐carrying cattle in post‐screening tests on individuals. We determined that the percentage of DRB3*009:02‐carrying cattle in Kyushu Island was 10.56%. With its ease of use and reliable detection, this new method strengthens the laboratory typing for DRB3*009:02‐carrying cattle. Thus, our findings support the use of BLV ECs in the field.
Collapse
Affiliation(s)
- Kosuke Notsu
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hala El Daous
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan.,Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Shuya Mitoma
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan
| | - Junzo Norimine
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan.,Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan
| | - Satoshi Sekiguchi
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan.,Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
60
|
Zanella I, Focà E, Degli-Antoni M, Castelli F, Quiros-Roldan E. An HIV elite controller patient carrying the homozygous H63D variant in the homeostatic iron regulator gene: A case report. Medicine (Baltimore) 2021; 100:e27732. [PMID: 34766580 PMCID: PMC10545298 DOI: 10.1097/md.0000000000027732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/14/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
RATIONALE HIV elite controllers represent a rare subset of persons living with HIV, able to spontaneously control viral replication without antiviral therapy. HLA-B∗57 and HLA-B∗27 alleles are associated to efficient polyfunctional CD8+ T-cell response and are overrepresented in elite controllers but these alleles alone incompletely explain spontaneous HIV replication control in these subjects. Further mechanisms involved in innate and adaptive immune response and host genetics may contribute to this control. In this context, the homeostatic iron regulator (HFE) gene encodes a major histocompatibility complex-class-I-like molecule involved in both innate immunity, acting also through autophagy regulation, and iron homeostasis, strictly related to immune functions and susceptibility to infections. PATIENT CONCERNS Homozygousity for the p.His63Asp (H63D) variant in the HFE gene was identified in an 80-year-old HIV-infected woman with spontaneous control of viral replication. DIAGNOSIS HIV-1 RNA was undetectable in patient's serum with a routine assay and an ultra-sensitive assay (<1 copy/mL) during the 30 years follow-up. CD4+ and CD8+ T cell counts were stable and normal during all this period. INTERVENTIONS The patient had a history of absence of any physical ailment and no antiviral therapy has been prescribed during the 30 years of follow-up. The subject did not harbor HLA-B∗57 and HLA-B∗27 alleles. HFE gene was sequenced by Sanger, as part of a larger study on a cohort of HIV infected patients, aged >65 years and screened for polymorphisms in genes belonging to several pathways involved in neuroinflammation. OUTCOMES The woman had CD4+ and CD8+ T cell normal values and spontaneously controlled serum HIV-1 RNA levels for 30 years. LESSONS We assume that the interplay between the HFE H63D variant in homozygosity and innate immunity, perhaps through autophagy regulation, could play a role in HIV-1 replication control in our patient. This hypothesis needs to be explored in in vitro and in vivo studies. Understanding mechanisms involved in spontaneous control of HIV-1 replication remains indeed a challenge due to its possible implications for HIV cure research.
Collapse
Affiliation(s)
- Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia, Clinical Chemistry Laboratory, Cytogenetics and Molecular Genetics Section, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Emanuele Focà
- University Division of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili, Brescia, Italy
| | - Melania Degli-Antoni
- University Division of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili, Brescia, Italy
| | - Francesco Castelli
- University Division of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili, Brescia, Italy
| | - Eugenia Quiros-Roldan
- University Division of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
61
|
Two distinct mechanisms leading to loss of virological control in the rare group of antiretroviral therapy-naïve, transiently aviraemic children living with HIV. J Virol 2021; 96:e0153521. [PMID: 34757843 PMCID: PMC8791270 DOI: 10.1128/jvi.01535-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
HIV-specific CD8+ T-cells play a central role in immune control of adult HIV, but their contribution in paediatric infection is less well-characterised. Previously, we identified a group of ART-naïve children with persistently undetectable plasma viraemia, termed 'elite controllers', and a second group who achieved aviraemia only transiently. To investigate the mechanisms of failure to maintain aviraemia, we characterized in three transient aviraemics (TAs), each of whom expressed the disease-protective HLA-B*81:01, longitudinal HIV-specific T-cell activity and viral sequences. In two TAs, a CD8+ T-cell response targeting the immunodominant epitope TPQDLNTML ('Gag-TL9') was associated with viral control, followed by viral rebound and the emergence of escape variants with lower replicative capacity. Both TAs mounted variant-specific responses, but only at low functional avidity, resulting in immunological progression. By contrast, in TA-3, intermittent viraemic episodes followed aviraemia without virus escape or a diminished CD4+ T-cell count. High quality and magnitude of the CD8+ T-cell response was associated with aviraemia. We therefore identify two distinct mechanisms of loss of viral control. In one scenario, CD8+ T-cell responses initially cornered low replicative capacity escape variants, but with insufficient avidity to prevent viraemia and disease progression. In the other, loss of viral control was associated neither with virus escape nor progression, but with a decrease in the quality of the CD8+ T-cell response, followed by recovery of viral control in association with improved antiviral response. These data suggest the potential for a consistently strong and polyfunctional antiviral response to achieve long-term viral control without escape. IMPORTANCE Very early initiation of antiretroviral therapy (ART) in paediatric HIV infection offers a unique opportunity to limit the size and diversity of the viral reservoir. However, only exceptionally is ART alone sufficient to achieve remission. Additional interventions are therefore required that likely include contributions from host immunity. The HIV-specific T-cell response plays a central role in immune control of adult HIV, often mediated through protective alleles such as HLA-B*57/58:01/81:01. However, due to the tolerogenic and type 2 biased immune response in early life, HLA-I-mediated immune suppression of viraemia is seldom observed in children. We describe a rare group of HLA-B*81:01-positive, ART-naïve children who achieved aviraemia, albeit only transiently, and investigate the role of the CD8+ T-cell response in the establishment and loss of viral control. We identify a mechanism by which the HIV-specific response can achieve viraemic control without viral escape, that can be explored in strategies to achieve remission.
Collapse
|
62
|
Moysi E, Darko S, Gea-Mallorquí E, Petrovas C, Almeida JR, Wolinsky D, Peng Y, Jaye A, Stewart-Jones G, Douek DC, Koup RA, Dong T, Rowland-Jones S. Clonotypic architecture of a Gag-specific CD8+ T-cell response in chronic human HIV-2 infection. Eur J Immunol 2021; 51:2485-2500. [PMID: 34369597 DOI: 10.1002/eji.202048931] [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: 10/03/2020] [Revised: 06/07/2021] [Accepted: 08/05/2021] [Indexed: 11/08/2022]
Abstract
The dynamics of T-cell receptor (TCR) selection in chronic HIV-1 infection, and its association with clinical outcome, is well documented for an array of MHC-peptide complexes and disease stages. However, the factors that may contribute to the selection and expansion of CD8+ T-cells in chronic HIV-2 infection, especially at clonal level remain unclear. To address this question, we undertook a detailed molecular characterization of the clonotypic architecture of an HLA-B*3501 restricted Gag -specific CD8+ T-cell response in donors chronically infected with HIV-2 using a combination of flow cytometry, tetramer-specific CD8+ TCR clonotyping and in vitro assays. We show that the response to the NY9 epitope is hierarchical and narrow in terms of T-cell receptor alpha (TCRA) and beta (TCRB) gene usage yet clonotypically diverse. Furthermore, clonotypic dominance in shared origin cytotoxic T lymphocyte (CTL) clones was associated with a greater magnitude of cytokine production and antigen sensitivity at limiting antigen dilution as well as enhanced cross-reactivity for known HIV-2 variants. Hence, our data suggest that effector mobilization and expansion in human chronic HIV-2 infection may be linked to the qualitative features of specific CD8+ T-cell clonotypes, which could have implications for viral control and disease outcome. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Eirini Moysi
- Tissue Analysis Core, Vaccine Research Centre, Bethesda, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Samuel Darko
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, 20892, USA
| | - Ester Gea-Mallorquí
- Viral Immunology Unit, Nuffield Department of Medicine, Headington, Oxford, OX3 7FZ, United Kingdom
| | - Constantinos Petrovas
- Tissue Analysis Core, Vaccine Research Centre, Bethesda, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Jorge R Almeida
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, 20892, USA
| | - David Wolinsky
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, 20892, USA
| | - Yanchun Peng
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, United Kingdom
| | - Assan Jaye
- MRC Laboratories, The Gambia, PO Box 273, West Africa
| | - Guillaume Stewart-Jones
- Structural Biology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, 20892, USA
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Tao Dong
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, United Kingdom
| | - Sarah Rowland-Jones
- Viral Immunology Unit, Nuffield Department of Medicine, Headington, Oxford, OX3 7FZ, United Kingdom
| |
Collapse
|
63
|
Bakker NAM, Rotman J, van Beurden M, Zijlmans HJM, van Ruiten M, Samuels S, Nuijen B, Beijnen J, De Visser K, Haanen J, Schumacher T, de Gruijl TD, Jordanova ES, Kenter GG, van den Berg JH, van Trommel NE. HPV-16 E6/E7 DNA tattoo vaccination using genetically optimized vaccines elicit clinical and immunological responses in patients with usual vulvar intraepithelial neoplasia (uVIN): a phase I/II clinical trial. J Immunother Cancer 2021; 9:jitc-2021-002547. [PMID: 34341131 PMCID: PMC8330588 DOI: 10.1136/jitc-2021-002547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 11/23/2022] Open
Abstract
Background Usual vulvar intraepithelial neoplasia (uVIN) is a premalignancy caused by persistent infection with high-risk types of human papillomavirus (HPV), mainly type 16. Even though different treatment modalities are available (eg, surgical excision, laser evaporation or topical application of imiquimod), these treatments can be mutilating, patients often have recurrences and 2%–8% of patients develop vulvar carcinoma. Therefore, immunotherapeutic strategies targeting the pivotal oncogenic HPV proteins E6 and E7 are being explored to repress carcinogenesis. Method In this phase I/II clinical trial, 14 patients with HPV16+ uVIN were treated with a genetically enhanced DNA vaccine targeting E6 and E7. Safety, clinical responses and immunogenicity were assessed. Patients received four intradermal HPV-16 E6/E7 DNA tattoo vaccinations, with a 2-week interval, alternating between both upper legs. Biopsies of the uVIN lesions were taken at screening and +3 months after last vaccination. Digital photography of the vulva was performed at every check-up until 12 months of follow-up for measurement of the lesions. HPV16-specific T-cell responses were measured in blood over time in ex vivo reactivity assays. Results Vaccinations were well tolerated, although one grade 3 suspected unexpected serious adverse reaction was observed. Clinical responses were observed in 6/14 (43%) patients, with 2 complete responses and 4 partial responses (PR). 5/14 patients showed HPV-specific T-cell responses in blood, measured in ex vivo reactivity assays. Notably, all five patients with HPV-specific T-cell responses had a clinical response. Conclusions Our results indicate that HPV-16 E6/E7 DNA tattoo vaccination is a biologically active and safe treatment strategy in patients with uVIN, and suggest that T-cell reactivity against the HPV oncogenes is associated with clinical benefit. Trial registration number NTR4607.
Collapse
Affiliation(s)
- Noor Alida Maria Bakker
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Division of Tumor Biology and Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Jossie Rotman
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marc van Beurden
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Henry J Maa Zijlmans
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Maartje van Ruiten
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Sanne Samuels
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Jos Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Karin De Visser
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - John Haanen
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Ton Schumacher
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, -Cancer Center Amsterdam, Amsterdam UMC-Vrije Universiteit, Amsterdam, The Netherlands
| | - Ekaterina S Jordanova
- Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gemma G Kenter
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Joost H van den Berg
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Nienke E van Trommel
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| |
Collapse
|
64
|
Del Valle A, Acosta-Rivero N, Laborde RJ, Cruz-Leal Y, Cabezas S, Luzardo MC, Alvarez C, Labrada M, Rodríguez A, Rodríguez GL, Raymond J, Nogueira CV, Grubaugh D, Fernández LE, Higgins D, Lanio ME. Sticholysin II shows similar immunostimulatory properties to LLO stimulating dendritic cells and MHC-I restricted T cell responses of heterologous antigen. Toxicon 2021; 200:38-47. [PMID: 34237340 DOI: 10.1016/j.toxicon.2021.06.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 06/22/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
Induction of CD8+ T cell responses against tumor cells and intracellular pathogens is an important goal of modern vaccinology. One approach of translational interest is the use of liposomes encapsulating pore-forming proteins (PFPs), such as Listeriolysin O (LLO), which has shown efficacy at priming strong and sustained CD8+ T cell responses. Recently, we have demonstrated that Sticholysin II (StII), a PFP from the sea anemone Stichodactyla helianthus, co-encapsulated into liposomes with ovalbumin (OVA) was able to stimulate, antigen presenting cells, antigen-specific CD8+ T cells and anti-tumor activity in mice. In the present study, we aimed to compare StII and LLO in terms of their abilities to stimulate dendritic cells and to induce major histocompatibility complex (MHC) class I restricted T cell responses against OVA. Interestingly, StII exhibited similar abilities to LLO in vitro of inducing dendritic cells maturation, as measured by increased expression of CD40, CD80, CD86 and MHC-class II molecules, and of stimulating OVA cross-presentation to a CD8+ T cell line. Remarkably, using an ex vivo Enzyme-Linked ImmunoSpot Assay (ELISPOT) to monitor gamma interferon (INF-γ) producing effector memory CD8+ T cells, liposomal formulations containing either StII or LLO induced comparable frequencies of OVA-specific INF-γ producing CD8+ T cells in mice that were sustained in time. However, StII-containing liposomes stimulated antigen-specific memory CD8+ T cells with a higher potential to secrete IFN-γ than liposomes encapsulating LLO. This StII immunostimulatory property further supports its use for the rational design of T cell vaccines against cancers and intracellular pathogens. In summary, this study indicates that StII has immunostimulatory properties similar to LLO, despite being evolutionarily distant PFPs.
Collapse
Affiliation(s)
- A Del Valle
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - N Acosta-Rivero
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba.
| | - R J Laborde
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - Y Cruz-Leal
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - S Cabezas
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - M C Luzardo
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - C Alvarez
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - M Labrada
- Center of Molecular Immunology (CIM), Playa, La Habana, Cuba
| | - A Rodríguez
- Center of Molecular Immunology (CIM), Playa, La Habana, Cuba
| | - G L Rodríguez
- Center of Molecular Immunology (CIM), Playa, La Habana, Cuba
| | - J Raymond
- Center of Molecular Immunology (CIM), Playa, La Habana, Cuba
| | | | - D Grubaugh
- Harvard Medical School, Harvard University, USA
| | - L E Fernández
- Center of Molecular Immunology (CIM), Playa, La Habana, Cuba
| | - D Higgins
- Harvard Medical School, Harvard University, USA
| | - M E Lanio
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba.
| |
Collapse
|
65
|
Sandberg JT, Varnaitė R, Christ W, Chen P, Muvva JR, Maleki KT, García M, Dzidic M, Folkesson E, Skagerberg M, Ahlén G, Frelin L, Sällberg M, Eriksson LI, Rooyackers O, Sönnerborg A, Buggert M, Björkström NK, Aleman S, Strålin K, Klingström J, Ljunggren H, Blom K, Gredmark‐Russ S. SARS-CoV-2-specific humoral and cellular immunity persists through 9 months irrespective of COVID-19 severity at hospitalisation. Clin Transl Immunology 2021; 10:e1306. [PMID: 34257967 PMCID: PMC8256672 DOI: 10.1002/cti2.1306] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES Humoral and cellular immunity to SARS-CoV-2 following COVID-19 will likely contribute to protection from reinfection or severe disease. It is therefore important to characterise the initiation and persistence of adaptive immunity to SARS-CoV-2 amidst the ongoing pandemic. METHODS Here, we conducted a longitudinal study on hospitalised moderate and severe COVID-19 patients from the acute phase of disease into convalescence at 5 and 9 months post-symptom onset. Utilising flow cytometry, serological assays as well as B cell and T cell FluoroSpot assays, we assessed the magnitude and specificity of humoral and cellular immune responses during and after human SARS-CoV-2 infection. RESULTS During acute COVID-19, we observed an increase in germinal centre activity, a substantial expansion of antibody-secreting cells and the generation of SARS-CoV-2-neutralising antibodies. Despite gradually decreasing antibody levels, we show persistent, neutralising antibody titres as well as robust specific memory B cell responses and polyfunctional T cell responses at 5 and 9 months after symptom onset in both moderate and severe COVID-19 patients. CONCLUSION Our findings describe the initiation and, importantly, persistence of cellular and humoral SARS-CoV-2-specific immunological memory in hospitalised COVID-19 patients long after recovery, likely contributing towards protection against reinfection.
Collapse
Affiliation(s)
- John Tyler Sandberg
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Renata Varnaitė
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Wanda Christ
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Puran Chen
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Jagadeeswara R Muvva
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Kimia T Maleki
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Marina García
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Majda Dzidic
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Elin Folkesson
- Department of Infectious DiseasesKarolinska University HospitalStockholmSweden
- Department of Medicine Solna, Division of Infectious DiseasesKarolinska InstitutetStockholmSweden
| | - Magdalena Skagerberg
- Department of Infectious DiseasesKarolinska University HospitalStockholmSweden
- Division of Infectious Diseases and DermatologyDepartment of Medicine HuddingeKarolinska InstitutetStockholmSweden
| | - Gustaf Ahlén
- Division of Clinical Microbiology, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Lars Frelin
- Division of Clinical Microbiology, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Matti Sällberg
- Division of Clinical Microbiology, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Lars I Eriksson
- Function Perioperative Medicine and Intensive CareKarolinska University HospitalStockholmSweden
- Department of Physiology and PharmacologyKarolinska InstitutetStockholmSweden
| | - Olav Rooyackers
- Function Perioperative Medicine and Intensive CareKarolinska University HospitalStockholmSweden
- Department of Clinical Science, Technology and Intervention, Division of Anesthesiology and Intensive CareKarolinska InstitutetStockholmSweden
| | - Anders Sönnerborg
- Department of Infectious DiseasesKarolinska University HospitalStockholmSweden
- Division of Infectious Diseases and DermatologyDepartment of Medicine HuddingeKarolinska InstitutetStockholmSweden
- Division of Clinical Microbiology, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Niklas K Björkström
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
- Department of Clinical MicrobiologyKarolinska University HospitalStockholmSweden
| | - Soo Aleman
- Department of Infectious DiseasesKarolinska University HospitalStockholmSweden
- Division of Infectious Diseases and DermatologyDepartment of Medicine HuddingeKarolinska InstitutetStockholmSweden
| | - Kristoffer Strålin
- Department of Infectious DiseasesKarolinska University HospitalStockholmSweden
- Division of Infectious Diseases and DermatologyDepartment of Medicine HuddingeKarolinska InstitutetStockholmSweden
| | - Jonas Klingström
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Hans‐Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Kim Blom
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Sara Gredmark‐Russ
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
- Department of Infectious DiseasesKarolinska University HospitalStockholmSweden
| |
Collapse
|
66
|
SARS-CoV-2-specific T cell memory is sustained in COVID-19 convalescent patients for 10 months with successful development of stem cell-like memory T cells. Nat Commun 2021; 12:4043. [PMID: 34193870 PMCID: PMC8245549 DOI: 10.1038/s41467-021-24377-1] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/18/2021] [Indexed: 12/20/2022] Open
Abstract
Memory T cells contribute to rapid viral clearance during re-infection, but the longevity and differentiation of SARS-CoV-2-specific memory T cells remain unclear. Here we conduct ex vivo assays to evaluate SARS-CoV-2-specific CD4+ and CD8+ T cell responses in COVID-19 convalescent patients up to 317 days post-symptom onset (DPSO), and find that memory T cell responses are maintained during the study period regardless of the severity of COVID-19. In particular, we observe sustained polyfunctionality and proliferation capacity of SARS-CoV-2-specific T cells. Among SARS-CoV-2-specific CD4+ and CD8+ T cells detected by activation-induced markers, the proportion of stem cell-like memory T (TSCM) cells is increased, peaking at approximately 120 DPSO. Development of TSCM cells is confirmed by SARS-CoV-2-specific MHC-I multimer staining. Considering the self-renewal capacity and multipotency of TSCM cells, our data suggest that SARS-CoV-2-specific T cells are long-lasting after recovery from COVID-19, thus support the feasibility of effective vaccination programs as a measure for COVID-19 control. T cells are instrumental to protective immune responses against SARS-CoV-2, the pathogen responsible for the COVID-19 pandemic. Here the authors show that, in convalescent COVID-19 patients, memory T cell responses are detectable up to 317 days post-symptom onset, in which the presence of stem cell-like memory T cells further hints long-lasting immunity.
Collapse
|
67
|
Duerr R, Crosse KM, Valero-Jimenez AM, Dittmann M. SARS-CoV-2 Portrayed against HIV: Contrary Viral Strategies in Similar Disguise. Microorganisms 2021; 9:1389. [PMID: 34198973 PMCID: PMC8307803 DOI: 10.3390/microorganisms9071389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 11/16/2022] Open
Abstract
SARS-CoV-2 and HIV are zoonotic viruses that rapidly reached pandemic scale, causing global losses and fear. The COVID-19 and AIDS pandemics ignited massive efforts worldwide to develop antiviral strategies and characterize viral architectures, biological and immunological properties, and clinical outcomes. Although both viruses have a comparable appearance as enveloped viruses with positive-stranded RNA and envelope spikes mediating cellular entry, the entry process, downstream biological and immunological pathways, clinical outcomes, and disease courses are strikingly different. This review provides a systemic comparison of both viruses' structural and functional characteristics, delineating their distinct strategies for efficient spread.
Collapse
Affiliation(s)
- Ralf Duerr
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; (K.M.C.); (A.M.V.-J.); (M.D.)
| | | | | | | |
Collapse
|
68
|
Gilfillan CB, Hebeisen M, Rufer N, Speiser DE. Constant regulation for stable CD8 T-cell functional avidity and its possible implications for cancer immunotherapy. Eur J Immunol 2021; 51:1348-1360. [PMID: 33704770 PMCID: PMC8252569 DOI: 10.1002/eji.202049016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/16/2020] [Accepted: 03/05/2021] [Indexed: 12/30/2022]
Abstract
The functional avidity (FA) of cytotoxic CD8 T cells impacts strongly on their functional capabilities and correlates with protection from infection and cancer. FA depends on TCR affinity, downstream signaling strength, and TCR affinity-independent parameters of the immune synapse, such as costimulatory and inhibitory receptors. The functional impact of coreceptors on FA remains to be fully elucidated. Despite its importance, FA is infrequently assessed and incompletely understood. There is currently no consensus as to whether FA can be enhanced by optimized vaccine dose or boosting schedule. Recent findings suggest that FA is remarkably stable in vivo, possibly due to continued signaling modulation of critical receptors in the immune synapse. In this review, we provide an overview of the current knowledge and hypothesize that in vivo, codominant T cells constantly "equalize" their FA for similar function. We present a new model of constant FA regulation, and discuss practical implications for T-cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Connie B. Gilfillan
- Department of OncologyUniversity Hospital and University of LausanneLausanneSwitzerland
| | - Michael Hebeisen
- Department of OncologyUniversity Hospital and University of LausanneLausanneSwitzerland
| | - Nathalie Rufer
- Department of OncologyUniversity Hospital and University of LausanneLausanneSwitzerland
| | - Daniel E. Speiser
- Department of OncologyUniversity Hospital and University of LausanneLausanneSwitzerland
| |
Collapse
|
69
|
Yang H, Llano A, Cedeño S, von Delft A, Corcuera A, Gillespie GM, Knox A, Leneghan DB, Frater J, Stöhr W, Fidler S, Mothe B, Mak J, Brander C, Ternette N, Dorrell L. Incoming HIV virion-derived Gag Spacer Peptide 2 (p1) is a target of effective CD8 + T cell antiviral responses. Cell Rep 2021; 35:109103. [PMID: 33979627 DOI: 10.1016/j.celrep.2021.109103] [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: 10/06/2020] [Revised: 02/20/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022] Open
Abstract
Persistence of HIV through integration into host DNA in CD4+ T cells presents a major barrier to virus eradication. Viral integration may be curtailed when CD8+ T cells are triggered to kill infected CD4+ T cells through recognition of histocompatibility leukocyte antigen (HLA) class I-bound peptides derived from incoming virions. However, this has been reported only in individuals with "beneficial" HLA alleles that are associated with superior HIV control. Through interrogation of the pre-integration immunopeptidome, we obtain proof of early presentation of a virion-derived HLA-A∗02:01-restricted epitope, FLGKIWPSH (FH9), located in Gag Spacer Peptide 2 (SP2). FH9-specific CD8+ T cell responses are detectable in individuals with primary HIV infection and eliminate HIV-infected CD4+ T cells prior to virus production in vitro. Our data show that non-beneficial HLA class I alleles can elicit an effective antiviral response through early presentation of HIV virion-derived epitopes and also demonstrate the importance of SP2 as an immune target.
Collapse
Affiliation(s)
- Hongbing Yang
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK; National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK; Research In Viral Eradication of Reservoirs (RIVER) trial study group.
| | - Anuska Llano
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Samandhy Cedeño
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Annette von Delft
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK; Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Angelica Corcuera
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | | | - Andrew Knox
- Immunocore Ltd, Milton, Abingdon OX14 4RY, UK
| | | | - John Frater
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK; National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK; Research In Viral Eradication of Reservoirs (RIVER) trial study group
| | - Wolfgang Stöhr
- Medical Research Council Clinical Trials Unit, University College London, London WC1V 6LJ, UK; Research In Viral Eradication of Reservoirs (RIVER) trial study group
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London, National Institute for Health Research Imperial Biomedical Research Centre, London W2 1NY, UK; Research In Viral Eradication of Reservoirs (RIVER) trial study group
| | - Beatriz Mothe
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), 08500 Vic, Spain; Fundació Lluita contra la Sida, Infectious Disease Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Johnson Mak
- Institute for Glycomics, Griffith University Gold Coast, Southport QLD 4215, Australia
| | - Christian Brander
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), 08500 Vic, Spain; Institució Catalana de Recerca I Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Nicola Ternette
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Lucy Dorrell
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK; National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK; Immunocore Ltd, Milton, Abingdon OX14 4RY, UK; Research In Viral Eradication of Reservoirs (RIVER) trial study group.
| |
Collapse
|
70
|
Optimal Maturation of the SIV-Specific CD8 + T Cell Response after Primary Infection Is Associated with Natural Control of SIV: ANRS SIC Study. Cell Rep 2021; 32:108174. [PMID: 32966788 DOI: 10.1016/j.celrep.2020.108174] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 07/10/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022] Open
Abstract
Highly efficient CD8+ T cells are associated with natural HIV control, but it has remained unclear how these cells are generated and maintained. We have used a macaque model of spontaneous SIVmac251 control to monitor the development of efficient CD8+ T cell responses. Our results show that SIV-specific CD8+ T cells emerge during primary infection in all animals. The ability of CD8+ T cells to suppress SIV is suboptimal in the acute phase but increases progressively in controller macaques before the establishment of sustained low-level viremia. Controller macaques develop optimal memory-like SIV-specific CD8+ T cells early after infection. In contrast, a persistently skewed differentiation phenotype characterizes memory SIV-specific CD8+ T cells in non-controller macaques. Accordingly, the phenotype of SIV-specific CD8+ T cells defined early after infection appears to favor the development of protective immunity in controllers, whereas SIV-specific CD8+ T cells in non-controllers fail to gain antiviral potency, feasibly as a consequence of early defects imprinted in the memory pool.
Collapse
|
71
|
García-López M, Lens S, Pallett LJ, Testoni B, Rodríguez-Tajes S, Mariño Z, Bartres C, García-Pras E, Leonel T, Perpiñán E, Lozano JJ, Rodríguez-Frías F, Koutsoudakis G, Zoulim F, Maini MK, Forns X, Pérez-Del-Pulgar S. Viral and immune factors associated with successful treatment withdrawal in HBeAg-negative chronic hepatitis B patients. J Hepatol 2021; 74:1064-1074. [PMID: 33278456 PMCID: PMC8062913 DOI: 10.1016/j.jhep.2020.11.043] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/28/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Factors associated with a successful outcome upon nucleos(t)ide analogue (NA) treatment withdrawal in HBeAg-negative chronic hepatitis B (CHB) patients have yet to be clarified. The objective of this study was to analyse the HBV-specific T cell response, in parallel with peripheral and intrahepatic viral parameters, in patients undergoing NA discontinuation. METHODS Twenty-seven patients without cirrhosis with HBeAg-negative CHB with complete viral suppression (>3 years) were studied prospectively. Intrahepatic HBV-DNA (iHBV-DNA), intrahepatic HBV-RNA (iHBV-RNA), and covalently closed circular DNA (cccDNA) were quantified at baseline. Additionally, serum markers (HBV-DNA, HBsAg, HBV core-related antigen [HBcrAg] and HBV-RNA) and HBV-specific T cell responses were analysed at baseline and longitudinally throughout follow-up. RESULTS After a median follow-up of 34 months, 22/27 patients (82%) remained off-therapy, of whom 8 patients (30% of the total cohort) lost HBsAg. Baseline HBsAg significantly correlated with iHBV-DNA and iHBV-RNA, and these parameters were lower in patients who lost HBsAg. All patients had similar levels of detectable cccDNA regardless of their clinical outcome. Patients achieving functional cure had baseline HBsAg levels ≤1,000 IU/ml. Similarly, an increased frequency of functional HBV-specific CD8+ T cells at baseline was associated with sustained viral control off treatment. These HBV-specific T cell responses persisted, but did not increase, after treatment withdrawal. A similar, but not statistically significant trend, was observed for HBV-specific CD4+ T cell responses. CONCLUSIONS Decreased cccDNA transcription and low HBsAg levels are associated with HBsAg loss upon NA discontinuation in patients with HBeAg-negative CHB. The presence of functional HBV-specific T cells at baseline are associated with a successful outcome after treatment withdrawal. LAY SUMMARY Nucleos(t)ide analogue therapy can be discontinued in a high proportion of chronic hepatitis B patients without cirrhosis. The strength of HBV-specific immune T cell responses may contribute to successful viral control after antiviral treatment interruption. Our comprehensive study provides in-depth data on virological and immunological factors than can help guide individualised therapy in patients with chronic hepatitis B.
Collapse
Affiliation(s)
- Mireia García-López
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Sabela Lens
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Laura J Pallett
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, UK
| | - Barbara Testoni
- INSERM U1052-Cancer Research Center of Lyon (CRCL), University of Lyon, UMR_S1052, CRCL, Lyon, France
| | - Sergio Rodríguez-Tajes
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Zoe Mariño
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Concepción Bartres
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Ester García-Pras
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Thais Leonel
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Elena Perpiñán
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain
| | | | - Francisco Rodríguez-Frías
- Liver Pathology Unit, Department of Biochemistry and Microbiology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, CIBERehd, Barcelona, Spain
| | - George Koutsoudakis
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Fabien Zoulim
- INSERM U1052-Cancer Research Center of Lyon (CRCL), University of Lyon, UMR_S1052, CRCL, Lyon, France
| | - Mala K Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, UK
| | - Xavier Forns
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain.
| | - Sofía Pérez-Del-Pulgar
- Liver Unit, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERehd, Barcelona, Spain.
| |
Collapse
|
72
|
Xie CB, Zhou J, Mackay S, Pober JS. Complement-activated human endothelial cells stimulate increased polyfunctionality in alloreactive T cells. Am J Transplant 2021; 21:1902-1909. [PMID: 33415805 PMCID: PMC8096653 DOI: 10.1111/ajt.16485] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/07/2020] [Accepted: 12/30/2020] [Indexed: 01/25/2023]
Abstract
Antibody-mediated deposition of complement membrane attack complexes (MACs) on IFN-γ-primed human endothelial cells (ECs) triggers autocrine/paracrine IL-1β-mediated EC activation and IL-15 transpresentation to alloreactive effector memory T cells (TEM ), changes that enable ECs to increase T cell proliferation and cytokine release. Here, we report the use of single-cell microchip 32-plex proteomics to more deeply assess the functionality of the activated T cells and dependence upon EC-derived signals. Compared to control ECs, MAC-activated human ECs increase both the frequency and degree of polyfunctionality among both CD4+ and CD8+ -proliferated TEM , assessed as secreted proteins. IFN-γ and TNF-α remain the predominant cytokines made by alloreactive TEM , but a few CD4+ TEM also made IL-4 while more CD8+ TEM made perforin and granzyme B. Increased polyfunctionality was attenuated by treatment of the MAC-activated ECs with anti-IL-15 blocking antibody more effectively than IL-1 receptor blockade. The increased polyfunctionality of T cells resulting from interactions with MAC-activated ECs may further link binding of donor-specific antibody to T cell-mediated allograft pathologies.
Collapse
Affiliation(s)
- Catherine B. Xie
- Dept of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jing Zhou
- IsoPlexis Corporation, Branford, CT, USA
| | | | - Jordan S. Pober
- Dept of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
73
|
Immunologic Control of HIV-1: What Have We Learned and Can We Induce It? Curr HIV/AIDS Rep 2021; 18:211-220. [PMID: 33709324 DOI: 10.1007/s11904-021-00545-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW A large amount of data now exists on the virus-specific immune response associated with spontaneous or induced immunologic control of lentiviruses. This review focuses on how the current understanding of HIV-specific immunity might be leveraged into induction of immunologic control and what further research is needed to accomplish this goal. RECENT FINDINGS During chronic infection, the function most robustly associated with immunologic control of HIV-1 is CD8+ T cell cytotoxic capacity. This function has proven difficult to restore in HIV-specific CD8+ T cells of chronically infected progressors in vitro and in vivo. However, progress has been made in inducing an effective CD8+ T cell response prior to lentiviral infection in the macaque model and during acute lentiviral infection in non-human primates. Further study will likely accelerate the ability to induce an effective CD8+ T cell response as part of prophylactic or therapeutic strategies.
Collapse
|
74
|
Morgan J, Muskat K, Tippalagama R, Sette A, Burel J, Lindestam Arlehamn CS. Classical CD4 T cells as the cornerstone of antimycobacterial immunity. Immunol Rev 2021; 301:10-29. [PMID: 33751597 PMCID: PMC8252593 DOI: 10.1111/imr.12963] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/11/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
Tuberculosis is a significant health problem without an effective vaccine to combat it. A thorough understanding of the immune response and correlates of protection is needed to develop a more efficient vaccine. The immune response against Mycobacterium tuberculosis (Mtb) is complex and involves all aspects of the immune system, however, the optimal protective, non‐pathogenic T cell response against Mtb is still elusive. This review will focus on discussing CD4 T cell immunity against mycobacteria and its importance in Mtb infection with a primary focus on human studies. We will in particular discuss the large heterogeneity of immune cell subsets that have been revealed by recent immunological investigations at an unprecedented level of detail. These studies have identified specific classical CD4 T cell subsets important for immune responses against Mtb in various states of infection. We further discuss the functional attributes that have been linked to the various subsets such as upregulation of activation markers and cytokine production. Another important topic to be considered is the antigenic targets of Mtb‐specific immune responses, and how antigen reactivity is influenced by both disease state and environmental exposure(s). These are key points for both vaccines and immune diagnostics development. Ultimately, these factors are holistically considered in the definition and investigations of what are the correlates on protection and resolution of disease.
Collapse
Affiliation(s)
- Jeffrey Morgan
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Kaylin Muskat
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Rashmi Tippalagama
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Julie Burel
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | | |
Collapse
|
75
|
Alfageme-Abello O, Porret R, Perreau M, Perez L, Muller YD. Chimeric antigen receptor T-cell therapy for HIV cure. Curr Opin HIV AIDS 2021; 16:88-97. [PMID: 33560017 DOI: 10.1097/coh.0000000000000665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Cell-based immunotherapies have made enormous progress over the last decade with the approval of several anti-CD19-chimeric antigen receptor (CAR)-T cell therapies for haemato-oncological diseases. CARs are synthetic receptors comprising an antigen-specific extracellular domain fused to a hinge, transmembrane and intracellular signalling domains. The success obtained with CD19 CAR-T cells rekindled interest in using CAR-T cells to treat HIV seropositive patients. The purpose of this review is to discuss historical and recent developments of anti-HIV CARs. RECENT FINDINGS Since the first description of CD4+-based CARs in the early 90s, new generations of anti-HIV CARs were developed. They target the hetero-trimeric glycoprotein gp120/gp41 and consist of either a CD4+ extracellular domain or a VH/VL segment derived from broadly neutralizing antibodies. Recent efforts were employed in multiplexing CAR specificities, intracellular signalling domains and T cells resistance to HIV. SUMMARY Several new-anti HIV CAR-T cells were successfully tested in preclinical mice models and are now waiting to be evaluated in clinical trials. One of the key parameters to successfully using CAR-T cells in HIV treatment will depend on their capacity to control the HIV reservoir without causing off-targeting activities.
Collapse
Affiliation(s)
- Oscar Alfageme-Abello
- Lausanne University Hospital (CHUV), Department of Medicine, Division of Immunology and Allergy, Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
76
|
Kedzierska K, Koutsakos M. The ABC of Major Histocompatibility Complexes and T Cell Receptors in Health and Disease. Viral Immunol 2021; 33:160-178. [PMID: 32286182 PMCID: PMC7185345 DOI: 10.1089/vim.2019.0184] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A seminal discovery of major histocompatibility complex (MHC) restriction in T cell recognition by Peter Doherty and Rolf Zinkernagel has led to 45 years of exciting research on the mechanisms governing peptide MHC (pMHC) recognition by T cell receptors (TCRs) and their importance in health and disease. T cells provide a significant level of protection against viral, bacterial, and parasitic infections, as well as tumors, hence, the generation of protective T cell responses is a primary goal for cell-mediated vaccines and immunotherapies. Understanding the mechanisms underlying generation of optimal high-avidity effector T cell responses, memory development, maintenance, and recall is of major importance for the rational design of preventative and therapeutic vaccines/immunotherapies. In this review, we summarize the lessons learned over the last four decades and outline our current understanding of the basis and consequences of pMHC/TCR interactions on T cell development and function, and TCR diversity and composition, driving better clinical outcomes and prevention of viral escape. We also discuss the current models of T cell memory formation and determinants of immunodominant T cell responses in animal models and humans. As TCR composition and diversity can affect both the protective capacity of T cells and protection against viral escape, defining the spectrum of TCR selection has implications for improving the functional efficacy of effector T cell responsiveness and memory formation.
Collapse
Affiliation(s)
- Katherine Kedzierska
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| |
Collapse
|
77
|
Adams P, Iserentant G, Servais JY, Vandekerckhove L, Vanham G, Seguin-Devaux C. Cytotoxic CD8+ T Cells Expressing CXCR5 Are Detectable in HIV-1 Elite Controllers After Prolonged In Vitro Peptide Stimulation. Front Immunol 2021; 11:622343. [PMID: 33717056 PMCID: PMC7945035 DOI: 10.3389/fimmu.2020.622343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/21/2020] [Indexed: 11/23/2022] Open
Abstract
Antiretroviral therapy (ART) is not curative as HIV-1 persists in long-lived viral reservoirs. Consequently, patients are dependent on life-long drug adherence with possible side effects. To overcome these limitations strategies of a functional cure aim at ART free viral remission. In this study, we sought to identify detailed subsets of anti-viral CD8+ T cell immunity linked to natural long-term control of HIV-1 infection. Here, we analyzed HIV controllers and ART suppressed progressors for in vitro viral suppressive capacity (VSC) at baseline and after peptide stimulation. Functional properties and phenotypes of CD8+ T cells were assessed by IFN-γ ELISPOT and 18 color flow cytometry. HIV controllers showed significantly increased suppression at baseline as well as after peptide stimulation. IFN-γ secretion and the proliferation marker Ki67 positively correlated with VSC. Moreover, the detailed phenotype of three distinct multifunctional memory CD8+ T cell subsets were specific traits of HIV controllers of which two correlated convincingly with VSC. Our results underline the importance of multifunctional CD8+ T cell responses during natural control. Especially the role of CXCR5 expressing cytotoxic subsets emphasizes potential surveillance in sites of reservoir persistence and demand further study.
Collapse
Affiliation(s)
- Philipp Adams
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Departments of Biomedical and Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Gilles Iserentant
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Jean-Yves Servais
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | | | - Guido Vanham
- Departments of Biomedical and Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Carole Seguin-Devaux
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | | |
Collapse
|
78
|
Zhou H, Yang J, Tian J, Wang S. CD8 + T Lymphocytes: Crucial Players in Sjögren's Syndrome. Front Immunol 2021; 11:602823. [PMID: 33584670 PMCID: PMC7876316 DOI: 10.3389/fimmu.2020.602823] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/10/2020] [Indexed: 01/14/2023] Open
Abstract
Primary Sjögren's syndrome (pSS) is a chronic autoimmune disease associated with damage to multiple organs and glands. The most common clinical manifestations are dry eyes, dry mouth, and enlarged salivary glands. Currently, CD4+ T lymphocytes are considered to be key factors in the immunopathogenesis of pSS, but various studies have shown that CD8+ T lymphocytes contribute to acinar injury in the exocrine glands. Therefore, in this review, we discussed the classification and features of CD8+ T lymphocytes, specifically describing the role of CD8+ T lymphocytes in disease pathophysiology. Furthermore, we presented treatment strategies targeting CD8+ T cells to capitalize on the pathogenic and regulatory potential of CD8+ T lymphocytes in SS to provide promising new strategies for this inflammatory disease.
Collapse
Affiliation(s)
- Huimin Zhou
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jun Yang
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Jie Tian
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
79
|
Freen-van Heeren JJ. Toll-like receptor-2/7-mediated T cell activation: An innate potential to augment CD8 + T cell cytokine production. Scand J Immunol 2021; 93:e13019. [PMID: 33377182 DOI: 10.1111/sji.13019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/10/2020] [Accepted: 12/26/2020] [Indexed: 12/11/2022]
Abstract
CD8+ T cells are critical to combat pathogens and eradicate malignantly transformed cells. To exert their effector function and kill target cells, T cells produce copious amounts of effector molecules, including the pro-inflammatory cytokines interferon γ, tumour necrosis factor α and interleukin 2. TCR triggering alone is sufficient to induce cytokine secretion by effector and memory CD8+ T cells. However, T cells can also be directly activated by pathogen-derived molecules, such as through the triggering of Toll-like receptors (TLRs). TLR-mediated pathogen sensing by T cells results in the production of only interferon γ. However, in particular when the antigen load on target cells is low, or when TCR affinity to the antigen is limited, antigen-experienced T cells can benefit from costimulatory signals. TLR stimulation can also function in a costimulatory fashion to enhance TCR triggering. Combined TCR and TLR triggering enhances the proliferation, memory formation and effector function of T cells, resulting in enhanced production of interferon γ, tumour necrosis factor α and interleukin 2. Therefore, TLR ligands or the exploitation of TLR signalling could provide novel opportunities for immunotherapy approaches. In fact, CD19 CAR T cells bearing an intracellular TLR2 costimulatory domain were recently employed to treat cancer patients in a clinical trial. Here, the current knowledge regarding TLR2/7 stimulation-induced cytokine production by T cells is reviewed. Specifically, the transcriptional and post-transcriptional pathways engaged upon TLR2/7 sensing and TLR2/7 signalling are discussed. Finally, the potential uses of TLRs to enhance the anti-tumor effector function of T cells are explored.
Collapse
|
80
|
Loucif H, Dagenais-Lussier X, Beji C, Cassin L, Jrade H, Tellitchenko R, Routy JP, Olagnier D, van Grevenynghe J. Lipophagy confers a key metabolic advantage that ensures protective CD8A T-cell responses against HIV-1. Autophagy 2021; 17:3408-3423. [PMID: 33459125 DOI: 10.1080/15548627.2021.1874134] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although macroautophagy/autophagy has been proposed as a critical defense mechanism against HIV-1 by targeting viral components for degradation, its contribution as a catabolic process in providing optimal anti-HIV-1 immunity has never been addressed. The failure to restore proper antiviral CD8A/CD8 T-cell immunity, especially against HIV-1, is still the major limitation of current antiretroviral therapies. Consequently, it is of clinical imperative to provide new strategies to enhance the function of HIV-1-specific CD8A T-cells in patients under antiretroviral treatments (ART). Here, we investigated whether targeting autophagy activity could be an optional solution to make this possible. Our data show that, after both polyclonal and HIV-1-specific activation, CD8A T-cells from ART displayed reduced autophagy-dependent degradation of lysosomal contents when compared to naturally HIV-1 protected elite controllers (EC). We further confirmed in EC, by using specific BECN1 gene silencing and lysosomal inhibitors, the critical role of active autophagy in superior CD8A T-cell protection against HIV-1. More importantly, we found that an IL21 treatment was effective in rescuing the antiviral CD8A T-cell immunity from ART in an autophagy-dependent manner. Finally, we established that IL21-dependent rescue occurred due to the enhanced degradation of endogenous lipids via autophagy, referred to as lipophagy, which fueled the cellular rates of mitochondrial beta-oxidation. In summary, our data show that autophagy/lipophagy can be considered as a therapeutic tool to elicit functional antiviral CD8 T-cell responses. Our results also provide additional insights toward the development of improved T-cell-based prevention and cure strategies against HIV-1.Abbreviations: ART: patients under antiretroviral therapy; BaF: bafilomycin A1; BECN1: beclin 1; CEF: cytomegalo-, Epstein-Barr- and flu-virus peptide pool; Chloro.: chloroquine; EC: elite controllers; FAO: fatty acid beta-oxidation; HIVneg: HIV-1-uninfected control donors; IFNG/IFN-γ: interferon gamma; IL21: interleukin 21; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; PBMC: peripheral blood mononuclear cells; SQSTM1: sequestosome 1; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Hamza Loucif
- Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie
| | - Xavier Dagenais-Lussier
- Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie
| | - Cherifa Beji
- Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie
| | - Léna Cassin
- Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie
| | - Hani Jrade
- The Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Roman Tellitchenko
- Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie
| | - Jean-Pierre Routy
- Chronic Viral Illness Service and Division of Hematology, McGill University Health Centre, Glen Site, Montreal, QC, Canada
| | - David Olagnier
- Department of Biomedicine, Research Center for Innate Immunology, Aarhus University, Aarhus C, Denmark
| | - Julien van Grevenynghe
- Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie
| |
Collapse
|
81
|
Kervevan J, Chakrabarti LA. Role of CD4+ T Cells in the Control of Viral Infections: Recent Advances and Open Questions. Int J Mol Sci 2021; 22:E523. [PMID: 33430234 PMCID: PMC7825705 DOI: 10.3390/ijms22020523] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 12/26/2022] Open
Abstract
CD4+ T cells orchestrate adaptive immune responses through their capacity to recruit and provide help to multiple immune effectors, in addition to exerting direct effector functions. CD4+ T cells are increasingly recognized as playing an essential role in the control of chronic viral infections. In this review, we present recent advances in understanding the nature of CD4+ T cell help provided to antiviral effectors. Drawing from our studies of natural human immunodeficiency virus (HIV) control, we then focus on the role of high-affinity T cell receptor (TCR) clonotypes in mediating antiviral CD4+ T cell responses. Last, we discuss the role of TCR affinity in determining CD4+ T cell differentiation, reviewing the at times divergent studies associating TCR signal strength to the choice of a T helper 1 (Th1) or a T follicular helper (Tfh) cell fate.
Collapse
Affiliation(s)
- Jérôme Kervevan
- Control of Chronic Viral Infections Group (CIVIC), Virus and Immunity Unit, Institut Pasteur, 75724 Paris, France;
- CNRS UMR, 3569 Paris, France
| | - Lisa A. Chakrabarti
- Control of Chronic Viral Infections Group (CIVIC), Virus and Immunity Unit, Institut Pasteur, 75724 Paris, France;
- CNRS UMR, 3569 Paris, France
| |
Collapse
|
82
|
Abstract
Over the past four decades, research on the natural history of HIV infection has described how HIV wreaks havoc on human immunity and causes AIDS. HIV host genomic research, which aims to understand how human genetic variation affects our response to HIV infection, has progressed from early candidate gene studies to recent multi-omic efforts, benefiting from spectacular advances in sequencing technology and data science. In addition to invading cells and co-opting the host machinery for replication, HIV also stably integrates into our own genome. The study of the complex interactions between the human and retroviral genomes has improved our understanding of pathogenic mechanisms and suggested novel preventive and therapeutic approaches against HIV infection.
Collapse
Affiliation(s)
- Paul J. McLaren
- grid.415368.d0000 0001 0805 4386National HIV and Retrovirology Laboratory at the JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB Canada ,grid.21613.370000 0004 1936 9609Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB Canada
| | - Jacques Fellay
- grid.5333.60000000121839049School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland ,grid.419765.80000 0001 2223 3006Swiss Institute of Bioinformatics, Lausanne, Switzerland ,grid.8515.90000 0001 0423 4662Precision Medicine Unit, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
83
|
Buckland MS, Galloway JB, Fhogartaigh CN, Meredith L, Provine NM, Bloor S, Ogbe A, Zelek WM, Smielewska A, Yakovleva A, Mann T, Bergamaschi L, Turner L, Mescia F, Toonen EJM, Hackstein CP, Akther HD, Vieira VA, Ceron-Gutierrez L, Periselneris J, Kiani-Alikhan S, Grigoriadou S, Vaghela D, Lear SE, Török ME, Hamilton WL, Stockton J, Quick J, Nelson P, Hunter M, Coulter TI, Devlin L, Bradley JR, Smith KGC, Ouwehand WH, Estcourt L, Harvala H, Roberts DJ, Wilkinson IB, Screaton N, Loman N, Doffinger R, Lyons PA, Morgan BP, Goodfellow IG, Klenerman P, Lehner PJ, Matheson NJ, Thaventhiran JED. Treatment of COVID-19 with remdesivir in the absence of humoral immunity: a case report. Nat Commun 2020; 11:6385. [PMID: 33318491 PMCID: PMC7736571 DOI: 10.1038/s41467-020-19761-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
The response to the coronavirus disease 2019 (COVID-19) pandemic has been hampered by lack of an effective severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antiviral therapy. Here we report the use of remdesivir in a patient with COVID-19 and the prototypic genetic antibody deficiency X-linked agammaglobulinaemia (XLA). Despite evidence of complement activation and a robust T cell response, the patient developed persistent SARS-CoV-2 pneumonitis, without progressing to multi-organ involvement. This unusual clinical course is consistent with a contribution of antibodies to both viral clearance and progression to severe disease. In the absence of these confounders, we take an experimental medicine approach to examine the in vivo utility of remdesivir. Over two independent courses of treatment, we observe a temporally correlated clinical and virological response, leading to clinical resolution and viral clearance, with no evidence of acquired drug resistance. We therefore provide evidence for the antiviral efficacy of remdesivir in vivo, and its potential benefit in selected patients.
Collapse
Affiliation(s)
- Matthew S Buckland
- Department of Clinical Immunology, Barts Health, London, UK.
- UCL GOSH Institute of Child Health Division of Infection and Immunity, Section of Cellular and Molecular Immunology, London, UK.
| | - James B Galloway
- Centre for Rheumatic Diseases, King's College London, London, UK
| | | | - Luke Meredith
- Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Nicholas M Provine
- Peter Medawar Building for Pathogen Research, South Parks Rd, Oxford, OX1 3SY, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Stuart Bloor
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Ane Ogbe
- Peter Medawar Building for Pathogen Research, South Parks Rd, Oxford, OX1 3SY, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Wioleta M Zelek
- Systems Immunity Institute and Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Anna Smielewska
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrookes Hospital, Cambridge, UK
- PHE - Public Health England Laboratory, Cambridge. Box 236, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, UK
| | - Anna Yakovleva
- Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Tiffeney Mann
- Medical Research Council Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Lorinda Turner
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Frederica Mescia
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Erik J M Toonen
- R&D Department, Hycult Biotechnology, Frontstraat 2A, 5405 PB, Uden, The Netherlands
| | - Carl-Philipp Hackstein
- Peter Medawar Building for Pathogen Research, South Parks Rd, Oxford, OX1 3SY, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Hossain Delowar Akther
- Peter Medawar Building for Pathogen Research, South Parks Rd, Oxford, OX1 3SY, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Vinicius Adriano Vieira
- Peter Medawar Building for Pathogen Research, South Parks Rd, Oxford, OX1 3SY, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | | | - Jimstan Periselneris
- Respiratory Department, King's College Hospital NHS Foundation Trust, UK. Department of Clinical Virology, Addenbrookes, UK
| | | | | | - Devan Vaghela
- Department of Infectious Diseases, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Sara E Lear
- Department of Immunology, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - M Estée Török
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Department of Microbiology, Cambridge, UK
| | - William L Hamilton
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Joanne Stockton
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Josh Quick
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Peter Nelson
- Belfast Health and Social Care Trust, Belfast, Northern Ireland, UK
| | - Michael Hunter
- Belfast Health and Social Care Trust, Belfast, Northern Ireland, UK
| | - Tanya I Coulter
- Belfast Health and Social Care Trust, Belfast, Northern Ireland, UK
- Regional Immunology Service, Belfast Health and Social Care Trust, Belfast, Northern Ireland, UK
| | - Lisa Devlin
- Belfast Health and Social Care Trust, Belfast, Northern Ireland, UK
- Regional Immunology Service, Belfast Health and Social Care Trust, Belfast, Northern Ireland, UK
| | - John R Bradley
- NIHR BioResource and NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | | | | | - David J Roberts
- NHS Blood and Transplant, Oxford, UK
- Radcliffe Department of Medicine and BRC Haematology Theme, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Ian B Wilkinson
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Nicholas Loman
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Rainer Doffinger
- Respiratory Department, King's College Hospital NHS Foundation Trust, UK. Department of Clinical Virology, Addenbrookes, UK
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - B Paul Morgan
- Systems Immunity Institute and Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Ian G Goodfellow
- Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, South Parks Rd, Oxford, OX1 3SY, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Paul J Lehner
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Department of Infectious Diseases, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Nicholas J Matheson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Infectious Diseases, Cambridge University Hospitals NHS Trust, Cambridge, UK.
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK.
| | - James E D Thaventhiran
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK.
- Medical Research Council Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QW, UK.
- Cancer Research UK Cambridge Institute, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
84
|
Inflammation, HIV, and Immune Quiescence: Leveraging on Immunomodulatory Products to Reduce HIV Susceptibility. AIDS Res Treat 2020; 2020:8672850. [PMID: 33178456 PMCID: PMC7609152 DOI: 10.1155/2020/8672850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/23/2020] [Accepted: 10/15/2020] [Indexed: 12/29/2022] Open
Abstract
The relationship between inflammation and HIV has been a focus of research over the last decade. In HIV-infected individuals, increased HIV-associated immune activation significantly correlated to disease progression. While genital inflammation (GI) has been shown to significantly increase the risk of HIV acquisition and transmission, immune correlates for reduced risk remain limited. In certain HIV-exposed seronegative individuals, an immune quiescent phenotype characterized reduced risk. Immune quiescence is defined by specific, targeted, highly regulated immune responses that hinder overt inflammation or immune activation. Targeted management of inflammation, therefore, is a plausible strategy to mitigate HIV risk and slow disease progression. Nonsteroidal anti-inflammatory drugs (NSAIDs) such as hydroxychloroquine and aspirin have shown encouraging preliminary results in low-risk women by reducing systemic and genital immune activation. A topical NSAID, containing ibuprofen, is effective in treating vulvovaginal inflammation. Additionally, the glucocorticoids (GCs), prednisolone, and dexamethasone are used to treat HIV-associated immune activation. Collectively, these data inform on immune-modulating drugs to reduce HIV risk. However, the prolonged use of these pharmaceutical drugs is associated with adverse effects, both systemically and to a lesser extent topically. Natural products with their reduced side effects coupled with anti-inflammatory properties render them viable options. Lactic acid (LA) has immunomodulatory properties. LA regulates the genital microbiome by facilitating the growth of Lactobacillus species, while simultaneously limiting bacterial species that cause microbial dysbiosis and GI. Glycerol monolaurate, besides being anti-inflammatory, also inhibited SIV infections in rhesus macaques. The proposed pharmaceutical and natural products could be used in combination with either antiretrovirals for treatment or preexposure prophylaxis for HIV prevention. This review provides a summary on the associations between inflammation, HIV risk, and disease progression. Furthermore, we use the knowledge from immune quiescence to exploit the use of pharmaceutical and natural products as strategic interventions to manage inflammation, toward mitigating HIV infections.
Collapse
|
85
|
Bricker KM, Obregon-Perko V, Uddin F, Williams B, Uffman EA, Garrido C, Fouda GG, Geleziunas R, Robb M, Michael N, Barouch DH, Chahroudi A. Therapeutic vaccination of SIV-infected, ART-treated infant rhesus macaques using Ad48/MVA in combination with TLR-7 stimulation. PLoS Pathog 2020; 16:e1008954. [PMID: 33104758 PMCID: PMC7644092 DOI: 10.1371/journal.ppat.1008954] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/05/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
Globally, 1.8 million children are living with HIV-1. While antiretroviral therapy (ART) has improved disease outcomes, it does not eliminate the latent HIV-1 reservoir. Interventions to delay or prevent viral rebound in the absence of ART would be highly beneficial for HIV-1-infected children who now must remain on daily ART throughout their lifespan. Here, we evaluated therapeutic Ad48-SIV prime, MVA-SIV boost immunization in combination with the TLR-7 agonist GS-986 in rhesus macaque (RM) infants orally infected with SIVmac251 at 4 weeks of age and treated with a triple ART regimen beginning 4 weeks after infection. We hypothesized immunization would enhance SIV-specific T cell responses during ART-mediated suppression of viremia. Compared to controls, vaccinated infants had greater magnitude SIV-specific T cell responses (mean of 3475 vs 69 IFN-γ spot forming cells (SFC) per 106 PBMCs, respectively, P = 0.01) with enhanced breadth of epitope recognition and increased CD8+ and CD4+ T cell polyfunctionality (P = 0.004 and P = 0.005, respectively). Additionally, SIV-specific gp120 antibodies against challenge and vaccine virus strains were significantly elevated following MVA boost (P = 0.02 and P < 0.001, respectively). GS-986 led to expected immune stimulation demonstrated by activation of monocytes and T cells 24 hours post-dose. Despite the vaccine-induced immune responses, levels of SIV DNA in peripheral and lymph node CD4+ T cells were not significantly different from controls and a similar time to viral rebound and viral load set point were observed following ART interruption in both groups. We demonstrate infant RMs mount a robust immunological response to this immunization, but vaccination alone was not sufficient to impact viral reservoir size or modulate rebound dynamics following ART release. Our findings hold promise for therapeutic vaccination as a part of a combination cure approach in children and highlight the importance of a pediatric model to evaluate HIV-1 cure interventions in this unique setting of immune development. While antiretroviral therapy (ART) has improved disease outcome and reduced HIV-1 transmission, it is not a cure, as interruption of ART results in rapid viral rebound due to the persistent latent reservoir. Interventions to induce HIV-1 remission in the absence of ART would be highly beneficial to children living with HIV-1, sparing them from the associated adherence requirements, side effects, and cost of ART. Here, we used our previously established pediatric model of oral SIV infection and ART suppression of viremia in infant rhesus macaques (RMs) to evaluate the safety and efficacy of an Ad48-SIV prime, MVA-SIV boost therapeutic vaccine approach plus TLR-7 stimulation. Our study demonstrates this vaccination strategy is immunogenic in infants; however, unlike previously reported results in adult RMs using a similar approach, vaccination did not result in a difference in the level of CD4+ T cell-associated SIV DNA or viral rebound dynamics after ART interruption when compared to control infant RMs. These results highlight the importance of pre-clinical studies using pediatric models and indicate potential HIV-1 cure strategies may differentially impact adults and children.
Collapse
Affiliation(s)
- Katherine M. Bricker
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Veronica Obregon-Perko
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Ferzan Uddin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Brianna Williams
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Emilie A. Uffman
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States of America
| | - Carolina Garrido
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States of America
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States of America
- Departments of Molecular Genetics and Microbiology and Pediatrics, Duke University School of Medicine, Durham, NC, United States of America
| | - Romas Geleziunas
- Gilead Sciences, Inc., Foster City, CA, United States of America
| | - Merlin Robb
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Nelson Michael
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States of America
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
86
|
Ward AR, Mota TM, Jones RB. Immunological approaches to HIV cure. Semin Immunol 2020; 51:101412. [PMID: 32981836 DOI: 10.1016/j.smim.2020.101412] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
Combination antiretroviral therapy (ART) to treat human immunodeficiency virus (HIV) infection has proven remarkably successful - for those who can access and afford it - yet HIV infection persists indefinitely in a reservoir of cells, despite effective ART and despite host antiviral immune responses. An HIV cure is therefore the next aspirational goal and challenge, though approaches differ in their objectives - with 'functional cures' aiming for durable viral control in the absence of ART, and 'sterilizing cures' aiming for the more difficult to realize objective of complete viral eradication. Mechanisms of HIV persistence, including viral latency, anatomical sequestration, suboptimal immune functioning, reservoir replenishment, target cell-intrinsic immune resistance, and, potentially, target cell distraction of immune effectors, likely need to be overcome in order to achieve a cure. A small fraction of people living with HIV (PLWH) naturally control infection via immune-mediated mechanisms, however, providing both sound rationale and optimism that an immunological approach to cure is possible. Herein we review up to date knowledge and emerging evidence on: the mechanisms contributing to HIV persistence, as well as potential strategies to overcome these barriers; promising immunological approaches to achieve viral control and elimination of reservoir-harboring cells, including harnessing adaptive immune responses to HIV and engineered therapies, as well as enhancers of their functions and of complementary innate immune functioning; and combination strategies that are most likely to succeed. Ultimately, a cure must be safe, effective, durable, and, eventually, scalable in order to be widely acceptable and available.
Collapse
Affiliation(s)
- Adam R Ward
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA; Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA; PhD Program in Epidemiology, The George Washington University, Washington, DC, USA
| | - Talia M Mota
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - R Brad Jones
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA; Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA.
| |
Collapse
|
87
|
Abstract
Despite the success of anti-retroviral therapy (ART) in transforming HIV into a manageable disease, it has become evident that long-term ART will not eliminate the HIV reservoir and cure the infection. Alternative strategies to eradicate HIV infection, or at least induce a state of viral control and drug-free remission are therefore needed. Therapeutic vaccination aims to induce or enhance immunity to alter the course of a disease. In this review we provide an overview of the current state of therapeutic HIV vaccine research and summarize the obstacles that the field faces while highlighting potential ways forward for a strategy to cure HIV infection.
Collapse
|
88
|
Assmus LM, Guan J, Wu T, Farenc C, Sng XYX, Zareie P, Nguyen A, Nguyen AT, Tscharke DC, Thomas PG, Rossjohn J, Gras S, Croft NP, Purcell AW, La Gruta NL. Overlapping Peptides Elicit Distinct CD8 + T Cell Responses following Influenza A Virus Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:1731-1742. [PMID: 32868409 DOI: 10.4049/jimmunol.2000689] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/03/2020] [Indexed: 12/28/2022]
Abstract
The presentation of pathogen-derived peptides on MHC class I molecules is essential for the initiation of adaptive CD8+ T cell immunity, which in turn is critical for effective control of many significant human infections. The identification of immunogenic pathogen-derived epitopes and a detailed understanding of how they are recognized by TCRs is essential for the design of effective T cell-based vaccines. In this study, we have characterized the T cell recognition and immune responses in mice to two naturally presented influenza A virus-derived peptides previously identified from virally infected cells via mass spectrometry. These neuraminidase-derived peptides, NA181-190 (SGPDNGAVAV) and NA181-191 (SGPDNGAVAVL), are completely overlapping with the exception of a 1 aa extension at the C terminus of the longer peptide. This minor peptidic difference results in the induction of two completely independent and non-cross-reactive T cell populations that show distinct functional characteristics after influenza A virus infection of B6 mice. We show that the unique TCR reactivity to the overlapping peptides is present in the naive repertoire prior to immune expansion in B6 mice. Moreover, we provide a structural explanation underlying the distinct CD8+ T cell reactivities, which reinforces the concept that peptide length is a key determinant of Ag specificity in CD8+ T cell responses.
Collapse
Affiliation(s)
- Lisa M Assmus
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.,Institute of Experimental Immunology, University Hospital Bonn, 53105 Bonn, Germany
| | - Jing Guan
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Ting Wu
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Carine Farenc
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Xavier Y X Sng
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Pirooz Zareie
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Angela Nguyen
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Andrea T Nguyen
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - David C Tscharke
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Jamie Rossjohn
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia; and.,Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom
| | - Stephanie Gras
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Nathan P Croft
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia;
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia;
| | - Nicole L La Gruta
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia;
| |
Collapse
|
89
|
Sant S, Quiñones-Parra SM, Koutsakos M, Grant EJ, Loudovaris T, Mannering SI, Crowe J, van de Sandt CE, Rimmelzwaan GF, Rossjohn J, Gras S, Loh L, Nguyen THO, Kedzierska K. HLA-B*27:05 alters immunodominance hierarchy of universal influenza-specific CD8+ T cells. PLoS Pathog 2020; 16:e1008714. [PMID: 32750095 PMCID: PMC7428290 DOI: 10.1371/journal.ppat.1008714] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/14/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022] Open
Abstract
Seasonal influenza virus infections cause 290,000–650,000 deaths annually and severe morbidity in 3–5 million people. CD8+ T-cell responses towards virus-derived peptide/human leukocyte antigen (HLA) complexes provide the broadest cross-reactive immunity against human influenza viruses. Several universally-conserved CD8+ T-cell specificities that elicit prominent responses against human influenza A viruses (IAVs) have been identified. These include HLA-A*02:01-M158-66 (A2/M158), HLA-A*03:01-NP265-273, HLA-B*08:01-NP225-233, HLA-B*18:01-NP219-226, HLA-B*27:05-NP383-391 and HLA-B*57:01-NP199-207. The immunodominance hierarchies across these universal CD8+ T-cell epitopes were however unknown. Here, we probed immunodominance status of influenza-specific universal CD8+ T-cells in HLA-I heterozygote individuals expressing two or more universal HLAs for IAV. We found that while CD8+ T-cell responses directed towards A2/M158 were generally immunodominant, A2/M158+CD8+ T-cells were markedly diminished (subdominant) in HLA-A*02:01/B*27:05-expressing donors following ex vivo and in vitro analyses. A2/M158+CD8+ T-cells in non-HLA-B*27:05 individuals were immunodominant, contained optimal public TRBV19/TRAV27 TCRαβ clonotypes and displayed highly polyfunctional and proliferative capacity, while A2/M158+CD8+ T cells in HLA-B*27:05-expressing donors were subdominant, with largely distinct TCRαβ clonotypes and consequently markedly reduced avidity, proliferative and polyfunctional efficacy. Our data illustrate altered immunodominance patterns and immunodomination within human influenza-specific CD8+ T-cells. Accordingly, our work highlights the importance of understanding immunodominance hierarchies within individual donors across a spectrum of prominent virus-specific CD8+ T-cell specificities prior to designing T cell-directed vaccines and immunotherapies, for influenza and other infectious diseases. Annual influenza infections cause significant morbidity and morbidity globally. Established T-cell immunity directed at conserved viral regions provides some protection against influenza viruses and promotes rapid recovery, leading to better clinical outcomes. Killer CD8+ T-cells recognising viral peptides in a context of HLA-I glycoproteins, provide the broadest ever reported immunity across distinct influenza strains and subtypes. We asked whether the expression of certain HLA-I alleles affects CD8+ T cells responses. Our study clearly illustrates altered immunodominance hierarchies and immunodomination within broadly-cross-reactive influenza-specific CD8+ T-cells in individuals expressing two or more universal HLA-I alleles, key for T cell-directed vaccines and immunotherapies.
Collapse
Affiliation(s)
- Sneha Sant
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Sergio M. Quiñones-Parra
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Emma J. Grant
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Thomas Loudovaris
- Immunology and Diabetes Unit, St Vincent’s Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Stuart I. Mannering
- Immunology and Diabetes Unit, St Vincent’s Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Jane Crowe
- Deepdene Surgery, Deepdene, Victoria, Australia
| | - Carolien E. van de Sandt
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Guus F. Rimmelzwaan
- National Influenza Center and Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jamie Rossjohn
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - Stephanie Gras
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | - Liyen Loh
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Thi H. O. Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
- * E-mail: (THON); (KK)
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
- * E-mail: (THON); (KK)
| |
Collapse
|
90
|
Weichselbaum L, Azouz A, Smolen KK, Das J, Splittgerber M, Lepida A, Moreno C, Schreiber J, Sersté T, Trepo E, Libert F, Gustot T, Goriely S. Epigenetic basis for monocyte dysfunction in patients with severe alcoholic hepatitis. J Hepatol 2020; 73:303-314. [PMID: 32145259 DOI: 10.1016/j.jhep.2020.02.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Severe forms of alcohol-related liver disease are associated with increased susceptibility to infections which are associated with poor prognosis. The cellular and molecular mechanisms responsible for this altered host defense are incompletely understood. METHODS We performed whole blood phenotypic analysis and ex vivo stimulation with various pathogen-associated molecular patterns (PAMPs). We included 34 patients with alcohol-related cirrhosis (18 of whom had biopsy-proven severe alcoholic hepatitis [sAH]), 12 healthy controls and 11 patients with chronic alcohol consumption without significant liver disease. We also evaluated the transcriptomic (RNA-seq) and chromatin accessibility (ATAC-seq) profiles of CD14+ monocytes from a subset of patients. RESULTS Circulating monocytes and conventional dendritic cells (DCs) from patients with sAH displayed complex alterations characterized by increased expression of both activating and inhibitory surface markers and an impaired pro-inflammatory response upon stimulation with PAMPs representative of gram-negative bacteria (lipopolysaccharide, Pam3CSK4) or fungal pathogens (Zymosan). Their decreased ability to produce more than 1 cytokine (polyfunctionality) upon PAMP stimulation correlated with the risk of developing infection at 28 days or mortality at 90 days. The presence of acute-on-chronic liver failure in patients with sAH did not significantly modify the immune profile of monocytes and DCs. Moreover, CD14+ monocytes of patients with sAH displayed altered transcriptional and epigenomic profiles characterized by downregulation of key innate immune and metabolic pathways and upregulation of important immunomodulatory factors. CONCLUSIONS In patients with sAH, the altered transcriptional program and functional properties of monocytes that contribute to patients' susceptibility to infection have strong epigenetic determinants. LAY SUMMARY Patients with severe alcoholic hepatitis are at increased risk of infections, which contribute to the poor prognosis associated with the disease. Herein, we show that epigenetic determinants underly the immune cell dysfunction and inappropriate responses to pathogens that are associated with severe alcoholic hepatitis.
Collapse
Affiliation(s)
- Laura Weichselbaum
- Université Libre de Bruxelles, Institute for Medical Immunology (IMI), Gosselies, Belgium; Université Libre de Bruxelles, Laboratory of Experimental Gastroenterology, Brussels, Belgium
| | - Abdulkader Azouz
- Université Libre de Bruxelles, Institute for Medical Immunology (IMI), Gosselies, Belgium
| | - Kinga K Smolen
- Université Libre de Bruxelles, Institute for Medical Immunology (IMI), Gosselies, Belgium
| | - Jishnu Das
- Ragon Institute of MGH, MIT and Harvard; Department of Biological Engineering, MIT; Center for Systems Immunology, Department of Immunology, University of Pittsburgh School of Medicine; Department of Computational and Systems Biology, University of Pittsburgh School of Medicine
| | - Marion Splittgerber
- Université Libre de Bruxelles, Institute for Medical Immunology (IMI), Gosselies, Belgium
| | - Antonia Lepida
- Department of Gastroenterology and Hepato-Pancreatology, C.U.B. Erasme Hospital, Brussels, Belgium
| | - Christophe Moreno
- Department of Gastroenterology and Hepato-Pancreatology, C.U.B. Erasme Hospital, Brussels, Belgium; Université Libre de Bruxelles, Laboratory of Experimental Gastroenterology, Brussels, Belgium
| | - Jonas Schreiber
- Department of Gastroenterology and Hepato-Pancreatology, C.U.B. Erasme Hospital, Brussels, Belgium
| | - Thomas Sersté
- Department of Gastroenterology and Hepato-Pancreatology, C.U.B. Erasme Hospital, Brussels, Belgium; Department of Hepato-gastroenterology, C.U.B. Saint Pierre, Brussels, Belgium
| | - Eric Trepo
- Department of Gastroenterology and Hepato-Pancreatology, C.U.B. Erasme Hospital, Brussels, Belgium; Université Libre de Bruxelles, Laboratory of Experimental Gastroenterology, Brussels, Belgium
| | - Frederick Libert
- Université Libre de Bruxelles, BRIGHTcore ULB-VUB and Institute of Interdisciplinary Research in Human and Molecular Biology (IRIBHM), Brussels, Belgium
| | - Thierry Gustot
- Department of Gastroenterology and Hepato-Pancreatology, C.U.B. Erasme Hospital, Brussels, Belgium; Université Libre de Bruxelles, Laboratory of Experimental Gastroenterology, Brussels, Belgium; Inserm Unité 1149, Centre de Recherche sur l'inflammation (CRI), Paris, France.
| | - Stanislas Goriely
- Université Libre de Bruxelles, Institute for Medical Immunology (IMI), Gosselies, Belgium.
| |
Collapse
|
91
|
Acevedo-Saenz L, Perdomo-Celis F, Montoya CJ, Velilla PA. Polyfunctional CD8+ T-Cell Response to Autologous Peptides from Protease and Reverse Transcriptase of HIV-1 Clade B. Curr HIV Res 2020; 17:350-359. [PMID: 31622220 DOI: 10.2174/1570162x17666191017105910] [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: 08/02/2019] [Revised: 09/28/2019] [Accepted: 10/04/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND The diversity of the HIV proteome influences the cellular response and development of an effective vaccine, particularly due to the generation of viral variants with mutations located within CD8+ T-cell epitopes. These mutations can affect the recognition of the epitopes, that may result in the selection of HIV variants with mutated epitopes (autologous epitopes) and different CD8+ T-cell functional profiles. OBJECTIVE To determine the phenotype and functionality of CD8+ T-cell from HIV-infected Colombian patients in response to autologous and consensus peptides derived from HIV-1 clade B protease and reverse transcriptase (RT). METHODS By flow cytometry, we compared the ex vivo CD8+ T-cell responses from HIV-infected patients to autologous and consensus peptides derived from HIV-1 clade B protease and RT, restricted by HLA-B*35, HLA-B*44 and HLA-B*51 alleles. RESULTS Although autologous peptides restricted by HLA-B*35 and HLA-B*44 did not show any differences compared with consensus peptides, we observed the induction of a higher polyfunctional profile of CD8+ T-cells by autologous peptides restricted by HLA-B*51, particularly by the production of interferon-γ and macrophage inflammatory protein-1β. The response by different memory CD8+ T-cell populations was comparable between autologous vs. consensus peptides. In addition, the magnitude of the polyfunctional response induced by the HLA-B*51-restricted QRPLVTIRI autologous epitope correlated with low viremia. CONCLUSION Autologous peptides should be considered for the evaluation of HIV-specific CD8+ Tcell responses and to reveal some relevant epitopes that could be useful for therapeutic strategies aiming to promote polyfunctional CD8+ T-cell responses in a specific population of HIV-infected patients.
Collapse
Affiliation(s)
- Liliana Acevedo-Saenz
- Grupo Inmunovirologia, Departamento de Microbiologia y Parasitologia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia.,Grupo de Investigación Enfermería-CES, Facultad de Enfermería, Universidad CES, Medellin, Colombia
| | - Federico Perdomo-Celis
- Grupo Inmunovirologia, Departamento de Microbiologia y Parasitologia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Carlos J Montoya
- Grupo Inmunovirologia, Departamento de Microbiologia y Parasitologia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Paula A Velilla
- Grupo Inmunovirologia, Departamento de Microbiologia y Parasitologia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| |
Collapse
|
92
|
Lima NS, Takata H, Huang SH, Haregot A, Mitchell J, Blackmore S, Garland A, Sy A, Cartwright P, Routy JP, Michael NL, Appay V, Jones RB, Trautmann L. CTL Clonotypes with Higher TCR Affinity Have Better Ability to Reduce the HIV Latent Reservoir. THE JOURNAL OF IMMUNOLOGY 2020; 205:699-707. [PMID: 32591402 DOI: 10.4049/jimmunol.1900811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 05/23/2020] [Indexed: 01/09/2023]
Abstract
The success of the shock and kill strategy for the HIV cure depends both on the reactivation of the latent reservoir and on the ability of the immune system to eliminate infected cells. As latency reversal alone has not shown any impact in the size of the latent reservoir, ensuring that effector CTLs are able to recognize and kill HIV-infected cells could contribute to reservoir reduction. In this study, we investigated which functional aspects of human CTLs are associated with a better capacity to kill HIV-infected CD4+ T cells. We isolated Gag- and Nef-specific CTL clones with different TCR sequences from the PBMC of donors in acute and chronic infection. High-affinity clonotypes that showed IFN-γ production preserved even when the CD8 coreceptor was blocked, and clones with high Ag sensitivity exhibited higher efficiency at reducing the latent reservoir. Although intrinsic cytotoxic capacity did not differ according to TCR affinity, clonotypes with high TCR affinity showed a better ability to kill HIV-infected CD4+ T cells obtained from in vivo-infected PBMC and subjected to viral reactivation. Strategies aiming to specifically boost and maintain long-living memory CTLs with high TCR affinity in vivo prior to latency-reversing treatment might improve the efficacy of the shock and kill approach to reduce the latent reservoir.
Collapse
Affiliation(s)
- Noemia S Lima
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817.,Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814
| | - Hiroshi Takata
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817.,Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
| | - Szu-Han Huang
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY 10021.,Department of Microbiology, Immunology, and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Alexander Haregot
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | - Julie Mitchell
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817.,Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
| | - Stephen Blackmore
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | - Ayanna Garland
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | - Aaron Sy
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | | | - Jean-Pierre Routy
- Division of Hematology and Chronic Viral Illness Service, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Nelson L Michael
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910
| | - Victor Appay
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Université, INSERM, Paris 75005, France; and.,International Research Center of Medical Sciences, Kumamoto University, Kumamoto 860-8555, Japan
| | - R Brad Jones
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY 10021.,Department of Microbiology, Immunology, and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Lydie Trautmann
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910; .,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817.,Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
| |
Collapse
|
93
|
Ruiz-Mateos E, Poveda E, Lederman MM. Antiretroviral Treatment for HIV Elite Controllers? Pathog Immun 2020; 5:121-133. [PMID: 32582872 PMCID: PMC7307444 DOI: 10.20411/pai.v5i1.364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022] Open
Abstract
In most HIV-infected persons, the natural history of untreated infection is one of sustained viremia, progressive CD4 T cell depletion with resultant morbidity and mortality. The advent of effective combination antiretroviral therapy (ART) that controls HIV replication has altered this landscape dramatically. Yet a rare population of HIV-infected persons-elite controllers (EC)-can control HIV replication such that plasma levels of virus are "undetectable" without ART. The EC phenotype is heterogeneous, with some subjects durably controlling the virus-persistent elite controllers-and some eventually losing viral control-transient elite controllers. Overall, EC tend to have robust HIV-specific T cell responses and in some cases, mainly in transient elite controllers, elevated activation and inflammation indices that diminish with ART suggesting that endogenous defenses against this persistent pathogen come at the cost of heightened activation/inflammation. A limited data set suggests that cardiovascular disease risk as well as the occur-rence of other morbid events may be greater in the overall EC population than in treated HIV infection. ART in EC decreases activation indices but does not appear to increase circulating CD4 T cell numbers nor do we know if it alters clinical outcomes. Thus, it is difficult to recommend or discourage a decision to start ART in the EC population but the authors lean toward treatment particularly in those EC whose activation indices are high and those who are progressively losing circulating CD4 T cell numbers. Biomarkers that can reliably predict loss of virologic control and immune failure are needed.
Collapse
Affiliation(s)
- Ezequiel Ruiz-Mateos
- Clinic Unit of Infectious Diseases; Microbiology and Preventive Medicine; Institute of Biomedicine of Seville; Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Eva Poveda
- Group of Virology and Pathogenesis; Galicia Sur Health Research Institute (IIS Galicia Sur)-Complexo Hospitalario Universitario de Vigo; SERGAS-UVigo; Vigo, Spain
| | - Michael M. Lederman
- Division of Infectious Diseases; Center for AIDS Research; Case Western Reserve University and University Hospitals; Cleveland Medical Center; Cleveland, Ohio
| |
Collapse
|
94
|
Imai N, Tawara I, Yamane M, Muraoka D, Shiku H, Ikeda H. CD4 + T cells support polyfunctionality of cytotoxic CD8 + T cells with memory potential in immunological control of tumor. Cancer Sci 2020; 111:1958-1968. [PMID: 32304127 PMCID: PMC7293103 DOI: 10.1111/cas.14420] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/25/2020] [Accepted: 04/13/2020] [Indexed: 12/24/2022] Open
Abstract
Polyfunctionality/multifunctionality of effector T cells at the single cell level has been shown as an important parameter to predict the quality of T cell response and immunological control of infectious disease and malignancy. However, the fate of polyfunctional CD8+ CTLs and the factors that control the polyfunctionality of T cells remain largely unknown. Here we show that the acquisition of polyfunctionality on the initial stimulation is a sensitive immune correlate of CTL survival and memory formation. CD8+ T cells with high polyfunctionality, assessed with γ‐interferon and tumor necrosis factor‐α production and surface mobilization of the degranulation marker CD107a, showed enhanced Bcl‐2 expression, low apoptosis, and increased CD127highKLRG1low memory precursor phenotype. Consistent with these observations, CD8+ T cells were found to acquire high frequency of cells with polyfunctionality when stimulated in conditions known to enhance memory formation, such as the presence of CD4+ T cells, interleukin (IL)‐2, or IL‐21. Utilizing T‐cell receptor (TCR) transgenic mouse‐derived CD8+ T cells that express a TCR specific for a tumor‐derived neoantigen, we showed that polyfunctional tumor‐specific CTLs generated in the presence of CD4+ T cells showed long persistence in vivo and induced enhanced tumor regression when adoptively transferred into mice with progressing tumor. Acquisition of polyfunctionality thus impacts CTL survival and memory formation associated with immunological control of tumor.
Collapse
Affiliation(s)
- Naoko Imai
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Makiko Yamane
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Daisuke Muraoka
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroshi Shiku
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hiroaki Ikeda
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
95
|
Sng XYX, Li J, Zareie P, Assmus LM, Lee JKC, Jones CM, Turner SJ, Daley SR, Quinn KM, La Gruta NL. The Impact of MHC Class I Dose on Development and Maintenance of the Polyclonal Naive CD8+ T Cell Repertoire. THE JOURNAL OF IMMUNOLOGY 2020; 204:3108-3116. [DOI: 10.4049/jimmunol.2000081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
|
96
|
Freen-van Heeren JJ, Popović B, Guislain A, Wolkers MC. Human T cells employ conserved AU-rich elements to fine-tune IFN-γ production. Eur J Immunol 2020; 50:949-958. [PMID: 32112565 PMCID: PMC7384093 DOI: 10.1002/eji.201948458] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/27/2020] [Accepted: 02/27/2020] [Indexed: 12/25/2022]
Abstract
Long‐lasting CD8+ T cell responses are critical in combatting infections and tumors. The pro‐inflammatory cytokine IFN‐γ is a key effector molecule herein. We recently showed that in murine T cells the production of IFN‐γ is tightly regulated through adenylate uridylate–rich elements (AREs) that are located in the 3′ untranslated region (UTR) of the Ifng mRNA molecule. Loss of AREs resulted in prolonged cytokine production in activated T cells and boosted anti‐tumoral T cell responses. Here, we investigated whether these findings can be translated to primary human T cells. Utilizing CRISPR‐Cas9 technology, we deleted the ARE region from the IFNG 3′ UTR in peripheral blood‐derived human T cells. Loss of AREs stabilized the IFNG mRNA in T cells and supported a higher proportion of IFN‐γ protein‐producing T cells. Importantly, combining MART‐1 T cell receptor engineering with ARE‐Del gene editing showed that this was also true for antigen‐specific activation of T cells. MART‐1‐specific ARE‐Del T cells showed higher percentages of IFN‐γ producing T cells in response to MART‐1 expressing tumor cells. Combined, our study reveals that ARE‐mediated posttranscriptional regulation is conserved between murine and human T cells. Furthermore, generating antigen‐specific ARE‐Del T cells is feasible, a feature that could potentially be used for therapeutical purposes.
Collapse
Affiliation(s)
- Julian J Freen-van Heeren
- Department of Hematopoiesis, Sanquin Research-Amsterdam UMC Landsteiner Laboratory, Amsterdam, The Netherlands.,Department of Hematopoiesis, Oncode Institute, Amsterdam, The Netherlands
| | - Branka Popović
- Department of Hematopoiesis, Sanquin Research-Amsterdam UMC Landsteiner Laboratory, Amsterdam, The Netherlands.,Department of Hematopoiesis, Oncode Institute, Amsterdam, The Netherlands
| | - Aurélie Guislain
- Department of Hematopoiesis, Sanquin Research-Amsterdam UMC Landsteiner Laboratory, Amsterdam, The Netherlands.,Department of Hematopoiesis, Oncode Institute, Amsterdam, The Netherlands
| | - Monika C Wolkers
- Department of Hematopoiesis, Sanquin Research-Amsterdam UMC Landsteiner Laboratory, Amsterdam, The Netherlands.,Department of Hematopoiesis, Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
97
|
Salerno F, Turner M, Wolkers MC. Dynamic Post-Transcriptional Events Governing CD8+ T Cell Homeostasis and Effector Function. Trends Immunol 2020; 41:240-254. [DOI: 10.1016/j.it.2020.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 12/31/2022]
|
98
|
Li SY, Zhang ZN, Jiang YJ, Fu YJ, Shang H. Transcriptional insights into the CD8 + T cell response in mono-HIV and HCV infection. J Transl Med 2020; 18:96. [PMID: 32093694 PMCID: PMC7038596 DOI: 10.1186/s12967-020-02252-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/31/2020] [Indexed: 12/04/2022] Open
Abstract
Background Disease progression in the absence of therapy varies significantly in mono-HIV and HCV infected individuals. Virus-specific CD8+ T cells play an important role in restricting lentiviral replication and determining the rate of disease progression during HIV and HCV mono- and co-infection. Thus, understanding the similarities in the characteristics of CD8+ T cells in mono-HIV and HCV infection at the transcriptomic level contributes to the development of antiviral therapy. In this study, a meta-analysis of CD8+ T cell gene expression profiles derived from mono-HIV and HCV infected individuals at different stages of disease progression, was conducted to understand the common changes experienced by CD8+ T cells. Methods Five microarray datasets, reporting CD8+ T cell mRNA expression of the mono-HIV and HCV infected patients, were retrieved from Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) were identified via integrative meta-analysis of expression data (INMEX) program. Network analysis methods were used to assess protein–protein interaction (PPI) networks, Gene Ontology (GO) terms and pathway enrichment for DEGs. MirDIP and miRDB online prediction tools were used to predict potential microRNAs (miRNAs) targeting hub genes. Results First, we identified 625 and 154 DEGs in the CD8+ T cells originating from mono-HIV and HCV chronic progressor patients, respectively, compared to healthy individuals. Among them, interferon-stimulated genes (ISGs) including ISG15, IFIT3, ILI44L, CXCL8, FPR1 and TLR2, were upregulated after mono-HIV and HCV infection. Pathway enrichment analysis of DEGs showed that the “cytokine–cytokine receptor interaction” and “NF-kappa B” signaling pathways were upregulated after mono-HIV and HCV infection. In addition, we identified 92 and 50 DEGs in the CD8+ T cells of HIV non-progressor and HCV resolver patients, respectively, compared with corresponding chronic progressors. We observed attenuated mitosis and reduced ISG expression in HIV non-progressors and HCV resolvers compared with the corresponding chronic progressors. Finally, we identified miRNA-143-3p, predicted to target both IFIT3 in HIV and STAT5A in HCV infection. Conclusions We identified DEGs and transcriptional patterns in mono-HIV and HCV infected individuals at different stages of disease progression and identified miRNA-143-3p with potential to intervene disease progression, which provides a new strategy for developing targeted therapies.
Collapse
Affiliation(s)
- Si-Yao Li
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Zi-Ning Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yong-Jun Jiang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Ya-Jing Fu
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China. .,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
99
|
Combination anti-HIV-1 antibody therapy is associated with increased virus-specific T cell immunity. Nat Med 2020; 26:222-227. [PMID: 32015556 PMCID: PMC7018622 DOI: 10.1038/s41591-019-0747-1] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
Abstract
Combination antiretroviral therapy (ART) is highly effective in controlling human immunodeficiency virus (HIV)-1 but requires lifelong medication due to the existence of a latent viral reservoir1,2. Potent broadly neutralizing antibodies (bNAbs) represent a potential alternative or adjuvant to ART. In addition to suppressing viremia, bNAbs may have T cell immunomodulatory effects as seen for other forms of immunotherapy3. However, this has not been established in individuals who are infected with HIV-1. Here, we document increased HIV-1 Gag-specific CD8+ T cell responses in the peripheral blood of all nine study participants who were infected with HIV-1 with suppressed blood viremia, while receiving bNAb therapy during ART interruption4. Increased CD4+ T cell responses were detected in eight individuals. The increased T cell responses were due both to newly detectable reactivity to HIV-1 Gag epitopes and the expansion of pre-existing measurable responses. These data demonstrate that bNAb therapy during ART interruption is associated with enhanced HIV-1-specific T cell responses. Whether these augmented T cell responses can contribute to bNAb-mediated viral control remains to be determined. T cell responses specific for HIV-1 Gag peptides increased in HIV-positive recipients of two broadly neutralizing antibodies with prolonged suppression of blood viremia during antiretroviral treatment interruption.
Collapse
|
100
|
Ambalathingal GR, Francis RS, Corvino D, Srihari S, Aftab BT, Smith C, Khanna R. Proteome-wide analysis of T-cell response to BK polyomavirus in healthy virus carriers and kidney transplant recipients reveals a unique transcriptional and functional profile. Clin Transl Immunology 2020; 9:e01102. [PMID: 31956413 PMCID: PMC6960379 DOI: 10.1002/cti2.1102] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/15/2019] [Accepted: 12/19/2019] [Indexed: 11/29/2022] Open
Abstract
Objectives Cellular immunity against BK polyomavirus (BKV)‐encoded antigens plays a crucial role in long‐term protection against virus‐associated pathogenesis in transplant recipients. However, in‐depth understanding on dynamics of these cellular immune responses is required to develop better immune monitoring and immunotherapeutic strategies. Methods Here, we have conducted a proteome‐wide analysis of BKV‐specific T‐cell responses in a cohort of 53 healthy individuals and 26 kidney transplant recipients to delineate the functional and transcriptional profile of these effector cells and compared these characteristics to T cells directed against cytomegalovirus, which is also known to cause significant morbidity in transplant recipients. Results Profiling of BKV‐specific CD4+ and CD8+ T cells revealed that kidney transplant recipients with high levels of circulating viraemia showed significantly reduced T‐cell reactivity against large T and/or small T antigens when compared to healthy donors. Interestingly, T cells specific for these antigens showed strong cross‐recognition to orthologous JC virus (JCV) peptides, including those exhibiting varying degrees of sequence identity. Ex vivo functional and phenotypic characterisation revealed that the majority of BKV‐specific T cells from renal transplant recipients expressed low levels of the key transcriptional regulators T‐bet and eomesodermin, which was coincident with undetectable expression of granzyme B and perforin. However, in vitro stimulation of T cells with BKV epitopes selectively enhanced the expression of T‐bet, granzyme B and cellular trafficking molecules (CCR4, CD49d and CD103) with minimal change in eomesodermin and perforin. Conclusions These observations provide an important platform for the future development of immune monitoring and adoptive T‐cell therapy strategies for BKV‐associated diseases in transplant recipients, which may also be exploited for similar therapeutic value in JCV‐associated clinical complications.
Collapse
Affiliation(s)
- George R Ambalathingal
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development Tumour Immunology Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia
| | - Ross S Francis
- Department of Nephrology Princess Alexandra Hospital Woolloongabba QLD Australia.,School of Medicine University of Queensland Brisbane QLD Australia
| | - Dillon Corvino
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development Tumour Immunology Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia.,School of Medicine University of Queensland Brisbane QLD Australia
| | - Sriganesh Srihari
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development Tumour Immunology Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia
| | - Blake T Aftab
- Department of Preclinical and Translational Sciences Atara Biotherapeutics Los Angeles CA USA
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development Tumour Immunology Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development Tumour Immunology Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia.,School of Medicine University of Queensland Brisbane QLD Australia
| |
Collapse
|