1
|
Sohai DK, Keller MD, Hanley PJ, Hoq F, Kukadiya D, Datar A, Reynolds E, Copertino DC, Lazarski C, McCann CD, Tanna J, Shibli A, Lang H, Zhang A, Chansky PA, Motta C, Huynh TT, Dwyer B, Wilson A, Lynch R, Mota TM, Conce Alberto WD, Brumme ZL, Kinloch NN, Cruz CRY, MacLaren Ehui L, Henn S, Brad Jones R, Bollard CM. Autologous HIV-specific T cell therapy targeting conserved epitopes is well-tolerated in six adults with HIV: an open-label, single-arm phase 1 study. Nat Commun 2025; 16:4510. [PMID: 40374689 PMCID: PMC12081906 DOI: 10.1038/s41467-025-59810-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 05/06/2025] [Indexed: 05/17/2025] Open
Abstract
Novel cellular therapies may enable HIV control or cure. HIV-specific T cells targeting conserved immunogenic protein regions of HIV Gag/Pol and the entirety of HIV Nef, termed HST-NEETs, eliminate HIV infected cells in vitro. Here we enroll seven participants in an open-label, single-arm phase 1 study (NCT03485963) to evaluate the safety (primary endpoint) of two autologous administrations of HST-NEET products without prescribed lymphodepletion. Adults with well-controlled HIV on anti-retroviral therapy are eligible. Six participants completed safety monitoring. No serious product-related toxicities are observed. Secondary endpoints are to assess expansion and persistence of HIV-reactive T cell clones, and changes to the HIV reservoir for each infused participant. HIV-specific T cell and HIV anti-Env antibody responses increase in two participants after infusion two. A trend towards decreasing levels of intact proviruses is observed in 2 participants. Three participants show persistence of HIV-reactive, product-associated T cell clones for ≥40 weeks post infusions. HST-NEETs infusions are well-tolerated. Future trials are needed to evaluate the efficacy of HST-NEETs in this population.
Collapse
Affiliation(s)
- Danielle K Sohai
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Michael D Keller
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Fahmida Hoq
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Divyesh Kukadiya
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Anushree Datar
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Emily Reynolds
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Dennis C Copertino
- Infectious Diseases Division, Department of Medicine, Weill-Cornell Medicine, New York, NY, USA
| | - Christopher Lazarski
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Chase D McCann
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Jay Tanna
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Abeer Shibli
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Haili Lang
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Anqing Zhang
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Pamela A Chansky
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Cecilia Motta
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Tan T Huynh
- Infectious Diseases Division, Department of Medicine, Weill-Cornell Medicine, New York, NY, USA
| | - Bridget Dwyer
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Andrew Wilson
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Rebecca Lynch
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Talia M Mota
- Infectious Diseases Division, Department of Medicine, Weill-Cornell Medicine, New York, NY, USA
| | | | - Zabrina L Brumme
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada
| | - Natalie N Kinloch
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada
| | - Conrad Russell Y Cruz
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA
| | | | - Sarah Henn
- Whitman-Walker Health, Washington, DC, USA
| | - R Brad Jones
- Infectious Diseases Division, Department of Medicine, Weill-Cornell Medicine, New York, NY, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, and Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA.
- Integrated Biomedical Sciences, Department of Microbiology, Immunology, and Tropical Medicine, and Department of Pediatrics, The George Washington University, Washington, DC, USA.
| |
Collapse
|
2
|
Traut CC, Jones JL, Sanders RA, Clark LR, Hamill MM, Stavrakis G, Sop J, Beckey TP, Keller SC, Gilliams EA, Cochran WV, Laeyendecker O, Manabe YC, Mostafa HH, Thomas DL, Hansoti B, Gebo KA, Blankson JN. Orthopoxvirus-Specific T-Cell Responses in Convalescent Mpox Patients. J Infect Dis 2024; 229:54-58. [PMID: 37380166 PMCID: PMC10786252 DOI: 10.1093/infdis/jiad245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/07/2023] [Accepted: 06/24/2023] [Indexed: 06/30/2023] Open
Abstract
Orthopoxvirus-specific T-cell responses were analyzed in 10 patients who had recovered from Mpox including 7 people with human immunodeficiency virus (PWH). Eight participants had detectable virus-specific T-cell responses, including a PWH who was not on antiretroviral therapy and a PWH on immunosuppressive therapy. These 2 participants had robust polyfunctional CD4+ T-cell responses to peptides from the 121L vaccinia virus (VACV) protein. T-cells from 4 of 5 HLA-A2-positive participants targeted at least 1 previously described HLA-A2-restricted VACV epitope, including an epitope targeted in 2 participants. These results advance our understanding of immunity in convalescent Mpox patients.
Collapse
Affiliation(s)
- Caroline C Traut
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Joyce L Jones
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Renata A Sanders
- Department of Pediatrics, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Laura R Clark
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Matthew M Hamill
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Georgia Stavrakis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Joel Sop
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Tyler P Beckey
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Sara C Keller
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | | | - Willa V Cochran
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Oliver Laeyendecker
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Intramural Research Program, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Yukari C Manabe
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Heba H Mostafa
- Department of Pathology, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - David L Thomas
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Bhakti Hansoti
- Department of Emergency Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Kelly A Gebo
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Joel N Blankson
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Tang J, Fu M, Xu C, Xue B, Zhou A, Chen S, Zhao H, Zhou Y, Chen J, Yang Q, Chen X. Development of a novel virus-like particle-based vaccine for preventing tick-borne encephalitis virus infection. Virol Sin 2023; 38:767-777. [PMID: 37328107 PMCID: PMC10590693 DOI: 10.1016/j.virs.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/12/2023] [Indexed: 06/18/2023] Open
Abstract
Tick-borne encephalitis virus (TBEV) is an important tick-borne pathogen that poses as a serious public health concern. The coverage and immunogenicity of the currently available vaccines against TBEV are relatively low; therefore, it is crucial to develop novel and effective vaccines against TBEV. The present study describes a novel strategy for the assembly of virus-like particles (VLPs) by co-expressing the structural (core/prM/E) and non-structural (NS2B/NS3Pro) proteins of TBEV. The efficacy of the VLPs was subsequently evaluated in C57BL/6 mice, and the resultant IgG serum could neutralize both Far-Eastern and European subtypes of TBEV. These findings indicated that the VLP-based vaccine elicited the production of cross-subtype reactive antibodies. The VLPs provided protection to mice lacking the type I interferon receptor (IFNAR-/-) against lethal TBEV challenge, with undetectable viral load in brain and intestinal tissues. Furthermore, the group that received the VLP vaccine did not exhibit significant pathological changes and the inflammatory factors were significantly suppressed compared to the control group. Immunization with the VLP vaccine induced the production of multiple-cytokine-producing antiviral CD4+ T cells in vivo, including TNF-α+, IL-2+, and IFN-γ+ T cells. Altogether, the findings suggest that noninfectious VLPs can serve as a potentially safe and effective vaccine candidate against diverse subtypes of TBEV.
Collapse
Affiliation(s)
- Jielin Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Muqing Fu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Chonghui Xu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Bao Xue
- Guangzhou National Laboratory, Guangzhou, 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Anqi Zhou
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Sijie Chen
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - He Zhao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yuan Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jizheng Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qi Yang
- Guangzhou National Laboratory, Guangzhou, 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
4
|
Rakshit S, Babji S, Parthiban C, Madhavan R, Adiga V, J SE, Chetan Kumar N, Ahmed A, Shivalingaiah S, Shashikumar N, V M, Johnson AR, Ramesh N, B RG, Asokan M, Mayor S, Kang G, D'souza G, Dias M, Vyakarnam A. Polyfunctional CD4 T-cells correlating with neutralising antibody is a hallmark of COVISHIELD TM and COVAXIN ® induced immunity in COVID-19 exposed Indians. NPJ Vaccines 2023; 8:134. [PMID: 37709772 PMCID: PMC10502007 DOI: 10.1038/s41541-023-00731-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/31/2023] [Indexed: 09/16/2023] Open
Abstract
Detailed characterisation of immune responses induced by COVID-19 vaccines rolled out in India: COVISHIELDTM (CS) and COVAXIN® (CO) in a pre-exposed population is only recently being discovered. We addressed this issue in subjects who received their primary series of vaccination between November 2021 and January 2022. Both vaccines are capable of strongly boosting Wuhan Spike-specific neutralising antibody, polyfunctional Th1 cytokine producing CD4+ T-cells and single IFN-γ + CD8+ T-cells. Consistent with inherent differences in vaccine platform, the vector-based CS vaccine-induced immunity was of greater magnitude, breadth, targeting Delta and Omicron variants compared to the whole-virion inactivated vaccine CO, with CS vaccinees showing persistent CD8+ T-cells responses until 3 months post primary vaccination. This study provides detailed evidence on the magnitude and quality of CS and CO vaccine induced responses in subjects with pre-existing SARS-CoV-2 immunity in India, thereby mitigating vaccine hesitancy arguments in such a population, which remains a global health challenge.
Collapse
Affiliation(s)
- Srabanti Rakshit
- Division of Infectious Diseases, St. John's Research Institute, Bangalore, Karnataka, India
| | - Sudhir Babji
- The Wellcome Trust Research Laboratory, Christian Medical College, Vellore, Tamil Nadu, India
| | - Chaitra Parthiban
- Division of Infectious Diseases, St. John's Research Institute, Bangalore, Karnataka, India
| | - Ramya Madhavan
- The Wellcome Trust Research Laboratory, Christian Medical College, Vellore, Tamil Nadu, India
| | - Vasista Adiga
- Division of Infectious Diseases, St. John's Research Institute, Bangalore, Karnataka, India
- Department of Biotechnology, PES University, Bangalore, Karnataka, India
| | - Sharon Eveline J
- Division of Infectious Diseases, St. John's Research Institute, Bangalore, Karnataka, India
| | - Nirutha Chetan Kumar
- Division of Infectious Diseases, St. John's Research Institute, Bangalore, Karnataka, India
| | - Asma Ahmed
- Division of Infectious Diseases, St. John's Research Institute, Bangalore, Karnataka, India
| | | | - Nandini Shashikumar
- Division of Infectious Diseases, St. John's Research Institute, Bangalore, Karnataka, India
| | - Mamatha V
- St. John's Medical College, Bangalore, Karnataka, India
| | | | - Naveen Ramesh
- St. John's Medical College, Bangalore, Karnataka, India
| | | | | | - Satyajit Mayor
- National Centre for Biological Sciences, Bengaluru, Karnataka, India
| | - Gagandeep Kang
- The Wellcome Trust Research Laboratory, Christian Medical College, Vellore, Tamil Nadu, India
| | - George D'souza
- Department of Pulmonary Medicine, St. John's Medical College, Bangalore, Karnataka, India
| | - Mary Dias
- Division of Infectious Diseases, St. John's Research Institute, Bangalore, Karnataka, India
- St. John's Medical College, Bangalore, Karnataka, India
| | - Annapurna Vyakarnam
- Division of Infectious Diseases, St. John's Research Institute, Bangalore, Karnataka, India.
- Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Science & Medicine, King's College, London, UK.
| |
Collapse
|
5
|
Augusto DG, Murdolo LD, Chatzileontiadou DSM, Sabatino JJ, Yusufali T, Peyser ND, Butcher X, Kizer K, Guthrie K, Murray VW, Pae V, Sarvadhavabhatla S, Beltran F, Gill GS, Lynch KL, Yun C, Maguire CT, Peluso MJ, Hoh R, Henrich TJ, Deeks SG, Davidson M, Lu S, Goldberg SA, Kelly JD, Martin JN, Vierra-Green CA, Spellman SR, Langton DJ, Dewar-Oldis MJ, Smith C, Barnard PJ, Lee S, Marcus GM, Olgin JE, Pletcher MJ, Maiers M, Gras S, Hollenbach JA. A common allele of HLA is associated with asymptomatic SARS-CoV-2 infection. Nature 2023; 620:128-136. [PMID: 37468623 PMCID: PMC10396966 DOI: 10.1038/s41586-023-06331-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 06/15/2023] [Indexed: 07/21/2023]
Abstract
Studies have demonstrated that at least 20% of individuals infected with SARS-CoV-2 remain asymptomatic1-4. Although most global efforts have focused on severe illness in COVID-19, examining asymptomatic infection provides a unique opportunity to consider early immunological features that promote rapid viral clearance. Here, postulating that variation in the human leukocyte antigen (HLA) loci may underly processes mediating asymptomatic infection, we enrolled 29,947 individuals, for whom high-resolution HLA genotyping data were available, in a smartphone-based study designed to track COVID-19 symptoms and outcomes. Our discovery cohort (n = 1,428) comprised unvaccinated individuals who reported a positive test result for SARS-CoV-2. We tested for association of five HLA loci with disease course and identified a strong association between HLA-B*15:01 and asymptomatic infection, observed in two independent cohorts. Suggesting that this genetic association is due to pre-existing T cell immunity, we show that T cells from pre-pandemic samples from individuals carrying HLA-B*15:01 were reactive to the immunodominant SARS-CoV-2 S-derived peptide NQKLIANQF. The majority of the reactive T cells displayed a memory phenotype, were highly polyfunctional and were cross-reactive to a peptide derived from seasonal coronaviruses. The crystal structure of HLA-B*15:01-peptide complexes demonstrates that the peptides NQKLIANQF and NQKLIANAF (from OC43-CoV and HKU1-CoV) share a similar ability to be stabilized and presented by HLA-B*15:01. Finally, we show that the structural similarity of the peptides underpins T cell cross-reactivity of high-affinity public T cell receptors, providing the molecular basis for HLA-B*15:01-mediated pre-existing immunity.
Collapse
Affiliation(s)
- Danillo G Augusto
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
- Department of Biological Sciences, The University of North Carolina at Charlotte, Charlotte, NC, USA
- Programa de Pós-Graduação em Genética, Universidade Federal do Paraná, Curitiba, Brazil
| | - Lawton D Murdolo
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Demetra S M Chatzileontiadou
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Joseph J Sabatino
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Tasneem Yusufali
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Noah D Peyser
- Division of Cardiology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Xochitl Butcher
- Division of Cardiology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Kerry Kizer
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Karoline Guthrie
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Victoria W Murray
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Vivian Pae
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Sannidhi Sarvadhavabhatla
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Fiona Beltran
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Gurjot S Gill
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Kara L Lynch
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Cassandra Yun
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Colin T Maguire
- Clinical and Translational Science Institute, University of Utah, Salt Lake City, UT, USA
| | - Michael J Peluso
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Rebecca Hoh
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Timothy J Henrich
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Steven G Deeks
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Michelle Davidson
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Scott Lu
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Sarah A Goldberg
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - J Daniel Kelly
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- F.I. Proctor Foundation, University of California, San Francisco, CA, USA
| | - Jeffrey N Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Cynthia A Vierra-Green
- CIBMTR (Center for International Blood and Marrow Transplant Research), National Marrow Donor Program/Be The Match, Minneapolis, MN, USA
| | - Stephen R Spellman
- CIBMTR (Center for International Blood and Marrow Transplant Research), National Marrow Donor Program/Be The Match, Minneapolis, MN, USA
| | | | - Michael J Dewar-Oldis
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development Brisbane, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Peter J Barnard
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Sulggi Lee
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Gregory M Marcus
- Division of Cardiology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Jeffrey E Olgin
- Division of Cardiology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Mark J Pletcher
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- Division of General Internal Medicine, University of California, San Francisco, CA, USA
| | - Martin Maiers
- CIBMTR (Center for International Blood and Marrow Transplant Research), National Marrow Donor Program/Be The Match, Minneapolis, MN, USA
| | - Stephanie Gras
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jill A Hollenbach
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA.
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.
| |
Collapse
|
6
|
NKG2D signaling shifts the balance of CD8 T cells from single cytokine- to polycytokine-producing effector cells. Mol Immunol 2023; 155:1-6. [PMID: 36634520 PMCID: PMC9992161 DOI: 10.1016/j.molimm.2022.12.013] [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: 11/08/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023]
Abstract
CD8 T cells play a critical role in immunity against intracellular pathogens and cancer. A primary objective of T cell-based vaccine strategies is the induction of durable and effective immune responses. Achieving this goal involves more than simply boosting the numbers of responding T cells. Of particular interest is the induction of CD8 T cells with polycytokine capability, specifically with the ability of CD8 T cells to co-produce IFNγ, TNFα and IL-2. The presence of these polycytokine-producing CD8 T cells correlates strongly with protection against foreign pathogens and cancer. Therefore, approaches capable of inducing such polyfunctional responses are needed. NKG2D engagement on CD8 T cells has been shown to result in increased effector response. However, the manner in which NKG2D engagement results in improved CD8 T cell effector response is unclear. Here we demonstrate in vitro and in vivo that NKG2D engagement by its natural ligand, Rae-1ε, shifts the balance from single cytokine to polycytokine (IL-2, IFNγ, and TFNα) production. These data define a previously unrecognized process in which NKG2D costimulation on CD8 T cells results in improved effector responses.
Collapse
|
7
|
Spencer Clinton JL, Hoornweg TE, Tan J, Peng R, Schaftenaar W, Rutten VPMG, de Haan CAM, Ling PD. EEHV1A glycoprotein B subunit vaccine elicits humoral and cell-mediated immune responses in mice. Vaccine 2022; 40:5131-5140. [PMID: 35879117 DOI: 10.1016/j.vaccine.2022.07.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 10/16/2022]
Abstract
Asian elephants are an endangered species facing many threats, including severe hemorrhagic disease (HD) caused by the elephant endotheliotropic herpesvirus (EEHV). EEHV-HD is the leading cause of death in captive juvenile Asian elephants in North America and Europe, and also affects elephants in their natural range countries. Significant challenges exist for successful treatment of EEHV-HD, which include timely recognition of disease onset and limited availability of highly effective treatment options. To address this problem, our goal is to prevent lethal disease in young elephants by developing a vaccine that elicits robust and durable humoral and cell-mediated immunity against EEHV. EEHV glycoprotein B (gB) is a major target for cellular and humoral immunity in elephants previously exposed to EEHV. Therefore, we generated a vaccine containing recombinant EEHV1A gB together with a liposome formulated TLR-4 and saponin combination adjuvant (SLA-LSQ). CD-1 mice that received one or two vaccinations with the vaccine elicited significant anti-gB antibody and polyfunctional CD4+ and CD8+ T cell responses, while no adverse effects of vaccination were observed. Overall, our findings demonstrate that an adjuvanted gB protein subunit vaccine stimulates robust humoral and cell-mediated immune responses and supports its potential use in elephants.
Collapse
Affiliation(s)
- Jennifer L Spencer Clinton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Tabitha E Hoornweg
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands.
| | - Jie Tan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Rongsheng Peng
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Willem Schaftenaar
- Veterinary Advisor EAZA Elephant TAG, Rotterdam Zoo, Blijdorplaan 8, 3041 JG Rotterdam, The Netherlands.
| | - Victor P M G Rutten
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands; Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria, South Africa.
| | - Cornelis A M de Haan
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands.
| | - Paul D Ling
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Kumar Singh A, Padwal V, Palav H, Velhal S, Nagar V, Patil P, Patel V. Highly dampened HIV-specific cytolytic effector T cell responses define viremic non-progression. Immunobiology 2022; 227:152234. [PMID: 35671626 DOI: 10.1016/j.imbio.2022.152234] [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: 02/25/2022] [Accepted: 05/30/2022] [Indexed: 11/05/2022]
Abstract
This study reports on HIV-specific T cell responses in HIV-1 infected Viremic Non-Progressors (VNPs), a rare group of people living with HIV that exhibit asymptomatic infection over several years accompanied by stable CD4+ T cell counts in spite of ongoing viral replication. We attempted to identify key virus-specific functional attributes that could underlie the apparently paradoxical virus-host equilibrium observed in VNPs. Our results revealed modulation of HIV-specific CD4+ and CD8+ effector T cell responses in VNPs towards a dominant non-cytolytic profile with concomitantly diminished degranulation (CD107a+) ability. Further, the HIV specific CD8+ effector T cell response was primarily enriched for MIP-1β producing cells. As expected, concordant with better viral suppression, VCs exhibit a robust cytolytic T cell response. Interestingly, PuPs shared features common to both these responses but did not exhibit a CD4+ central memory IFN-γ producing Gag-specific response that was shared by both non-progressor (VC and VNP) groups, suggesting CD4 helper response is critical for non-progression. Our study also revealed that cytolytic response in VNPs is primarily limited to polyfunctional cells while both monofunctional and polyfunctional cells significantly contribute to cytolytic responses in VCs. To further understand mechanisms underlying the unique HIV-specific effector T cell response described here in VNPs we also evaluated and demonstrated a possible role for altered gut homing in these individuals. Our findings inform immunotherapeutic interventions to achieve functional cures in the context of ART resistance and serious non AIDS events.
Collapse
Affiliation(s)
- Amit Kumar Singh
- Viral Immunopathogenesis Laboratory, ICMR-National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India
| | - Varsha Padwal
- Viral Immunopathogenesis Laboratory, ICMR-National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India
| | - Harsha Palav
- Viral Immunopathogenesis Laboratory, ICMR-National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India
| | - Shilpa Velhal
- Viral Immunopathogenesis Laboratory, ICMR-National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India
| | - Vidya Nagar
- Department of Medicine, Grant Medical College & Sir J. J. Group of Hospitals, Mumbai, Maharashtra, India
| | - Priya Patil
- Department of Medicine, Grant Medical College & Sir J. J. Group of Hospitals, Mumbai, Maharashtra, India
| | - Vainav Patel
- Viral Immunopathogenesis Laboratory, ICMR-National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India.
| |
Collapse
|
9
|
Blunck BN, Angelo LS, Henke D, Avadhanula V, Cusick M, Ferlic-Stark L, Zechiedrich L, Gilbert BE, Piedra PA. Adult Memory T Cell Responses to the Respiratory Syncytial Virus Fusion Protein During a Single RSV Season (2018-2019). Front Immunol 2022; 13:823652. [PMID: 35422803 PMCID: PMC9002099 DOI: 10.3389/fimmu.2022.823652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Respiratory Syncytial Virus (RSV) is ubiquitous and re-infection with both subtypes (RSV/A and RSV/B) is common. The fusion (F) protein of RSV is antigenically conserved, induces neutralizing antibodies, and is a primary target of vaccine development. Insight into the breadth and durability of RSV-specific adaptive immune response, particularly to the F protein, may shed light on susceptibility to re-infection. We prospectively enrolled healthy adult subjects (n = 19) and collected serum and peripheral blood mononuclear cells (PBMCs) during the 2018–2019 RSV season. Previously, we described their RSV-specific antibody responses and identified three distinct antibody kinetic profiles associated with infection status: uninfected (n = 12), acutely infected (n = 4), and recently infected (n = 3). In this study, we measured the longevity of RSV-specific memory T cell responses to the F protein following natural RSV infection. We stimulated PBMCs with overlapping 15-mer peptide libraries spanning the F protein derived from either RSV/A or RSV/B and found that memory T cell responses mimic the antibody responses for all three groups. The uninfected group had stable, robust memory T cell responses and polyfunctionality. The acutely infected group had reduced polyfunctionality of memory T cell response at enrollment compared to the uninfected group, but these returned to comparable levels by end-of-season. The recently infected group, who were unable to maintain high levels of RSV-specific antibody following infection, similarly had decreased memory T cell responses and polyfunctionality during the RSV season. We observed subtype-specific differences in memory T cell responses and polyfunctionality, with RSV/A stimulating stronger memory T cell responses with higher polyfunctionality even though RSV/B was the dominant subtype in circulation. A subset of individuals demonstrated an overall deficiency in the generation of a durable RSV-specific adaptive immune response. Because memory T cell polyfunctionality may be associated with protection against re-infection, this latter group would likely be at greater risk of re-infection. Overall, these results expand our understanding of the longevity of the adaptive immune response to the RSV fusion protein and should be considered in future vaccine development efforts.
Collapse
Affiliation(s)
- Brittani N Blunck
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Laura S Angelo
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - David Henke
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Vasanthi Avadhanula
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Matthew Cusick
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Laura Ferlic-Stark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Lynn Zechiedrich
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, United States.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, United States
| | - Brian E Gilbert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
10
|
Dowell AC, Haigh TA, Ryan GB, Turner JE, Long HM, Taylor GS. Cytotoxic CD4+ T-cells specific for EBV capsid antigen BORF1 are maintained in long-term latently infected healthy donors. PLoS Pathog 2021; 17:e1010137. [PMID: 34882759 PMCID: PMC8691624 DOI: 10.1371/journal.ppat.1010137] [Citation(s) in RCA: 7] [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: 05/14/2021] [Revised: 12/21/2021] [Accepted: 11/22/2021] [Indexed: 12/15/2022] Open
Abstract
Epstein Barr Virus (EBV) infects more than 95% of the population whereupon it establishes a latent infection of B-cells that persists for life under immune control. Primary EBV infection can cause infectious mononucleosis (IM) and long-term viral carriage is associated with several malignancies and certain autoimmune diseases. Current efforts developing EBV prophylactic vaccination have focussed on neutralising antibodies. An alternative strategy, that could enhance the efficacy of such vaccines or be used alone, is to generate T-cell responses capable of recognising and eliminating newly EBV-infected cells before the virus initiates its growth transformation program. T-cell responses against the EBV structural proteins, brought into the newly infected cell by the incoming virion, are prime candidates for such responses. Here we show the structural EBV capsid proteins BcLF1, BDLF1 and BORF1 are frequent targets of T-cell responses in EBV infected people, identify new CD8+ and CD4+ T-cell epitopes and map their HLA restricting alleles. Using T-cell clones we demonstrate that CD4+ but not CD8+ T-cell clones specific for the capsid proteins can recognise newly EBV-infected B-cells and control B-cell outgrowth via cytotoxicity. Using MHC-II tetramers we show a CD4+ T-cell response to an epitope within the BORF1 capsid protein epitope is present during acute EBV infection and in long-term viral carriage. In common with other EBV-specific CD4+ T-cell responses the BORF1-specific CD4+ T-cells in IM patients expressed perforin and granzyme-B. Unexpectedly, perforin and granzyme-B expression was sustained over time even when the donor had entered the long-term infected state. These data further our understanding of EBV structural proteins as targets of T-cell responses and how CD4+ T-cell responses to EBV change from acute disease into convalescence. They also identify new targets for prophylactic EBV vaccine development. Epstein-Barr virus is a widespread herpesvirus carried by most individuals. Whilst infection is usually asymptomatic, development of a prophylactic vaccine against EBV is desirable because of the virus’s association with infectious mononucleosis in primary infection and several cancers and autoimmune diseases during long-term virus carriage. Identifying T-cell responses that can recognise newly infected B-cells at very early stages of infection may provide novel targets for T-cell vaccination. Here we characterise T-cell responses against three virus proteins, BcLF1, BDLF1 and BORF1 that, as structural proteins of the virus particle, are delivered into the cell by the infecting virus. We find that all three proteins are recognised by T-cells from infected individuals. Moreover, isolated structural antigen-specific CD4+ T-cells rapidly recognise newly infected B-cells and prevent their outgrowth in vitro. As reported for CD4+ T-cells against other EBV proteins, structural antigen-specific CD4+ T-cells induced by primary EBV infection have cytotoxic function. However, we also demonstrate that, unusually, this cytotoxic function is retained in memory T-cells present in long-term infected individuals. Structural antigens may therefore represent useful targets for prophylactic EBV vaccine development to induce CD4+ T-cells able to rapidly eliminate virus-infected cells.
Collapse
Affiliation(s)
- Alexander C. Dowell
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Tracey A. Haigh
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gordon B. Ryan
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - James E. Turner
- Department for Health, University of Bath, Claverton Down, Bath, United Kingdom
| | - Heather M. Long
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Graham S. Taylor
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- * E-mail:
| |
Collapse
|
11
|
Kumar NP, Padmapriyadarsini C, Rajamanickam A, Bhavani PK, Nancy A, Jayadeepa B, Selvaraj N, Asokan D, Renji RM, Venkataramani V, Tripathy S, Babu S. BCG vaccination induces enhanced frequencies of memory T cells and altered plasma levels of common γc cytokines in elderly individuals. PLoS One 2021; 16:e0258743. [PMID: 34758029 PMCID: PMC8580239 DOI: 10.1371/journal.pone.0258743] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/26/2021] [Indexed: 12/03/2022] Open
Abstract
BCG vaccination is known to induce innate immune memory, which confers protection against heterologous infections. However, the effect of BCG vaccination on the conventional adaptive immune cells subsets is not well characterized. We investigated the impact of BCG vaccination on the frequencies of T cell subsets and common gamma c (γc) cytokines in a group of healthy elderly individuals (age 60–80 years) at one month post vaccination as part of our clinical study to examine the effect of BCG on COVID-19. Our results demonstrate that BCG vaccination induced enhanced frequencies of central (p<0.0001) and effector memory (p<0.0001) CD4+ T cells and diminished frequencies of naïve (p<0.0001), transitional memory (p<0.0001), stem cell memory (p = 0.0001) CD4+ T cells and regulatory T cells. In addition, BCG vaccination induced enhanced frequencies of central (p = 0.0008), effector (p<0.0001) and terminal effector memory (p<0.0001) CD8+ T cells and diminished frequencies of naïve (p<0.0001), transitional memory (p<0.0001) and stem cell memory (p = 0.0034) CD8+T cells. BCG vaccination also induced enhanced plasma levels of IL-7 (p<0.0001) and IL-15 (p = 0.0020) but diminished levels of IL-2 (p = 0.0033) and IL-21 (p = 0.0020). Thus, BCG vaccination was associated with enhanced memory T cell subsets as well as memory enhancing γc cytokines in elderly individuals, suggesting its ability to induce non-specific adaptive immune responses.
Collapse
Affiliation(s)
- Nathella Pavan Kumar
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
- * E-mail: (NPK); (SB)
| | | | | | - Perumal Kannabiran Bhavani
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Arul Nancy
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
| | - Bharathi Jayadeepa
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Nandhini Selvaraj
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
| | - Dinesh Asokan
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | | | | | - Srikanth Tripathy
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
- Dr D Y Patil Medical College, Hospital and Research Centre, Pune, India
| | - Subash Babu
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
- * E-mail: (NPK); (SB)
| |
Collapse
|
12
|
Carre C, Wong G, Narang V, Tan C, Chong J, Chin HX, Xu W, Lu Y, Chua M, Poidinger M, Tambyah P, Nyunt M, Ng TP, Larocque D, Hessler C, Bosco N, Quemeneur L, Larbi A. Endoplasmic reticulum stress response and bile acid signatures associate with multi-strain seroresponsiveness during elderly influenza vaccination. iScience 2021; 24:102970. [PMID: 34471863 PMCID: PMC8387917 DOI: 10.1016/j.isci.2021.102970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 03/25/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022] Open
Abstract
The elderly are an important target for influenza vaccination, and the determination of factors that underlie immune responsiveness is clinically valuable. We evaluated the immune and metabolic profiles of 205 elderly Singaporeans administered with Vaxigrip. Despite high seroprotection rates, we observed heterogeneity in the response. We stratified the cohort into complete (CR) or incomplete responders (IR), where IR exhibited signs of accelerated T cell aging. We found a higher upregulation of genes associated with the B-cell endoplasmic-reticulum stress response in CR, where XBP-1 acts as a key upstream regulator. B-cells from IR were incapable of matching the level of XBP-1 upregulation observed in CR after inducing ER stress with tunicamycin in vitro. Metabolic signatures also distinguished CR and IR - as CR presented with a greater diversity of bile acids. Our findings suggest that the ER-stress pathway activation could improve influenza vaccination in the elderly.
Collapse
Affiliation(s)
| | - Glenn Wong
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Vipin Narang
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Crystal Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Joni Chong
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Hui Xian Chin
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Weili Xu
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Yanxia Lu
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Michelle Chua
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| | - Paul Tambyah
- Division of Infectious Diseases, National University Hospital, Singapore
| | - Ma Nyunt
- Division of Infectious Diseases, National University Hospital, Singapore
| | - Tze Pin Ng
- Division of Infectious Diseases, National University Hospital, Singapore
| | | | | | - Nabil Bosco
- Nestlé Research, Nestlé Institute of Health Sciences, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | | | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A∗STAR), Immunos, Singapore
| |
Collapse
|
13
|
Effects of therapeutic vaccination on the control of SIV in rhesus macaques with variable responsiveness to antiretroviral drugs. PLoS One 2021; 16:e0253265. [PMID: 34138927 PMCID: PMC8211199 DOI: 10.1371/journal.pone.0253265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023] Open
Abstract
A therapeutic vaccine that induces lasting control of HIV infection could eliminate the need for lifelong adherence to antiretroviral therapy. This study investigated a therapeutic DNA vaccine delivered with a single adjuvant or a novel combination of adjuvants to augment T cell immunity in the blood and gut-associated lymphoid tissue in SIV-infected rhesus macaques. Animals that received DNA vaccines expressing SIV proteins, combined with plasmids expressing adjuvants designed to increase peripheral and mucosal T cell responses, including the catalytic subunit of the E. coli heat-labile enterotoxin, IL-12, IL-33, retinaldehyde dehydrogenase 2, soluble PD-1 and soluble CD80, were compared to mock-vaccinated controls. Following treatment interruption, macaques exhibited variable levels of viral rebound, with four animals from the vaccinated groups and one animal from the control group controlling virus at median levels of 103 RNA copies/ml or lower (controllers) and nine animals, among all groups, exhibiting immediate viral rebound and median viral loads greater than 103 RNA copies/ml (non-controllers). Although there was no significant difference between the vaccinated and control groups in protection from viral rebound, the variable virological outcomes during treatment interruption enabled an examination of immune correlates of viral replication in controllers versus non-controllers regardless of vaccination status. Lower viral burden in controllers correlated with increased polyfunctional SIV-specific CD8+ T cells in mesenteric lymph nodes and blood prior to and during treatment interruption. Notably, higher frequencies of colonic CD4+ T cells and lower Th17/Treg ratios prior to infection in controllers correlated with improved responses to ART and control of viral rebound. These results indicate that mucosal immune responses, present prior to infection, can influence efficacy of antiretroviral therapy and the outcome of immunotherapeutic vaccination, suggesting that therapies capable of modulating host mucosal responses may be needed to achieve HIV cure.
Collapse
|
14
|
Isaguliants M, Krotova O, Petkov S, Jansons J, Bayurova E, Mezale D, Fridrihsone I, Kilpelainen A, Podschwadt P, Agapkina Y, Smirnova O, Kostic L, Saleem M, Latyshev O, Eliseeva O, Malkova A, Gorodnicheva T, Wahren B, Gordeychuk I, Starodubova E, Latanova A. Cellular Immune Response Induced by DNA Immunization of Mice with Drug Resistant Integrases of HIV-1 Clade A Offers Partial Protection against Growth and Metastatic Activity of Integrase-Expressing Adenocarcinoma Cells. Microorganisms 2021; 9:1219. [PMID: 34199989 PMCID: PMC8226624 DOI: 10.3390/microorganisms9061219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 02/07/2023] Open
Abstract
Therapeutic DNA-vaccination against drug-resistant HIV-1 may hinder emergence and spread of drug-resistant HIV-1, allowing for longer successful antiretroviral treatment (ART) up-to relief of ART. We designed DNA-vaccines against drug-resistant HIV-1 based on consensus clade A integrase (IN) resistant to raltegravir: IN_in_r1 (L74M/E92Q/V151I/N155H/G163R) or IN_in_r2 (E138K/G140S/Q148K) carrying D64V abrogating IN activity. INs, overexpressed in mammalian cells from synthetic genes, were assessed for stability, route of proteolytic degradation, and ability to induce oxidative stress. Both were found safe in immunotoxicity tests in mice, with no inherent carcinogenicity: their expression did not enhance tumorigenic or metastatic potential of adenocarcinoma 4T1 cells. DNA-immunization of mice with INs induced potent multicytokine T-cell response mainly against aa 209-239, and moderate IgG response cross-recognizing diverse IN variants. DNA-immunization with IN_in_r1 protected 60% of mice from challenge with 4Tlluc2 cells expressing non-mutated IN, while DNA-immunization with IN_in_r2 protected only 20% of mice, although tumor cells expressed IN matching the immunogen. Tumor size inversely correlated with IN-specific IFN-γ/IL-2 T-cell response. IN-expressing tumors displayed compromised metastatic activity restricted to lungs with reduced metastases size. Protective potential of IN immunogens relied on their immunogenicity for CD8+ T-cells, dependent on proteasomal processing and low level of oxidative stress.
Collapse
Affiliation(s)
- Maria Isaguliants
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
- Department of Research, Riga Stradins University, LV-1007 Riga, Latvia; (J.J.); (D.M.); (I.F.)
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia
| | - Olga Krotova
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Stefan Petkov
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Juris Jansons
- Department of Research, Riga Stradins University, LV-1007 Riga, Latvia; (J.J.); (D.M.); (I.F.)
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia
| | - Ekaterina Bayurova
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia
| | - Dzeina Mezale
- Department of Research, Riga Stradins University, LV-1007 Riga, Latvia; (J.J.); (D.M.); (I.F.)
| | - Ilze Fridrihsone
- Department of Research, Riga Stradins University, LV-1007 Riga, Latvia; (J.J.); (D.M.); (I.F.)
| | - Athina Kilpelainen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Philip Podschwadt
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Yulia Agapkina
- Department of Chemistry and Belozersky Institute of Physicochemical Biology, Moscow State University, 119991 Moscow, Russia;
| | - Olga Smirnova
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Linda Kostic
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Mina Saleem
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Oleg Latyshev
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
| | - Olesja Eliseeva
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
| | - Anastasia Malkova
- Institute of Medical Biological Research and Technologies, 143090 Krasnoznamensk, Russia;
| | | | - Britta Wahren
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Ilya Gordeychuk
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, 127994 Moscow, Russia
| | - Elizaveta Starodubova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasia Latanova
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
15
|
Li H, Omange RW, Liang B, Toledo N, Hai Y, Liu LR, Schalk D, Crecente-Campo J, Dacoba TG, Lambe AB, Lim SY, Li L, Kashem MA, Wan Y, Correia-Pinto JF, Seaman MS, Liu XQ, Balshaw RF, Li Q, Schultz-Darken N, Alonso MJ, Plummer FA, Whitney JB, Luo M. Vaccine targeting SIVmac251 protease cleavage sites protects macaques against vaginal infection. J Clin Invest 2021; 130:6429-6442. [PMID: 32853182 DOI: 10.1172/jci138728] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/20/2020] [Indexed: 01/03/2023] Open
Abstract
After over 3 decades of research, an effective anti-HIV vaccine remains elusive. The recently halted HVTN702 clinical trial not only further stresses the challenge to develop an effective HIV vaccine but also emphasizes that unconventional and novel vaccine strategies are urgently needed. Here, we report that a vaccine focusing the immune response on the sequences surrounding the 12 viral protease cleavage sites (PCSs) provided greater than 80% protection to Mauritian cynomolgus macaques against repeated intravaginal SIVmac251 challenges. The PCS-specific T cell responses correlated with vaccine efficacy. The PCS vaccine did not induce immune activation or inflammation known to be associated with increased susceptibility to HIV infection. Machine learning analyses revealed that the immune microenvironment generated by the PCS vaccine was predictive of vaccine efficacy. Our study demonstrates, for the first time to our knowledge, that a vaccine which targets only viral maturation, but lacks full-length Env and Gag immunogens, can prevent intravaginal infection in a stringent macaque/SIV challenge model. Targeting HIV maturation thus offers a potentially novel approach to developing an effective HIV vaccine.
Collapse
Affiliation(s)
- Hongzhao Li
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robert W Omange
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Binhua Liang
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nikki Toledo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yan Hai
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lewis R Liu
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Dane Schalk
- Scientific Protocol Implementation Unit, Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | - Jose Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Tamara G Dacoba
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | | | - So-Yon Lim
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Lin Li
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Mohammad Abul Kashem
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yanmin Wan
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Jorge F Correia-Pinto
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiao Qing Liu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robert F Balshaw
- Centre for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Nancy Schultz-Darken
- Scientific Protocol Implementation Unit, Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | - Maria J Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Francis A Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - James B Whitney
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Ma Luo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| |
Collapse
|
16
|
Hu X, Felber BK, Valentin A. Assessing Antigen-Specific Cellular Immune Responses upon HIV /SIV Plasmid DNA Vaccination in the Nonhuman Primate Model. Methods Mol Biol 2021; 2197:113-131. [PMID: 32827134 PMCID: PMC10802792 DOI: 10.1007/978-1-0716-0872-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Reliable detection and quantification of antigen-specific T cells are critical for assessing the immunogenicity of vaccine candidates. In this chapter, we describe the use of ELISpot and flow cytometry-based assays for efficient detection, mapping, and functional characterization of memory T lymphocytes in different tissues of rhesus macaques immunized with plasmid DNA. Flow cytometric assays provide a large amount of information, both phenotypic and functional, about individual cells, while the ELISpot is well suited for high throughput sample screening.
Collapse
Affiliation(s)
- Xintao Hu
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA.
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| |
Collapse
|
17
|
Abstract
As our understanding of mucosal immunity increases, it is becoming clear that the host response to HIV-1 is more complex and nuanced than originally believed. The mucosal landscape is populated with a variety of specialized cell types whose functions include combating infectious agents while preserving commensal microbiota, maintaining barrier integrity, and ensuring immune homeostasis. Advances in multiparameter flow cytometry, gene expression analysis and bioinformatics have allowed more detailed characterization of these cell types and their roles in host defense than was previously possible. This review provides an overview of existing literature on immunity to HIV-1 and SIVmac in mucosal tissues of the female reproductive tract and the gastrointestinal tract, focusing on major effector cell populations and briefly summarizing new information on tissue resident memory T cells, Treg, Th17, Th22 and innate lymphocytes (ILC), subsets that have been studied primarily in the gastrointestinal mucosa.
Collapse
Affiliation(s)
- Barbara L Shacklett
- Department of Medical Microbiology and Immunology.,Division of Infectious Diseases, Department of Medicine, School of Medicine, University of California, Davis, CA 95616
| |
Collapse
|
18
|
Munusamy Ponnan S, Pattabiram S, Thiruvengadam K, Goyal R, Singla N, Mukherjee J, Chatrath S, Bergin P, T. Kopycinski J, Gilmour J, Kumar S, Muthu M, Subramaniam S, Swaminathan S, Prasad Tripathy S, Luke HE. Induction and maintenance of bi-functional (IFN-γ + IL-2+ and IL-2+ TNF-α+) T cell responses by DNA prime MVA boosted subtype C prophylactic vaccine tested in a Phase I trial in India. PLoS One 2019; 14:e0213911. [PMID: 30921340 PMCID: PMC6438518 DOI: 10.1371/journal.pone.0213911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 03/04/2019] [Indexed: 01/09/2023] Open
Abstract
Effective vaccine design relies on accurate knowledge of protection against a pathogen, so as to be able to induce relevant and effective protective responses against it. An ideal Human Immunodeficiency virus (HIV) vaccine should induce humoral as well as cellular immune responses to prevent initial infection of host cells or limit early events of viral dissemination. A Phase I HIV-1 prophylactic vaccine trial sponsored by the International AIDS Vaccine Initiative (IAVI) was conducted in India in 2009.The trial tested a HIV-1 subtype C vaccine in a prime-boost regimen, comprising of a DNA prime (ADVAX) and Modified Vaccine Ankara (MVA) (TBC-M4) boost. The trial reported that the vaccine regimen was safe, well tolerated, and resulted in enhancement of HIV-specific immune responses. However, preliminary immunological studies were limited to vaccine-induced IFN-γ responses against the Env and Gag peptides. The present study is a retrospective study to characterize in detail the nature of the vaccine-induced cell mediated immune responses among volunteers, using Peripheral Blood Mononuclear Cells (PBMC) that were archived during the trial. ELISpot was used to measure IFN-γ responses and polyfunctional T cells were analyzed by intracellular multicolor flow cytometry. It was observed that DNA priming and MVA boosting induced Env and Gag specific bi-functional and multi-functional CD4+ and CD8+ T cells expressing IFN-γ, TNF-α and IL-2. The heterologous prime-boost regimen appeared to be slightly superior to the homologous prime-boost regimen in inducing favorable cell mediated immune responses. These results suggest that an in-depth analysis of vaccine-induced cellular immune response can aid in the identification of correlates of an effective immunogenic response, and inform future design of HIV vaccines.
Collapse
Affiliation(s)
- Sivasankaran Munusamy Ponnan
- Department of HIV, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Sathyamurthy Pattabiram
- Department of HIV, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Kannan Thiruvengadam
- Department of HIV, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Rajat Goyal
- International AIDS Vaccine Initiative, New Delhi, India
| | - Nikhil Singla
- International AIDS Vaccine Initiative, New Delhi, India
| | | | | | - Philip Bergin
- IAVI Human Immunology Laboratory, Imperial College, London, United Kingdom
| | | | - Jill Gilmour
- IAVI Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Sriram Kumar
- Department of HIV, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Malathy Muthu
- Department of HIV, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Sudha Subramaniam
- Department of HIV, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Soumya Swaminathan
- Department of HIV, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Srikanth Prasad Tripathy
- Department of HIV, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Hanna Elizabeth Luke
- Department of HIV, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
- * E-mail:
| |
Collapse
|
19
|
Won SY, Hunt K, Guak H, Hasaj B, Charland N, Landry N, Ward BJ, Krawczyk CM. Characterization of the innate stimulatory capacity of plant-derived virus-like particles bearing influenza hemagglutinin. Vaccine 2018; 36:8028-8038. [DOI: 10.1016/j.vaccine.2018.10.099] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 10/29/2018] [Accepted: 10/31/2018] [Indexed: 01/29/2023]
|
20
|
Doescher J, Jeske S, Weissinger SE, Brunner C, Laban S, Bölke E, Hoffmann TK, Whiteside TL, Schuler PJ. Polyfunctionality of CD4+ T lymphocytes is increased after chemoradiotherapy of head and neck squamous cell carcinoma. Strahlenther Onkol 2018; 194:392-402. [DOI: 10.1007/s00066-018-1289-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/21/2018] [Indexed: 10/17/2022]
|
21
|
Pillet S, Aubin É, Trépanier S, Poulin JF, Yassine-Diab B, Ter Meulen J, Ward BJ, Landry N. Humoral and cell-mediated immune responses to H5N1 plant-made virus-like particle vaccine are differentially impacted by alum and GLA-SE adjuvants in a Phase 2 clinical trial. NPJ Vaccines 2018; 3:3. [PMID: 29387473 PMCID: PMC5780465 DOI: 10.1038/s41541-017-0043-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 12/11/2017] [Accepted: 12/15/2017] [Indexed: 12/19/2022] Open
Abstract
The hemagglutinination inhibition (HI) response remains the gold standard used for the licensure of influenza vaccines. However, cell-mediated immunity (CMI) deserves more attention, especially when evaluating H5N1 influenza vaccines that tend to induce poor HI response. In this study, we measured the humoral response (HI) and CMI (flow cytometry) during a Phase II dose-ranging clinical trial (NCT01991561). Subjects received two intramuscular doses, 21 days apart, of plant-derived virus-like particles (VLP) presenting the A/Indonesia/05/2005 H5N1 influenza hemagglutinin protein (H5) at the surface of the VLP (H5VLP). The vaccine was co-administrated with Alhydrogel® or with a glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE). We demonstrated that low doses (3.75 or 7.5 μg H5VLP) of GLA-SE-adjuvanted vaccines induced HI responses that met criteria for licensure at both antigen doses tested. Alhydrogel adjuvanted vaccines induced readily detectable HI response that however failed to meet licensure criteria at any of three doses (10, 15 and 20 μg) tested. The H5VLP also induced a sustained (up to 6 months) polyfunctional and cross-reactive HA-specific CD4+ T cell response in all vaccinated groups. Interestingly, the frequency of central memory Th1-primed precursor cells before the boost significantly correlated with HI titers 21 days after the boost. The ability of the low dose GLA-SE-adjuvanted H5VLP to elicit both humoral response and a sustained cross-reactive CMI in healthy adults is very attractive and could result in significant dose-sparing in a pandemic situation.
Collapse
Affiliation(s)
- Stéphane Pillet
- 1Medicago Inc., Québec, G1V 3V9 QC Canada.,2Research Institute of the McGill University Health Centre, Montreal, H4A 3J1 QC Canada
| | - Éric Aubin
- 1Medicago Inc., Québec, G1V 3V9 QC Canada
| | | | | | | | - Jan Ter Meulen
- Immune Design, Seattle, WA 98102 USA.,Immune Design, San Francisco, CA 94080-7006 USA
| | - Brian J Ward
- 2Research Institute of the McGill University Health Centre, Montreal, H4A 3J1 QC Canada
| | | |
Collapse
|
22
|
Sahay B, Aranyos AM, McAvoy A, Yamamoto JK. Utilization of Feline ELISpot to Evaluate the Immunogenicity of a T Cell-Based FIV MAP Vaccine. Methods Mol Biol 2018; 1808:197-219. [PMID: 29956186 DOI: 10.1007/978-1-4939-8567-8_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The prototype and the commercial dual-subtype feline immunodeficiency virus (FIV) vaccines conferred protection against homologous FIV strains as well as heterologous FIV strains from the vaccine subtypes with closely related envelope (Env) sequences. Such protection was mediated by the FIV neutralizing antibodies (NAbs) induced by the vaccines. Remarkably, both prototype and commercial FIV vaccines also conferred protection against heterologous FIV subtypes with highly divergent Env sequences from the vaccine strains. Such protection was not mediated by the vaccine-induced NAbs but was mediated by a potent FIV-specific T-cell immunity generated by the vaccines (Aranyos et al., Vaccine 34: 1480-1488, 2016). The protective epitopes on the FIV vaccine antigen were identified using feline interleukin-2 (IL-2) and interferon-γ (IFNγ) ELISpot assays with overlapping FIV peptide stimulation of the peripheral blood mononuclear cells (PBMC) from cats immunized with prototype FIV vaccine. Two of the protective FIV peptide epitopes were identified on FIV p24 protein and another two protective peptide epitopes were found on FIV reverse transcriptase. In the current study, the multiple antigenic peptides (MAPs) of the four protective FIV peptides were combined with an adjuvant as the FIV MAP vaccine. The laboratory cats were immunized with the MAP vaccine to evaluate whether significant levels of vaccine-specific cytokine responses can be generated to the FIV MAPs and their peptides at post-second and post-third vaccinations. The PBMC from vaccinated cats and non-vaccinated control cats were tested for IL-2, IFNγ, and IL-10 ELISpot responses to the FIV MAPs and peptides. These results were compared to the results from CD4+ and CD8+ T-cell proliferation to the FIV MAPs and peptides. Current study demonstrates that IL-2 and IFNγ ELISpot responses can be used to detect memory responses of the T cells from vaccinated cats after the second and third vaccinations.
Collapse
Affiliation(s)
- Bikash Sahay
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Alek M Aranyos
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Andrew McAvoy
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Janet K Yamamoto
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
23
|
Abstract
Despite major advances in antiretroviral therapy against HIV-1, an effective HIV vaccine is urgently required to reduce the number of new cases of HIV infections in the world. Vaccines are the ultimate tool in the medical arsenal to control and prevent the spread of infectious diseases such as HIV/AIDS. Several failed phase-IIb to –III clinical vaccine trials against HIV-1 in the past generated a plethora of information that could be used for better designing of an effective HIV vaccine in the future. Most of the tested vaccine candidates produced strong humoral responses against the HIV proteins; however, failed to protect due to: 1) the low levels and the narrow breadth of the HIV-1 neutralizing antibodies and the HIV-specific antibody-dependent Fc-mediated effector activities, 2) the low levels and the poor quality of the anti-HIV T-cell responses, and 3) the excessive responses to immunodominant non-protective HIV epitopes, which in some cases blocked the protective immunity and/or enhanced HIV infection. The B-cell epitopes on HIV for producing broadly neutralizing antibodies (bNAbs) against HIV have been extensively characterized, and the next step is to develop bNAb epitope immunogen for HIV vaccine. The bNAb epitopes are often conformational epitopes and therefore more difficult to construct as vaccine immunogen and likely to include immunodominant non-protective HIV epitopes. In comparison, T-cell epitopes are short linear peptides which are easier to construct into vaccine immunogen free of immunodominant non-protective epitopes. However, its difficulty lies in identifying the T-cell epitopes conserved among HIV subtypes and induce long-lasting, potent polyfunctional T-cell and cytotoxic T lymphocyte (CTL) activities against HIV. In addition, these protective T-cell epitopes must be recognized by the HLA prevalent in the country(s) targeted for the vaccine trial. In conclusion, extending from the findings from previous vaccine trials, future vaccines should combine both T- and B-cell epitopes as vaccine immunogen to induce multitude of broad and potent immune effector activities required for sterilizing protection against global HIV subtypes.
Collapse
Affiliation(s)
- Bikash Sahay
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| | - Cuong Q Nguyen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| | - Janet K Yamamoto
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| |
Collapse
|
24
|
Effects of gold nanoparticle-based vaccine size on lymph node delivery and cytotoxic T-lymphocyte responses. J Control Release 2017; 256:56-67. [DOI: 10.1016/j.jconrel.2017.04.024] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/19/2017] [Accepted: 04/17/2017] [Indexed: 01/05/2023]
|
25
|
Xiang B, Baybutt TR, Berman-Booty L, Magee MS, Waldman SA, Alexeev VY, Snook AE. Prime-Boost Immunization Eliminates Metastatic Colorectal Cancer by Producing High-Avidity Effector CD8 + T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:3507-3514. [PMID: 28341670 PMCID: PMC5435941 DOI: 10.4049/jimmunol.1502672] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/27/2017] [Indexed: 11/19/2022]
Abstract
Heterologous prime-boost immunization with plasmid DNA and viral vector vaccines is an emerging approach to elicit CD8+ T cell-mediated immunity targeting pathogens and tumor Ags that is superior to either monotherapy. Yet, the mechanisms underlying the synergy of prime-boost strategies remain incompletely defined. In this study, we examine a DNA and adenovirus (Ad5) combination regimen targeting guanylyl cyclase C (GUCY2C), a receptor expressed by intestinal mucosa and universally expressed by metastatic colorectal cancer. DNA immunization efficacy was optimized by i.m. delivery via electroporation, yet it remained modest compared with Ad5. Sequential immunization with DNA and Ad5 produced superior antitumor efficacy associated with increased TCR avidity, whereas targeted disruption of TCR avidity enhancement eliminated GUCY2C-specific antitumor efficacy, without affecting responding T cell number or cytokine profile. Indeed, functional TCR avidity of responding GUCY2C-specific CD8+ T cells induced by various prime or prime-boost regimens correlated with antitumor efficacy, whereas T cell number and cytokine profile were not. Importantly, although sequential immunization with DNA and Ad5 maximized antitumor efficacy through TCR avidity enhancement, it produced no autoimmunity, reflecting sequestration of GUCY2C to intestinal apical membranes and segregation of mucosal and systemic immunity. Together, TCR avidity enhancement may be leveraged by prime-boost immunization to improve GUCY2C-targeted colorectal cancer immunotherapeutic efficacy and patient outcomes without concomitant autoimmune toxicity.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- CD8-Positive T-Lymphocytes/physiology
- CD8-Positive T-Lymphocytes/transplantation
- Cells, Cultured
- Colorectal Neoplasms/immunology
- Colorectal Neoplasms/therapy
- Cytotoxicity, Immunologic
- Immunity, Mucosal
- Immunization, Secondary
- Immunotherapy, Adoptive/methods
- Intestinal Mucosa/physiology
- Mice
- Mice, Inbred BALB C
- Neoplasm Metastasis
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Enterotoxin
- Receptors, Guanylate Cyclase-Coupled/genetics
- Receptors, Guanylate Cyclase-Coupled/metabolism
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
- Tumor Burden
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Bo Xiang
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107
| | - Trevor R Baybutt
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107
| | - Lisa Berman-Booty
- Department of Discovery Toxicology, Bristol-Myers Squibb, Princeton, NJ 08543
| | - Michael S Magee
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107
- Bluebird Bio, Cambridge, MA 02141; and
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107
| | - Vitali Y Alexeev
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Adam E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107;
| |
Collapse
|
26
|
Schmitt MER, Sitte S, Voehringer D. CD4 T Helper Cells Instruct Lymphopenia-Induced Memory-Like CD8 T Cells for Control of Acute LCMV Infection. Front Immunol 2017; 7:622. [PMID: 28066432 PMCID: PMC5174106 DOI: 10.3389/fimmu.2016.00622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/07/2016] [Indexed: 01/12/2023] Open
Abstract
Lymphopenic conditions lead to expansion of memory-like T cells (TML), which develop from naïve T cells by spontaneous proliferation. TML cells are often increased in the elderly population, AIDS patients, and patients recovering from radio- or chemotherapy. At present, it is unclear whether TML cells can efficiently respond to foreign antigen and participate in antiviral immunity. To address this question, we analyzed the immune response during acute low-dose infection with lymphocytic choriomeningitis virus-WE in T cell lymphopenic CD4Cre/R-diphtheria toxin alpha (DTA) mice in which most peripheral T cells show a TML phenotype. On day 8 after infection, the total number of effector T cells and polyfunctional IFN-γ and TNF-α producing CD8 T cells were three- to fivefold reduced in CD4Cre/R-DTA mice as compared to controls. Viral clearance and the humoral immune response were severely impaired in CD4Cre/R-DTA mice although CTLs efficiently killed transferred target cells in vivo. Transfer of naïve CD4 T cells but not anti-PD-L1 blockade restored the expansion of antigen-specific polyfunctional CD8 T cells and resulted in lower viral titers. This finding indicates that under lymphopenic conditions endogenous CD4 TML cell lack the capacity to promote expansion of CTLs. However, CD8 TML cells retain sufficient functional plasticity to participate in antiviral immunity in the presence of appropriate help by fully functional CD4 T cells. This capacity might be exploited to develop treatments for improvement of CD8 T cell functions under various clinical settings of lymphopenia.
Collapse
Affiliation(s)
- Michaela E R Schmitt
- Department of Infection Biology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Selina Sitte
- Department of Infection Biology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - David Voehringer
- Department of Infection Biology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| |
Collapse
|
27
|
Liu W, Xu Y, Yan J, Shen H, Yang E, Wang H. Ag85B synergizes with ESAT-6 to induce efficient and long-term immunity of C57BL/6 mice primed with recombinant Bacille Calmette-Guerin. Exp Ther Med 2016; 13:208-214. [PMID: 28123491 PMCID: PMC5245152 DOI: 10.3892/etm.2016.3944] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/04/2016] [Indexed: 11/05/2022] Open
Abstract
The latest probable scenario in vaccination strategies is to prime one live attenuated vaccine candidate followed by boost dose of second vaccine candidate. In the present study, we primed the mice with a recombinant Bacille Calmette-Guerin (BCG) comprising Ag85B and ESAT-6 followed by boost doses of Ag85B, ESAT-6 and Ag85B-ESAT-6 fusion protein in the DDA adjuvant, separately. After boost doses of 8 and 12 weeks, the levels of antigen-stimulated T cells secreting interferon (IFN)-γ, the content of the IFN-γ, tumor necrosis factor-α and interleukin-4 in the splenocytes in vitro culture supernatant, the antigen-specific immunoglobulin (Ig)G titer from mouse serum, IgG subclass and the population of antigen-specific CD4+ and CD8+ T cells were detected. The present study showed that recombinant BCG along with boost doses of Ag85B or ESAT-6 individually did not induce efficient T-helper (Th) 1-type immune response. On the other hand, recombinant BCG with boost doses of Ag85B-ESAT-6 fusion protein enhanced longer lasting predominant Th1 immune response. This result suggested that Ag85B might synergize with ESAT-6 protein in order to produce better as well as effective immune response. Thus, the present study concluded recombinant BCG with boost doses of Ag85B-ESAT-6 fusion protein could be a good strategy to improve the immune protective efficacy.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China
| | - Ying Xu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China
| | - Jingran Yan
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China
| | - Hongbo Shen
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China
| | - Enzhuo Yang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China
| | - Honghai Wang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China
| |
Collapse
|
28
|
Pillet S, Aubin É, Trépanier S, Bussière D, Dargis M, Poulin JF, Yassine-Diab B, Ward BJ, Landry N. A plant-derived quadrivalent virus like particle influenza vaccine induces cross-reactive antibody and T cell response in healthy adults. Clin Immunol 2016; 168:72-87. [PMID: 26987887 DOI: 10.1016/j.clim.2016.03.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 02/22/2016] [Accepted: 03/07/2016] [Indexed: 01/09/2023]
Abstract
Recent issues regarding efficacy of influenza vaccines have re-emphasized the need of new approaches to face this major public health issue. In a phase 1-2 clinical trial, healthy adults received one intramuscular dose of a seasonal influenza plant-based quadrivalent virus-like particle (QVLP) vaccine or placebo. The hemagglutination inhibition (HI) titers met all the European licensure criteria for the type A influenza strains at the 3μg/strain dose and for all four strains at the higher dosages 21days after immunization. High HI titers were maintained for most of the strains 6months after vaccination. QVLP vaccine induced a substantial and sustained increase of hemagglutinin-specific polyfunctional CD4 T cells, mainly transitional memory and TEMRA effector IFN-γ(+) CD4 T cells. A T cells cross-reactive response was also observed against A/Hong-Kong/1/1968 H3N2 and B/Massachusetts/2/2012. Plant-based QVLP offers an attractive alternative manufacturing method for producing effective and HA-strain matching seasonal influenza vaccines.
Collapse
Affiliation(s)
- Stéphane Pillet
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9; Research Institute of the McGill University Health Centre, 2155 Guy Street, 5th Floor, Montreal, QC, Canada, H3H 2R9
| | - Éric Aubin
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9
| | - Sonia Trépanier
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9
| | - Diane Bussière
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9
| | - Michèle Dargis
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9
| | | | - Bader Yassine-Diab
- ImmuneCarta, 201 Avenue du Président-Kennedy, Montreal, QC, Canada, H2X 3Y7
| | - Brian J Ward
- Research Institute of the McGill University Health Centre, 2155 Guy Street, 5th Floor, Montreal, QC, Canada, H3H 2R9
| | - Nathalie Landry
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9.
| |
Collapse
|
29
|
Snyder LD, Chan C, Kwon D, Yi JS, Martissa JA, Copeland CAF, Osborne RJ, Sparks SD, Palmer SM, Weinhold KJ. Polyfunctional T-Cell Signatures to Predict Protection from Cytomegalovirus after Lung Transplantation. Am J Respir Crit Care Med 2016; 193:78-85. [PMID: 26372850 DOI: 10.1164/rccm.201504-0733oc] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Cytomegalovirus (CMV), which is one of the most common infections after lung transplantation, is associated with chronic lung allograft dysfunction and worse post-transplantation survival. Current approaches for at-risk patients include a fixed duration of antiviral prophylaxis despite the associated cost and side effects. OBJECTIVES We sought to identify a specific immunologic signature that predicted protection from subsequent CMV. METHODS CMV-seropositive lung transplantation recipients were included in the discovery (n = 43) and validation (n = 28) cohorts. Polyfunctional CMV-specific immunity was assessed by stimulating peripheral blood mononuclear cells with CMV pp65 or IE-1 peptide pools and then by measuring T-cell expression of CD107a, IFN-γ, tumor necrosis factor-α (TNF-α), and IL-2. Recipients were prospectively monitored for subsequent viremia. A Cox proportional hazards regression model that considered cytokine responses individually and in combination was used to create a predictive model for protection from CMV reactivation. This model was then applied to the validation cohort. MEASUREMENTS AND MAIN RESULTS Using the discovery cohort, we identified a specific combination of polyfunctional T-cell subsets to pp65 that predicted protection from subsequent CMV viremia (concordance index 0.88 [SE, 0.087]). The model included both protective (CD107a(-)/IFN-γ(+)/IL-2(+)/TNF-α(+) CD4(+) T cells, CD107a(-)/IFN-γ(+)/IL-2(+)/TNF-α(+) CD8(+) T cells) and detrimental (CD107a(+)/IFN-γ(+)/IL-2(-)/TNF-α(-) CD8(+) T cells) subsets. The model was robust in the validation cohort (concordance index 0.81 [SE, 0.103]). CONCLUSIONS We identified and validated a specific T-cell polyfunctional response to CMV antigen stimulation that provides a clinically useful prediction of subsequent cytomegalovirus risk. This novel diagnostic approach could inform the optimal duration of individual prophylaxis.
Collapse
Affiliation(s)
| | - Cliburn Chan
- 2 Department of Biostatistics and Bioinformatics
| | | | | | | | | | | | | | | | - Kent J Weinhold
- 3 Department of Surgery, and.,4 Department of Immunology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
30
|
McAulay KA, Jarrett RF. Human leukocyte antigens and genetic susceptibility to lymphoma. ACTA ACUST UNITED AC 2016; 86:98-113. [PMID: 26189878 DOI: 10.1111/tan.12604] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Familial aggregation, coupled with ethnic variation in incidence, suggests that inherited susceptibility plays a role in the development of lymphoma, and the search for genetic risk factors has highlighted the contribution of the human leukocyte antigen (HLA) complex. In a landmark study published almost 50 years ago, Hodgkin lymphoma (HL) was the first disease to be associated with HLA variation. It is now clear that Epstein-Barr virus (EBV)-positive and -negative HL are strongly associated with specific HLA polymorphisms but these differ by EBV status of the tumours. HLA class I alleles are consistently associated with EBV-positive HL while a polymorphism in HLA class II is the strongest predictor of risk of EBV-negative HL. Recent investigations, particularly genome-wide association studies (GWAS), have also revealed associations between HLA and common types of non-Hodgkin lymphoma (NHL). Follicular lymphoma is strongly associated with two distinct haplotypes in HLA class II whereas diffuse large B-cell lymphoma is most strongly associated with HLA-B*08. Although chronic lymphocytic leukaemia is associated with variation in HLA class II, the strongest signals in GWAS are from non-HLA polymorphisms, suggesting that inherited susceptibility is explained by co-inheritance of multiple low risk variants. Associations between B-cell derived lymphoma and HLA variation suggest that antigen presentation, or lack of, plays an important role in disease pathogenesis but the precise mechanisms have yet to be elucidated.
Collapse
Affiliation(s)
- K A McAulay
- MRC - University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - R F Jarrett
- MRC - University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
31
|
Fresnay S, McArthur MA, Magder L, Darton TC, Jones C, Waddington CS, Blohmke CJ, Angus B, Levine MM, Pollard AJ, Sztein MB. Salmonella Typhi-specific multifunctional CD8+ T cells play a dominant role in protection from typhoid fever in humans. J Transl Med 2016; 14:62. [PMID: 26928826 PMCID: PMC4772330 DOI: 10.1186/s12967-016-0819-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 02/17/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Typhoid fever, caused by the human-restricted organism Salmonella Typhi (S. Typhi), is a major public health problem worldwide. Development of novel vaccines remains imperative, but is hampered by an incomplete understanding of the immune responses that correlate with protection. METHODS Recently, a controlled human infection model was re-established in which volunteers received ~10(3) cfu wild-type S. Typhi (Quailes strain) orally. Twenty-one volunteers were evaluated for their cell-mediated immune (CMI) responses. Ex vivo PBMC isolated before and up to 1 year after challenge were exposed to three S. Typhi-infected targets, i.e., autologous B lymphoblastoid cell-lines (B-LCL), autologous blasts and HLA-E restricted AEH B-LCL cells. CMI responses were evaluated using 14-color multiparametric flow cytometry to detect simultaneously five intracellular cytokines/chemokines (i.e., IL-17A, IL-2, IFN-g, TNF-a and MIP-1b) and a marker of degranulation/cytotoxic activity (CD107a). RESULTS Herein we provide the first evidence that S. Typhi-specific CD8+ responses correlate with clinical outcome in humans challenged with wild-type S. Typhi. Higher multifunctional S. Typhi-specific CD8+ baseline responses were associated with protection against typhoid and delayed disease onset. Moreover, following challenge, development of typhoid fever was accompanied by decreases in circulating S. Typhi-specific CD8+ T effector/memory (TEM) with gut homing potential, suggesting migration to the site(s) of infection. In contrast, protection against disease was associated with low or no changes in circulating S. Typhi-specific TEM. CONCLUSIONS These studies provide novel insights into the protective immune responses against typhoid disease that will aid in selection and development of new vaccine candidates.
Collapse
Affiliation(s)
- Stephanie Fresnay
- Center for Vaccine Development, University of Maryland School of Medicine, 685 W. Baltimore Street, Suite 480, Baltimore, MD, 21201, USA.
| | - Monica A McArthur
- Center for Vaccine Development, University of Maryland School of Medicine, 685 W. Baltimore Street, Suite 480, Baltimore, MD, 21201, USA.
| | - Laurence Magder
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Thomas C Darton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Claire Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Claire S Waddington
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Christoph J Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Brian Angus
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Myron M Levine
- Center for Vaccine Development, University of Maryland School of Medicine, 685 W. Baltimore Street, Suite 480, Baltimore, MD, 21201, USA.
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Marcelo B Sztein
- Center for Vaccine Development, University of Maryland School of Medicine, 685 W. Baltimore Street, Suite 480, Baltimore, MD, 21201, USA.
| |
Collapse
|
32
|
Wahid R, Fresnay S, Levine MM, Sztein MB. Immunization with Ty21a live oral typhoid vaccine elicits crossreactive multifunctional CD8+ T-cell responses against Salmonella enterica serovar Typhi, S. Paratyphi A, and S. Paratyphi B in humans. Mucosal Immunol 2015; 8:1349-59. [PMID: 25872480 PMCID: PMC4607552 DOI: 10.1038/mi.2015.24] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/06/2015] [Indexed: 02/04/2023]
Abstract
Previously we have extensively characterized Salmonella enterica serovar Typhi (S. Typhi)-specific cell-mediated immune (CMI) responses in volunteers orally immunized with the licensed Ty21a typhoid vaccine. In this study we measured Salmonella-specific multifunctional (MF) CD8+ T-cell responses to further investigate whether Ty21a elicits crossreactive CMI against S. Paratyphi A and S. Paratyphi B that also cause enteric fever. Ty21a-elicited crossreactive CMI responses against all three Salmonella serotypes were predominantly observed in CD8+ T effector/memory (T(EM)) and, to a lesser extent, in CD8+CD45RA+ T(EM) (T(EMRA)) subsets. These CD8+ T-cell responses were largely mediated by MF cells coproducing interferon-γ and macrophage inflammatory protein-1β and expressing CD107a with or without tumor necrosis factor-α. Significant proportions of Salmonella-specific MF cells expressed the gut-homing molecule integrin α4β7. In most subjects, similar MF responses were observed to S. Typhi and S. Paratyphi B, but not to S. Paratyphi A. These results suggest that Ty21a elicits MF CMI responses against Salmonella that could be critical in clearing the infection. Moreover, because S. Paratyphi A is a major public concern and Ty21a was shown in field studies not to afford cross-protection to S. Paratyphi A, these results will be important in developing a S. Typhi/S. Paratyphi A bivalent vaccine against enteric fevers.
Collapse
Affiliation(s)
- Rezwanul Wahid
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Stephanie Fresnay
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Myron M. Levine
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marcelo B. Sztein
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Cárdenas D, Vélez G, Orfao A, Herrera MV, Solano J, Olaya M, Uribe AM, Saavedra C, Duarte M, Rodríguez M, López M, Fiorentino S, Quijano S. Epstein-Barr virus-specific CD8(+) T lymphocytes from diffuse large B cell lymphoma patients are functionally impaired. Clin Exp Immunol 2015; 182:173-83. [PMID: 26174440 PMCID: PMC4608507 DOI: 10.1111/cei.12682] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2015] [Indexed: 12/13/2022] Open
Abstract
Epstein-Barr virus (EBV) is a persistent virus with oncogenic capacity that has been implicated in the development of aggressive B cell lymphomas, primarily in immunosuppressed individuals, although it can be present in immunocompetent individuals. Changes in the function and clonal diversity of T lymphocytes might be implied by viral persistence and lymphoma development. The aim of the present study was to evaluate the frequency, phenotype, function and clonotypical distribution of EBV-specific T cells after peripheral blood stimulation with a virus lysate in newly diagnosed patients with diffuse large B cell lymphoma (DLBCL) aged more than 50 years without prior histories of clinical immunosuppression compared with healthy controls. Our results showed impaired EBV-specific immune responses among DLBCL patients that were associated primarily with decreased numbers of central and effector memory CD8(+) T lymphocytes. In contrast to healthy controls, only a minority of the patients showed CD4(+)/tumour necrosis factor (TNF)-α(+) T cells expressing T cell receptor (TCR)-Vβ17 and CD8(+)/TNF-α(+) T cells with TCR-Vβ5·2, Vβ9 and Vβ18 in response to EBV. Notably, the production of TNF-α was undetectable among TCR-Vβ5·3(+), Vβ11(+), Vβ12(+), Vβ16(+) and Vβ23(+) CD8(+) T cells. In addition, we observed decreased numbers of CD4(+)/TNF-α(+) and CD8(+)/TNF-α(+), CD8(+)/interleukin (IL)-2(+) and CD8(+)/TNF-α(+)/IL-2(+) T lymphocytes in the absence of T cells capable of producing TNF-α, IL-2 and IFN-γ after EBV stimulation simultaneously. Moreover, DLBCL patients displayed higher IL-10 levels both under baseline conditions and after EBV stimulation. These findings were also observed in patients with positive EBV viral loads. Prospective studies including a large number of patients are needed to confirm these findings.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/virology
- Epstein-Barr Virus Infections/blood
- Epstein-Barr Virus Infections/immunology
- Epstein-Barr Virus Infections/virology
- Female
- Flow Cytometry
- Herpesvirus 4, Human/immunology
- Herpesvirus 4, Human/physiology
- Host-Pathogen Interactions/immunology
- Humans
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-10/immunology
- Interleukin-10/metabolism
- Interleukin-2/immunology
- Interleukin-2/metabolism
- Lymphocyte Count
- Lymphoma, Large B-Cell, Diffuse/blood
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/virology
- Male
- Middle Aged
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/metabolism
- Viral Load/immunology
Collapse
Affiliation(s)
- D Cárdenas
- Grupo De Inmunobiología Y Biología Celular Departamento De Microbiología Facultad De Ciencias Pontificia Universidad JaverianaBogotá, Colombia
| | - G Vélez
- Grupo De Inmunobiología Y Biología Celular Departamento De Microbiología Facultad De Ciencias Pontificia Universidad JaverianaBogotá, Colombia
| | - A Orfao
- Servicio General De Citometría Y Departamento De Medicina, Centro De Investigación Del Cáncer (Instituto De Biología Molecular Y Celular Del Cáncer and IBSAL; CSIC-USAL), Universidad De SalamancaSalamanca, España
| | - M V Herrera
- Servicio De Hematología Hospital Universitario San Ignacio-Centro De Oncología Javeriano
| | - J Solano
- Servicio De Hematología Hospital Universitario San Ignacio-Centro De Oncología Javeriano
| | - M Olaya
- Departamento de Patología, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio
| | - A M Uribe
- Departamento de Patología, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio
| | - C Saavedra
- Grupo De Patología Fundación Santa Fe De Bogotá
| | - M Duarte
- Servicio De Hematología Fundación Santa Fe De BogotáBogotá, Colombia
| | - M Rodríguez
- Servicio De Hematología Fundación Santa Fe De BogotáBogotá, Colombia
| | - M López
- Fundación Cardiovascular De ColombiaFloridablanca, Colombia
| | - S Fiorentino
- Grupo De Inmunobiología Y Biología Celular Departamento De Microbiología Facultad De Ciencias Pontificia Universidad JaverianaBogotá, Colombia
| | - S Quijano
- Grupo De Inmunobiología Y Biología Celular Departamento De Microbiología Facultad De Ciencias Pontificia Universidad JaverianaBogotá, Colombia
| |
Collapse
|
34
|
Aberle JH, Schwaiger J, Aberle SW, Stiasny K, Scheinost O, Kundi M, Chmelik V, Heinz FX. Human CD4+ T Helper Cell Responses after Tick-Borne Encephalitis Vaccination and Infection. PLoS One 2015; 10:e0140545. [PMID: 26465323 PMCID: PMC4605778 DOI: 10.1371/journal.pone.0140545] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/28/2015] [Indexed: 12/30/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV) is a human-pathogenic flavivirus that is endemic in large parts of Europe and Asia and causes severe neuroinvasive illness. A formalin-inactivated vaccine induces strong neutralizing antibody responses and confers protection from TBE disease. CD4+ T cell responses are essential for neutralizing antibody production, but data on the functionalities of TBEV-specific CD4+ T cells in response to vaccination or infection are lacking. This study provides a comprehensive analysis of the cytokine patterns of CD4+ T cell responses in 20 humans after TBE vaccination in comparison to those in 18 patients with TBEV infection. Specifically, Th1-specific cytokines (IFN-γ, IL-2, TNF-α), CD40 ligand and the Th1 lineage-specifying transcription factor Tbet were determined upon stimulation with peptides covering the TBEV structural proteins contained in the vaccine (C-capsid, prM/M-membrane and E-envelope). We show that TBEV-specific CD4+ T cell responses are polyfunctional, but the cytokine patterns after vaccination differed from those after infection. TBE vaccine responses were characterized by lower IFN-γ responses and high proportions of TNF-α+IL-2+ cells. In vaccine-induced responses—consistent with the reduced IFN-γ expression patterns—less than 50% of TBEV peptides were detected by IFN-γ+ cells as compared to 96% detected by IL-2+ cells, indicating that the single use of IFN-γ as a read-out strongly underestimates the magnitude and breadth of such responses. The results provide important insights into the functionalities of CD4+ T cells that coordinate vaccine responses and have direct implications for future studies that address epitope specificity and breadth of these responses.
Collapse
Affiliation(s)
- Judith H. Aberle
- Department of Virology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| | - Julia Schwaiger
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | - Stephan W. Aberle
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | - Ondrej Scheinost
- Laboratory of Molecular Genetics, Hospital České Budĕjovice, České Budĕjovice, Czech Republic
| | - Michael Kundi
- Centre for Public Health, Medical University of Vienna, Vienna, Austria
| | - Vaclav Chmelik
- Department of Infectious Diseases, Hospital České Budĕjovice, České Budĕjovice, Czech Republic
| | - Franz X. Heinz
- Department of Virology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
A comparative phase 1 clinical trial to identify anti-infective mechanisms of vitamin D in people with HIV infection. AIDS 2015; 29:1127-35. [PMID: 25870995 PMCID: PMC4516350 DOI: 10.1097/qad.0000000000000666] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objectives: To determine if there is a biological mechanism that explains the association between HIV disease progression and increased mortality with low circulating vitamin D levels; specifically, to determine if restoring vitamin D levels induced T-cell functional changes important for antiviral immunity. Design: This was a pilot, open-label, three-arm prospective phase 1 study. Methods: We recruited 28 patients with low plasma vitamin D (<50 nmol/l 25-hydroxyvitamin D3), comprising 17 HIV+ patients (11 on HAART, six treatment-naive) and 11 healthy controls, who received a single dose of 200 000 IU oral cholecalciferol. Advanced T-cell flow cytometry methods measured CD4+ T-cell function associated with viral control in blood samples at baseline and 1-month after vitamin D supplementation. Results: One month of vitamin D supplementation restored plasma levels to sufficiency (>75 nmol/l) in 27 of 28 patients, with no safety issues. The most striking change was in HIV+ HAART+ patients, where increased frequencies of antigen-specific T cells expressing macrophage inflammatory protein (MIP)-1β – an important anti-HIV blocking chemokine – were observed, with a concomitant increase in plasma MIP-1β, both of which correlated significantly with vitamin D levels. In addition, plasma cathelicidin – a vitamin D response gene with broad antimicrobial activity – was enhanced. Conclusion: Vitamin D supplementation modulates disease-relevant T-cell functions in HIV-infected patients, and may represent a useful adjunct to HAART therapy.
Collapse
|
36
|
Chattopadhyay PK, Roederer M. A mine is a terrible thing to waste: high content, single cell technologies for comprehensive immune analysis. Am J Transplant 2015; 15:1155-61. [PMID: 25708158 DOI: 10.1111/ajt.13193] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/22/2014] [Accepted: 12/26/2014] [Indexed: 01/25/2023]
Abstract
In recent years, an incredible variety of single cell technologies have become available to analyze immune responses. These technologies include polychromatic flow cytometry, mass cytometry, highly multiplexed single cell qPCR, RNA sequencing, microtools, and high-resolution imaging. In this article, we review these platforms, describing their power and limitations for comprehensive analysis of the immune system. We relate the properties of these technologies to the various cellular states relevant to an immune response, in order to address which technologies are most appropriate for which settings.
Collapse
Affiliation(s)
- P K Chattopadhyay
- Vaccine Research Center, National Institutes of Health, Bethesda, MD
| | | |
Collapse
|
37
|
Zehn D, Wherry EJ. Immune Memory and Exhaustion: Clinically Relevant Lessons from the LCMV Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 850:137-52. [PMID: 26324351 DOI: 10.1007/978-3-319-15774-0_10] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of dysfunctional or exhausted T cells is characteristic of immune responses to chronic viral infections and cancer. Exhausted T cells are defined by reduced effector function, sustained upregulation of multiple inhibitory receptors, an altered transcriptional program and perturbations of normal memory development and homeostasis. This review focuses on (a) illustrating milestone discoveries that led to our present understanding of T cell exhaustion, (b) summarizing recent developments in the field, and (c) identifying new challenges for translational research. Exhausted T cells are now recognized as key therapeutic targets in human infections and cancer. Much of our knowledge of the clinically relevant process of exhaustion derives from studies in the mouse model of Lymphocytic choriomeningitis virus (LCMV) infection. Studies using this model have formed the foundation for our understanding of human T cell memory and exhaustion. We will use this example to discuss recent advances in our understanding of T cell exhaustion and illustrate the value of integrated mouse and human studies and will emphasize the benefits of bi-directional mouse-to-human and human-to-mouse research approaches.
Collapse
Affiliation(s)
- D Zehn
- Division of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland,
| | | |
Collapse
|
38
|
Choi JH, Jonsson-Schmunk K, Qiu X, Shedlock DJ, Strong J, Xu JX, Michie KL, Audet J, Fernando L, Myers MJ, Weiner D, Bajrovic I, Tran LQ, Wong G, Bello A, Kobinger GP, Schafer SC, Croyle MA. A Single Dose Respiratory Recombinant Adenovirus-Based Vaccine Provides Long-Term Protection for Non-Human Primates from Lethal Ebola Infection. Mol Pharm 2014; 12:2712-31. [PMID: 25363619 PMCID: PMC4525323 DOI: 10.1021/mp500646d] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
![]()
As
the Ebola outbreak in West Africa continues and cases appear
in the United States and other countries, the need for long-lasting
vaccines to preserve global health is imminent. Here, we evaluate
the long-term efficacy of a respiratory and sublingual (SL) adenovirus-based
vaccine in non-human primates in two phases. In the first, a single
respiratory dose of 1.4 × 109 infectious virus particles
(ivp)/kg of Ad-CAGoptZGP induced strong Ebola glycoprotein (GP) specific
CD8+ and CD4+ T cell responses and Ebola GP-specific
antibodies in systemic and mucosal compartments and was partially
(67%) protective from challenge 62 days after immunization. The same
dose given by the SL route induced Ebola GP-specific CD8+ T cell responses similar to that of intramuscular (IM) injection,
however, the Ebola GP-specific antibody response was low. All primates
succumbed to infection. Three primates were then given the vaccine
in a formulation that improved the immune response to Ebola in rodents.
Three primates were immunized with 2.0 × 1010 ivp/kg
of vaccine by the SL route. Diverse populations of polyfunctional
Ebola GP-specific CD4+ and CD8+ T cells and
significant anti-Ebola GP antibodies were present in samples collected
150 days after respiratory immunization. The formulated vaccine was
fully protective against challenge 21 weeks after immunization. While
diverse populations of Ebola GP-specific CD4+ T cells were
produced after SL immunization, antibodies were not neutralizing and
the vaccine was unprotective. To our knowledge, this is the first
time that durable protection from a single dose respiratory adenovirus-based
Ebola vaccine has been demonstrated in primates.
Collapse
Affiliation(s)
- Jin Huk Choi
- †Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Kristina Jonsson-Schmunk
- †Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Xiangguo Qiu
- §Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
| | - Devon J Shedlock
- ∥Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jim Strong
- §Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
| | - Jason X Xu
- †Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Kelly L Michie
- ⊥The University of Texas College of Natural Sciences Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jonathan Audet
- §Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
| | - Lisa Fernando
- §Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
| | - Mark J Myers
- †Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - David Weiner
- ∥Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Irnela Bajrovic
- #Department of Biochemistry, College of Natural Sciences, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Lilian Q Tran
- †Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Gary Wong
- §Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
| | - Alexander Bello
- §Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
| | - Gary P Kobinger
- §Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada.,∥Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Stephen C Schafer
- †Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Maria A Croyle
- †Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States.,⊥The University of Texas College of Natural Sciences Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, United States.,¶Center for Infectious Disease, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
39
|
Toward development of a comprehensive external quality assurance program for polyfunctional intracellular cytokine staining assays. J Immunol Methods 2014; 409:44-53. [PMID: 24968072 DOI: 10.1016/j.jim.2014.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 05/23/2014] [Accepted: 05/28/2014] [Indexed: 11/21/2022]
Abstract
The External Quality Assurance Program Oversight Laboratory (EQAPOL) Flow Cytometry Program assesses the proficiency of NIH/NIAID/DAIDS-supported and potentially other interested research laboratories in performing Intracellular Cytokine Staining (ICS) assays. The goal of the EQAPOL Flow Cytometry External Quality Assurance Program (EQAP) is to provide proficiency testing and remediation for participating sites. The program is not punitive; rather, EQAPOL aims to help sites identify areas for improvement. EQAPOL utilizes a highly standardized ICS assay to minimize variability and readily identify those sites experiencing technical difficulties with their assays. Here, we report the results of External Proficiency 3 (EP3) where participating sites performed a 7-color ICS assay. On average, sites perform well in the Flow Cytometry EQAP (median score is "Good"). The most common technical issues identified by the program involve protocol adherence and data analysis; these areas have been the focus of site remediation. The EQAPOL Flow Cytometry team is now in the process of expanding the program to 8-color ICS assays. Evaluating polyfunctional ICS responses would align the program with assays currently being performed in support of HIV immune monitoring assays.
Collapse
|
40
|
Kidzeru EB, Hesseling AC, Passmore JAS, Myer L, Gamieldien H, Tchakoute CT, Gray CM, Sodora DL, Jaspan HB. In-utero exposure to maternal HIV infection alters T-cell immune responses to vaccination in HIV-uninfected infants. AIDS 2014; 28:1421-30. [PMID: 24785950 PMCID: PMC4333196 DOI: 10.1097/qad.0000000000000292] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE In sub-Saharan Africa, HIV-exposed uninfected (HEU) infants have higher morbidity and mortality than HIV-unexposed infants. To evaluate whether immune dysfunction contributes to this vulnerability of HEU infants, we conducted a longitudinal, observational cohort study to assess T-cell immune responses to infant vaccines (Mycobacterium bovis BCG and acellular pertussis) and staphylococcal enterotoxin B (SEB). In total, 46 HEU and 46 HIV-unexposed infants were recruited from Khayelitsha, Cape Town. METHODS Vaccine-specific T-cell proliferation (Ki67 expression) and intracellular expression of four cytokines [interferon-γ, interleukin (IL)-2, IL-13 and IL-17] were measured after whole blood stimulation with antigens at 6 and 14 weeks of age. RESULTS HEU infants demonstrated elevated BCG-specific CD4 and CD8 T-cell proliferative responses at 14 weeks (P = 0.041 and 0.002, respectively). These responses were significantly increased even after adjusting for birth weight, feeding mode and gestational age. Similar to BCG, increased CD4 and CD8 T-cell proliferation was evident in response to SEB stimulation (P = 0.004 and 0.002, respectively), although pertussis-specific T cells proliferated comparably between the two groups. Within HEU infants, maternal CD4 cell count and length of antenatal antiretroviral exposure had no effect on T-cell proliferation to BCG or SEB. HIV exposure significantly diminished measurable cytokine polyfunctionality in response to BCG, Bordetella pertussis and SEB stimulation. CONCLUSION These data show for the first time, when adjusting for confounders, that exposure to HIV in utero is associated with significant alterations to CD4 and CD8T-cell immune responses in infants to vaccines and nonspecific antigens.
Collapse
Affiliation(s)
- Elvis B. Kidzeru
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town
| | - Anneke C. Hesseling
- Desmond Tutu TB Centre, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow
| | - Jo-Ann S. Passmore
- Division of Virology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town
- National Health Laboratory Services, South Africa
| | - Landon Myer
- Division of Epidemiology & Biostatistics, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Hoyam Gamieldien
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town
- Division of Virology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town
| | | | - Clive M. Gray
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town
- National Health Laboratory Services, South Africa
| | | | - Heather B. Jaspan
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town
- Seattle Biomedical Research Institute, Seattle, Washington, USA
| |
Collapse
|
41
|
Epithelial immunization induces polyfunctional CD8+ T cells and optimal mousepox protection. J Virol 2014; 88:9472-5. [PMID: 24899206 DOI: 10.1128/jvi.01464-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We assessed several routes of immunization with vaccinia virus (VACV) in protecting mice against ectromelia virus (ECTV). By a wide margin, skin scarification provided the greatest protection. Humoral immunity and resident-memory T cells notwithstanding, several approaches revealed that circulating, memory CD8(+) T cells primed via scarification were functionally superior and conferred enhanced virus control. Immunization via the epithelial route warrants further investigation, as it may also provide enhanced defense against other infectious agents.
Collapse
|
42
|
Hillyer P, Raviv N, Gold DM, Dougherty D, Liu J, Johnson TR, Graham BS, Rabin RL. Subtypes of type I IFN differentially enhance cytokine expression by suboptimally stimulated CD4(+) T cells. Eur J Immunol 2013; 43:3197-208. [PMID: 24030809 DOI: 10.1002/eji.201243288] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 07/17/2013] [Accepted: 09/03/2013] [Indexed: 01/08/2023]
Abstract
Human type I interferons (IFNs) include IFN-β and 12 subtypes of IFN-α. During viral infection, infiltrating memory CD4(+) T cells are exposed to IFNs, but their impact on memory T-cell function is poorly understood. To address this, we pretreated PBMCs with different IFNs for 16 h before stimulation with Staphylococcus aureus enterotoxin B and measured cytokine expression by flow cytometry. IFN-α8 and -α10 most potently enhanced expression of IFN-γ, IL-2, and IL-4. Potency among the subtypes differed most at doses between 10 and 100 U/mL. While enhancement of IL-2 and IL-4 correlated with the time of preincubation with type I IFN, IFN-γ production was enhanced best when IFN-α was added immediately preceding or simultaneously with T-cell stimulation. Comparison of T-cell responses to multiple doses of Staphylococcus aureus enterotoxin B and to peptide libraries from RSV or CMV demonstrated that IFN-α best enhanced cytokine expression when CD4(+) T cells were suboptimally stimulated. We conclude that type I IFNs enhance Th1 and Th2 function with dose dependency and subtype specificity, and best when T-cell stimulation is suboptimal. While type I IFNs may beneficially enhance CD4(+) T-cell memory responses to vaccines or viral pathogens, they may also enhance the function of resident Th2 cells and exacerbate allergic inflammation.
Collapse
Affiliation(s)
- Philippa Hillyer
- Laboratory of Immunobiochemistry, Division of Bacterial, Parasitic and Allergenic Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Bethesda, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Immune monitoring in cancer vaccine clinical trials: critical issues of functional flow cytometry-based assays. BIOMED RESEARCH INTERNATIONAL 2013; 2013:726239. [PMID: 24195078 PMCID: PMC3806162 DOI: 10.1155/2013/726239] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/19/2013] [Indexed: 11/17/2022]
Abstract
The development of immune monitoring assays is essential to determine the immune responses against tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs) and their possible correlation with clinical outcome in cancer patients receiving immunotherapies. Despite the wide range of techniques used, to date these assays have not shown consistent results among clinical trials and failed to define surrogate markers of clinical efficacy to antitumor vaccines. Multiparameter flow cytometry- (FCM-) based assays combining different phenotypic and functional markers have been developed in the past decade for informative and longitudinal analysis of polyfunctional T-cells. These technologies were designed to address the complexity and functional heterogeneity of cancer biology and cellular immunity and to define biomarkers predicting clinical response to anticancer treatment. So far, there is still a lack of standardization of some of these immunological tests. The aim of this review is to overview the latest technologies for immune monitoring and to highlight critical steps involved in some of the FCM-based cellular immune assays. In particular, our laboratory is focused on melanoma vaccine research and thus our main goal was the validation of a functional multiparameter test (FMT) combining different functional and lineage markers to be applied in clinical trials involving patients with melanoma.
Collapse
|
44
|
Ortiz YM, Arias LF, Álvarez CM, García LF. Memory phenotype and polyfunctional T cells in kidney transplant patients. Transpl Immunol 2013; 28:127-37. [DOI: 10.1016/j.trim.2013.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/19/2013] [Accepted: 02/20/2013] [Indexed: 10/27/2022]
|
45
|
Knowlton ER, Lepone LM, Li J, Rappocciolo G, Jenkins FJ, Rinaldo CR. Professional antigen presenting cells in human herpesvirus 8 infection. Front Immunol 2013; 3:427. [PMID: 23346088 PMCID: PMC3549500 DOI: 10.3389/fimmu.2012.00427] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/24/2012] [Indexed: 12/18/2022] Open
Abstract
Professional antigen presenting cells (APC), i.e., dendritic cells (DC), monocytes/macrophages, and B lymphocytes, are critically important in the recognition of an invading pathogen and presentation of antigens to the T cell-mediated arm of immunity. Human herpesvirus 8 (HHV-8) is one of the few human viruses that primarily targets these APC for infection, altering their cytokine profiles, manipulating their surface expression of MHC molecules, and altering their ability to activate HHV-8-specific T cells. This could be why T cell responses to HHV-8 antigens are not very robust. Of these APC, only B cells support complete, lytic HHV-8 infection. However, both complete and abortive virus replication cycles in APC could directly affect viral pathogenesis and progression to Kaposi's sarcoma (KS) and HHV-8-associated B cell cancers. In this review, we discuss the effects of HHV-8 infection on professional APC and their relationship to the development of KS and B cell lymphomas.
Collapse
Affiliation(s)
- Emilee R Knowlton
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
46
|
Costa C, Saldan A, Cavallo R. Evaluation of virus-specific cellular immune response in transplant patients. World J Virol 2012; 1:150-3. [PMID: 24175220 PMCID: PMC3782278 DOI: 10.5501/wjv.v1.i6.150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 08/23/2012] [Accepted: 11/07/2012] [Indexed: 02/05/2023] Open
Abstract
Virus-specific immune responses have a major impact on the outcome of the infection. Viral agents that are characterized by latency, such as herpesviruses and polyomaviruses, require a continuous immune control to reduce the extent of viral reactivation, as viral clearance cannot be accomplished, independently from the anti-viral treatment. In transplant patients, morbidity and mortality related to viral infections are significantly increased. In fact, the key steps of activation of T-cells are major target for anti-rejection immunosuppressive therapy and anti-viral immune response may be altered when infected cells and cellular effectors of immune response coexist in a transplanted organ. The role of cellular immune response in controlling viral replication and the main methods employed for its evaluation will be discussed. In particular, the main features, including both advantages and limitations, of available assays, including intracellular cytokine staining, major histocompatibility complex - multimer-based assays, Elispot assay, and QuantiFERON test, will be described. The potential applications of these assays in the transplant context will be discussed, particularly in relation to cytomegalovirus and polyomavirus BK infection. The relevance of introducing viro-immunological monitoring, beside virological monitoring, in order to identify the risk profile for viral infections in the transplant patients will allows for define a patient-tailored clinical management, particular in terms of modulation of immunosuppressive therapy and anti-viral administration.
Collapse
Affiliation(s)
- Cristina Costa
- Cristina Costa, Alda Saldan, Rossana Cavallo, Virology Unit, University Hospital San Giovanni Battista di Torino, 10126 Turin, Italy
| | | | | |
Collapse
|
47
|
Karulin AY, Quast S, Hesse MD, Lehmann PV. Neuroantigen-Specific CD4 Cells Expressing Interferon-γ (IFN-γ), Interleukin (IL)-2 and IL-3 in a Mutually Exclusive Manner Prevail in Experimental Allergic Encephalomyelitis (EAE). Cells 2012; 1:576-96. [PMID: 24710491 PMCID: PMC3901106 DOI: 10.3390/cells1030576] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 07/28/2012] [Accepted: 07/31/2012] [Indexed: 12/14/2022] Open
Abstract
Experimental allergic encephalomyelitis (EAE) is mediated by neuroantigen-specific pro-inflammatory T cells of the Th1 and Th17 effector class. Th-17 cells can be clearly defined by expression of IL-17, but not IFN-γ, IL-2 or IL-3. Th1 cells do not express IL-17, but it is unclear presently to what extent they co-express the cytokines canonically assigned to Th1 immunity (i.e., IFN-γ, IL-2 and IL-3) and whether CD4 cells producing these cytokines indeed belong to a single Th1 lineage. It is also unclear to what extent the Th1 response in EAE entails polyfunctional T cells that co-express IFN-γ and IL-2. Therefore, we dissected the Th1 cytokine signature of neuroantigen-specific CD4 cells studying at single cell resolution co-expression of IFN-γ, IL-2 and IL-3 using dual color cytokine ELISPOT analysis. Shortly after immunization, in the draining lymph nodes (dLN), the overall cytokine signature of the neuroantigen-specific CD4 cells was highly type 1-polarized, but IFN-γ, IL-2, and IL-3 were each secreted by different CD4 cells in a mutually exclusive manner. This single cell - single cytokine profile was stable through the course of chronic EAE-polyfunctional CD4 cells co-expressing IL-2 and IFN-γ presented less than 5% of the neuroantigen-specific T cells, even in the inflamed CNS itself. The neuroantigen-specific CD4 cells that expressed IFN-γ, IL-2 and IL-3 in a mutually exclusive manner exhibited similar functional avidities and kinetics of cytokine production, but showed different tissue distributions. These data suggest that Th1 cells do not belong to a single lineage, but different Th1 subpopulations jointly mediate Th1 immunity.
Collapse
Affiliation(s)
- Alexey Y Karulin
- Vice President R&D, Cellular Technology Ltd., 20521 Chagrin Blvd., Cleveland, OH 44122, USA.
| | - Stefan Quast
- Cognitive Neurology Section, Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Juelich, Germany.
| | - Maike D Hesse
- Cognitive Neurology Section, Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Juelich, Germany.
| | - Paul V Lehmann
- President and CEO, Cellular Technology Ltd., Cleveland, OH 44122, USA.
| |
Collapse
|
48
|
Bhadra R, Gigley JP, Khan IA. PD-1-mediated attrition of polyfunctional memory CD8+ T cells in chronic toxoplasma infection. J Infect Dis 2012; 206:125-34. [PMID: 22539813 DOI: 10.1093/infdis/jis304] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We reported earlier that during chronic toxoplasmosis CD8(+) T cells become functionally exhausted with concomitant PD-1 upregulation, leading to eventual host mortality. However, how immune exhaustion specifically mediates attrition of CD8 polyfunctionality, a hallmark of potent T-cell response, during persistent infections has not been addressed. In this study, we demonstrate that PD-1 is preferentially expressed on polyfunctional memory CD8(+) T cells, which renders them susceptible to apoptosis. In vitro blockade of the PD-1-PD-L1 pathway dramatically reduces apoptosis of polyfunctional and interferon γ(+)/granzyme B(-) memory but not effector CD8(+) T cells. In summary, the present report underscores the critical role of the PD-1-PD-L1 pathway in mediating attrition of this important CD8(+) T-cell subset and addresses the mechanistic basis of how αPD-L1 therapy reinvigorates polyfunctional CD8 response during chronic infections. The conclusions of this study can have profound immunotherapeutic implications in combating recrudescent toxoplasmosis as well other chronic infections.
Collapse
Affiliation(s)
- Rajarshi Bhadra
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington, DC DC20037, USA
| | | | | |
Collapse
|
49
|
Novel approach for improved assessment of phenotypic and functional characteristics of BKV-specific T-cell immunity. Transplantation 2012; 92:1269-77. [PMID: 22124284 DOI: 10.1097/tp.0b013e318234e0e5] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND BKV-associated nephropathy represents a serious complication of the posttransplant period in kidney transplant recipients. Monitoring BKV-specific immunity is of a special importance for estimation of clinical course in patients with BKV reactivation. Our recent data demonstrated that all five BKV antigens are immunogenic and elicit T-cell responses varying within patients. Therefore, all five BKV proteins should be evaluated for the assessment of BKV-specific immunity. However, analysis of five proteins performed separately is time- and cost-intensive and requires large amount of blood. METHODS Using novel approach of a mixture of overlapping peptide pools encompassing all five BKV antigens (viral protein [VP] 1, VP2, VP3, large tumor antigen, and small tumor antigen) and multiparameter flow cytometry, we evaluate BKV-specific T cells in patients with a previous/present severe long-lasting or transient BKV reactivation. Patients without BKV reactivation were used as control. RESULTS In this study, we show that using mixture of overlapping peptide pool results in the magnitude of CD4- and CD8-positive BKV-specific T-cell response, which is significantly higher compared with any frequencies detected by previously used single BKV antigen stimulation. Of interest, patients with a history of rapid BKV clearance had significantly higher frequency of multifunctional interferon gamma-γ/interleukin (IL)-2/tumor necrosis factor-α and IL-2/tumor necrosis factor-α CD4-positive T cells, suggesting protective potential of polyfunctional T cells. Furthermore, we did not find IL-17-producing BKV-specific memory T cells in patients recovered from BKV reactivation. CONCLUSIONS Here, we established a fast and sensitive approach allowing the most comprehensive assessment of the total BKV immunity performed to date and offer a new platform for further prospective studies.
Collapse
|
50
|
Ning RJ, Xu XQ, Chan KH, Chiang AKS. Long-term carriers generate Epstein-Barr virus (EBV)-specific CD4(+) and CD8(+) polyfunctional T-cell responses which show immunodominance hierarchies of EBV proteins. Immunology 2011; 134:161-71. [PMID: 21896011 DOI: 10.1111/j.1365-2567.2011.03476.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
T cells simultaneously producing multiple cytokines and possessing cytotoxic capacity termed polyfunctional cells (PFCs) are increasingly recognized as the immune correlate of protection against pathogenic viruses. We investigated co-expression of four cytokines (interferon-γ, macrophage inflammatory protein 1-α, tumour necrosis factor-α and interleukin-2) and degranulation capacity (CD107a surface expression) of Epstein-Barr virus (EBV) -specific CD4(+) and CD8(+) T cells upon stimulation by overlapping peptides of EBV lytic (BZLF1) and latent (EBNA1, EBNA3 and LMP2) proteins, in 20 healthy Chinese long-term carriers. Two patients with post-transplant lymphoproliferative disorder (PTLD), who had impaired T-cell immunity, were studied for comparison. Both EBV-specific CD4(+) and CD8(+) PFCs were readily generated in long-term carriers and showed immunodominance hierarchies of latent proteins (EBNA1 > EBNA3/LMP2 and EBNA3 > LMP2 > EBNA1 for CD4(+) and CD8(+) T cells, respectively), as evidenced by a higher proportion of PFCs generated by immunodominant EBV proteins than by subdominant viral proteins. In contrast, the proportion of EBV-specific PFCs was markedly decreased in patients with PTLD. The EBV-specific PFCs produced more cytokine per cell than single-functional T cells and comprised different subsets. Five-functional CD4(+) and CD8(+) T cells were detected and four-functional CD4(+) T cells were mainly CD107a negative and expressed all four cytokines whereas four-functional CD8(+) T cells were mainly CD107a positive and expressed three of the four cytokines (interleukin-2-negative). We conclude that EBV-specific PFCs are generated in much higher proportions in the long-term carriers than in the patients with PTLD and maintain the immunodominant characteristics of the virus.
Collapse
Affiliation(s)
- Raymond J Ning
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|