1
|
Ghahari N, Shegefti S, Alaei M, Amara A, Telittchenko R, Isnard S, Routy JP, Olagnier D, van Grevenynghe J. HSP60 controls mitochondrial ATP generation for optimal virus-specific IL-21-producing CD4 and cytotoxic CD8 memory T cell responses. Commun Biol 2024; 7:1688. [PMID: 39709477 DOI: 10.1038/s42003-024-07326-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
We have shown that virus-specific CD4 and CD8 memory T cells (TM) induce autophagy after T cell receptor (TCR) engagement to provide free glutamine and fatty acids, including in people living with HIV-1 (PLWH). These nutrients fuel mitochondrial ATP generation through glutaminolysis and fatty acid oxidation (FAO) pathways, to fulfill the bioenergetic demands for optimal IL-21 and cytotoxic molecule production in CD4 and CD8 cells, respectively. Here, we expand our knowledge on how the metabolic events that occur in the mitochondria of virus-specific TM down-stream of the autophagy are regulated. We show that HSP60 chaperone positively regulates the protein levels for multiple glutaminolysis- and FAO-related enzymes, thereby actively fueling the levels of cellular alpha-ketoglutarate (αKG) and related mitochondrial ATP-dependent antiviral T cell immunity in both CD4 and CD8 TM. Finally, we provide a way to rescue defective ATP generation in mitochondria and dependent effector functions in virus-specific TM including anti-HIV-1 protective responses, when HSP60 expression is impaired after TCR engagement in patients, in the form of dimethyl 2-oxoglutarate (DMKG) supplementation.
Collapse
Affiliation(s)
- Nazanin Ghahari
- Institut national de la recherche scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, 531 boulevard des Prairies, H7V 1M7, Laval, QC, Canada
| | - Saina Shegefti
- Institut national de la recherche scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, 531 boulevard des Prairies, H7V 1M7, Laval, QC, Canada
| | - Mahsa Alaei
- Institut national de la recherche scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, 531 boulevard des Prairies, H7V 1M7, Laval, QC, Canada
| | - Amine Amara
- Institut national de la recherche scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, 531 boulevard des Prairies, H7V 1M7, Laval, QC, Canada
| | - Roman Telittchenko
- Institut national de la recherche scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, 531 boulevard des Prairies, H7V 1M7, Laval, QC, Canada
| | - Stéphane Isnard
- Chronic Viral Illness Service and Division of Hematology, McGill University Health Centre, Glen site, H4A 3J1, Montreal, Quebec, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illness Service and Division of Hematology, McGill University Health Centre, Glen site, H4A 3J1, Montreal, Quebec, Canada
| | - David Olagnier
- Aarhus University; Department of Biomedicine, Aarhus C, 8000, Denmark
| | - Julien van Grevenynghe
- Institut national de la recherche scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, 531 boulevard des Prairies, H7V 1M7, Laval, QC, Canada.
| |
Collapse
|
2
|
Bliss CM, Nachbagauer R, Mariottini C, Cuevas F, Feser J, Naficy A, Bernstein DI, Guptill J, Walter EB, Berlanda-Scorza F, Innis BL, García-Sastre A, Palese P, Krammer F, Coughlan L. A chimeric haemagglutinin-based universal influenza virus vaccine boosts human cellular immune responses directed towards the conserved haemagglutinin stalk domain and the viral nucleoprotein. EBioMedicine 2024; 104:105153. [PMID: 38805853 PMCID: PMC11154122 DOI: 10.1016/j.ebiom.2024.105153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND The development of a universal influenza virus vaccine, to protect against both seasonal and pandemic influenza A viruses, is a long-standing public health goal. The conserved stalk domain of haemagglutinin (HA) is a promising vaccine target. However, the stalk is immunosubdominant. As such, innovative approaches are required to elicit robust immunity against this domain. In a previously reported observer-blind, randomised placebo-controlled phase I trial (NCT03300050), immunisation regimens using chimeric HA (cHA)-based immunogens formulated as inactivated influenza vaccines (IIV) -/+ AS03 adjuvant, or live attenuated influenza vaccines (LAIV), elicited durable HA stalk-specific antibodies with broad reactivity. In this study, we sought to determine if these vaccines could also boost T cell responses against HA stalk, and nucleoprotein (NP). METHODS We measured interferon-γ (IFN-γ) responses by Enzyme-Linked ImmunoSpot (ELISpot) assay at baseline, seven days post-prime, pre-boost and seven days post-boost following heterologous prime:boost regimens of LAIV and/or adjuvanted/unadjuvanted IIV-cHA vaccines. FINDINGS Our findings demonstrate that immunisation with adjuvanted cHA-based IIVs boost HA stalk-specific and NP-specific T cell responses in humans. To date, it has been unclear if HA stalk-specific T cells can be boosted in humans by HA-stalk focused universal vaccines. Therefore, our study will provide valuable insights for the design of future studies to determine the precise role of HA stalk-specific T cells in broad protection. INTERPRETATION Considering that cHA-based vaccines also elicit stalk-specific antibodies, these data support the further clinical advancement of cHA-based universal influenza vaccine candidates. FUNDING This study was funded in part by the Bill and Melinda Gates Foundation (BMGF).
Collapse
Affiliation(s)
- Carly M Bliss
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Cancer & Genetics and Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Mariottini
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Frans Cuevas
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jodi Feser
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Abdi Naficy
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - David I Bernstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffrey Guptill
- Duke Early Phase Clinical Research Unit, Duke Clinical Research Institute, Durham, NC, USA
| | - Emmanuel B Walter
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | | | - Bruce L Innis
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lynda Coughlan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; University of Maryland School of Medicine, Department of Microbiology and Immunology, Baltimore, MD 21201, USA; University of Maryland School of Medicine, Center for Vaccine Development and Global Health (CVD), Baltimore, MD 21201, USA.
| |
Collapse
|
3
|
Traska AK, Nowacki TM, Vollenberg R, Rennebaum F, Meier JA, Schomacher T, Reinartz Groba SN, Fischer J, Trebicka J, Tepasse PR. Immunomonitoring via ELISPOT Assay Reveals Attenuated T-Cell Immunity to CMV in Immunocompromised Liver-Transplant Patients. Cells 2024; 13:741. [PMID: 38727277 PMCID: PMC11083338 DOI: 10.3390/cells13090741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/12/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
Assessing immune responses to cytomegalovirus (CMV) after liver transplant in patients on immunosuppressive therapy remains challenging. In this study, employing ELISPOT assays, 52 liver-transplant recipients were evaluated for antiviral T-cell activity in peripheral blood mononuclear cells (PBMCs), measuring interferon-γ (IFN-γ) secretion upon stimulation with CMV-specific peptides (CMV peptide pool, CMV IE-1, and pp65 antigens). Parameters such as stimulation index, mean spot size, and mean spot count were measured. The study found that heightened immunosuppression, especially with prednisolone in triple therapy, significantly dampened CMV-specific immune responses. This was demonstrated by decreased IFN-γ production by CMV-specific T-cells (CMV peptide pool: p = 0.036; OR = 0.065 [95% CI: 0.005-0.840], pp65 antigen: p = 0.026; OR = 0.048 [95% CI: 0.003-0.699]). Increased immunosuppression correlated with reduced IFN-γ secretion per cell, reflected in smaller mean spot sizes for the CMV peptide pool (p = 0.019). Notably, shorter post-transplant intervals correlated with diminished antiviral T-cell IFN-γ release at two years (CMV peptide pool: p = 0.019; IE antigen: p = 0.010) and five years (CMV peptide pool: p = 0.0001; IE antigen: p = 0.002; pp65 antigen: p = 0.047), as did advancing age (pp65 antigen: p = 0.016, OR = 0.932, 95% CI: 0.881-0.987). Patients with undetectable CMV antigens had a notably higher risk of CMV reactivation within six months from blood collection, closely linked with triple immunosuppression and prednisolone use. These findings highlight the intricate interplay between immunosuppression, immune response dynamics, and CMV reactivation risk, emphasizing the necessity for tailored immunosuppressive strategies to mitigate CMV reactivation in liver-transplant recipients. It can be concluded that, particularly in the early months post-transplantation, the use of prednisolone as a third immunosuppressant should be critically reconsidered. Additionally, the use of prophylactic antiviral therapy effective against CMV in this context holds significant importance.
Collapse
Affiliation(s)
- Ann-Kristin Traska
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Münster, Germany; (A.-K.T.); (R.V.); (F.R.); (J.A.M.); (T.S.); (S.N.R.G.); (J.F.); (J.T.)
| | - Tobias Max Nowacki
- Department of Internal Medicine and Gastroenterology, Marienhospital Steinfurt, 48565 Steinfurt, Germany;
| | - Richard Vollenberg
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Münster, Germany; (A.-K.T.); (R.V.); (F.R.); (J.A.M.); (T.S.); (S.N.R.G.); (J.F.); (J.T.)
| | - Florian Rennebaum
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Münster, Germany; (A.-K.T.); (R.V.); (F.R.); (J.A.M.); (T.S.); (S.N.R.G.); (J.F.); (J.T.)
| | - Jörn Arne Meier
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Münster, Germany; (A.-K.T.); (R.V.); (F.R.); (J.A.M.); (T.S.); (S.N.R.G.); (J.F.); (J.T.)
| | - Tina Schomacher
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Münster, Germany; (A.-K.T.); (R.V.); (F.R.); (J.A.M.); (T.S.); (S.N.R.G.); (J.F.); (J.T.)
| | - Sara Noemi Reinartz Groba
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Münster, Germany; (A.-K.T.); (R.V.); (F.R.); (J.A.M.); (T.S.); (S.N.R.G.); (J.F.); (J.T.)
| | - Julia Fischer
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Münster, Germany; (A.-K.T.); (R.V.); (F.R.); (J.A.M.); (T.S.); (S.N.R.G.); (J.F.); (J.T.)
| | - Jonel Trebicka
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Münster, Germany; (A.-K.T.); (R.V.); (F.R.); (J.A.M.); (T.S.); (S.N.R.G.); (J.F.); (J.T.)
| | - Phil-Robin Tepasse
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Münster, Germany; (A.-K.T.); (R.V.); (F.R.); (J.A.M.); (T.S.); (S.N.R.G.); (J.F.); (J.T.)
| |
Collapse
|
4
|
Clutton GT, Weideman AMK, Mischell MA, Kallon S, Conrad SZ, Shaw FR, Warren JA, Lin L, Kuruc JD, Xu Y, Gay CM, Armistead PM, G. Hudgens M, Goonetilleke NP. CD3 downregulation identifies high-avidity human CD8 T cells. Clin Exp Immunol 2024; 215:279-290. [PMID: 37950348 PMCID: PMC10876116 DOI: 10.1093/cei/uxad124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/22/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023] Open
Abstract
CD8 T cells recognize infected and cancerous cells via their T-cell receptor (TCR), which binds peptide-MHC complexes on the target cell. The affinity of the interaction between the TCR and peptide-MHC contributes to the antigen sensitivity, or functional avidity, of the CD8 T cell. In response to peptide-MHC stimulation, the TCR-CD3 complex and CD8 co-receptor are downmodulated. We quantified CD3 and CD8 downmodulation following stimulation of human CD8 T cells with CMV, EBV, and HIV peptides spanning eight MHC restrictions, observing a strong correlation between the levels of CD3 and CD8 downmodulation and functional avidity, regardless of peptide viral origin. In TCR-transduced T cells targeting a tumor-associated antigen, changes in TCR-peptide affinity were sufficient to modify CD3 and CD8 downmodulation. Correlation analysis and generalized linear modeling indicated that CD3 downmodulation was the stronger correlate of avidity. CD3 downmodulation, simply measured using flow cytometry, can be used to identify high-avidity CD8 T cells in a clinical context.
Collapse
Affiliation(s)
- Genevieve T Clutton
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ann Marie K Weideman
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Melissa A Mischell
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sallay Kallon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shayla Z Conrad
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Fiona R Shaw
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joanna A Warren
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lin Lin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - JoAnn D Kuruc
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yinyan Xu
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cynthia M Gay
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Paul M Armistead
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael G. Hudgens
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nilu P Goonetilleke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
5
|
Janetzki S. Important Considerations for ELISpot Validation. Methods Mol Biol 2024; 2768:1-13. [PMID: 38502384 DOI: 10.1007/978-1-0716-3690-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
The ELISpot assay has a solid place in the immune monitoring field for over 40 years. It is an assay that can assess the function of single immune cells in a straightforward and easy-to-learn approach. Its use in basic research, translational, and clinical work has been documented in countless publications. Harmonization guidelines and invaluable tools for optimal assay performance and evaluation exist. However, the validation of an established ELISpot protocol has been left to diverse opinions about how to interpret and tackle typical validation parameters. This chapter addresses important considerations for ELISpot validation, including the interpretations of validation parameters for a meaningful description of assay performance.
Collapse
|
6
|
Mistretta B, Rankothgedera S, Castillo M, Rao M, Holloway K, Bhardwaj A, El Noafal M, Albarracin C, El-Zein R, Rezaei H, Su X, Akbani R, Shao XM, Czerniecki BJ, Karchin R, Bedrosian I, Gunaratne PH. Chimeric RNAs reveal putative neoantigen peptides for developing tumor vaccines for breast cancer. Front Immunol 2023; 14:1188831. [PMID: 37744342 PMCID: PMC10512078 DOI: 10.3389/fimmu.2023.1188831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/27/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction We present here a strategy to identify immunogenic neoantigen candidates from unique amino acid sequences at the junctions of fusion proteins which can serve as targets in the development of tumor vaccines for the treatment of breastcancer. Method We mined the sequence reads of breast tumor tissue that are usually discarded as discordant paired-end reads and discovered cancer specific fusion transcripts using tissue from cancer free controls as reference. Binding affinity predictions of novel peptide sequences crossing the fusion junction were analyzed by the MHC Class I binding predictor, MHCnuggets. CD8+ T cell responses against the 15 peptides were assessed through in vitro Enzyme Linked Immunospot (ELISpot). Results We uncovered 20 novel fusion transcripts from 75 breast tumors of 3 subtypes: TNBC, HER2+, and HR+. Of these, the NSFP1-LRRC37A2 fusion transcript was selected for further study. The 3833 bp chimeric RNA predicted by the consensus fusion junction sequence is consistent with a read-through transcription of the 5'-gene NSFP1-Pseudo gene NSFP1 (NSFtruncation at exon 12/13) followed by trans-splicing to connect withLRRC37A2 located immediately 3' through exon 1/2. A total of 15 different 8-mer neoantigen peptides discovered from the NSFP1 and LRRC37A2 truncations were predicted to bind to a total of 35 unique MHC class I alleles with a binding affinity of IC50<500nM.); 1 of which elicited a robust immune response. Conclusion Our data provides a framework to identify immunogenic neoantigen candidates from fusion transcripts and suggests a potential vaccine strategy to target the immunogenic neopeptides in patients with tumors carrying the NSFP1-LRRC37A2 fusion.
Collapse
Affiliation(s)
- Brandon Mistretta
- Department of Biology & Biochemistry, University of Houston, Houston, TX, United States
| | - Sakuni Rankothgedera
- Department of Biology & Biochemistry, University of Houston, Houston, TX, United States
| | - Micah Castillo
- Department of Biology & Biochemistry, University of Houston, Houston, TX, United States
| | - Mitchell Rao
- Department of Biology & Biochemistry, University of Houston, Houston, TX, United States
| | - Kimberly Holloway
- Department of Biology & Biochemistry, University of Houston, Houston, TX, United States
| | - Anjana Bhardwaj
- Department of Breast Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Maha El Noafal
- Department of Medicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Constance Albarracin
- Department of Pathology, The UT MD Anderson Cancer Center, Houston, TX, United States
| | - Randa El-Zein
- Department of Medicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Hengameh Rezaei
- Department of Biology & Biochemistry, University of Houston, Houston, TX, United States
| | - Xiaoping Su
- Department of Bioinformatics & Computational Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Rehan Akbani
- Department of Bioinformatics & Computational Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Xiaoshan M. Shao
- Biomedical Engineering Department, Institute for Computational Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Brian J. Czerniecki
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Rachel Karchin
- Biomedical Engineering Department, Institute for Computational Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Isabelle Bedrosian
- Department of Breast Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Preethi H. Gunaratne
- Department of Biology & Biochemistry, University of Houston, Houston, TX, United States
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, United States
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
7
|
Gorovits B, Azadeh M, Buchlis G, Fiscella M, Harrison T, Havert M, Janetzki S, Jawa V, Long B, Mahnke YD, McDermott A, Milton M, Nelson R, Vettermann C, Wu B. Evaluation of Cellular Immune Response to Adeno-Associated Virus-Based Gene Therapy. AAPS J 2023; 25:47. [PMID: 37101079 PMCID: PMC10132926 DOI: 10.1208/s12248-023-00814-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/14/2023] [Indexed: 04/28/2023] Open
Abstract
The number of approved or investigational late phase viral vector gene therapies (GTx) has been rapidly growing. The adeno-associated virus vector (AAV) technology continues to be the most used GTx platform of choice. The presence of pre-existing anti-AAV immunity has been firmly established and is broadly viewed as a potential deterrent for successful AAV transduction with a possibility of negative impact on clinical efficacy and a connection to adverse events. Recommendations for the evaluation of humoral, including neutralizing and total antibody based, anti-AAV immune response have been presented elsewhere. This manuscript aims to cover considerations related to the assessment of anti-AAV cellular immune response, including review of correlations between humoral and cellular responses, potential value of cellular immunogenicity assessment, and commonly used analytical methodologies and parameters critical for monitoring assay performance. This manuscript was authored by a group of scientists involved in GTx development who represent several pharma and contract research organizations. It is our intent to provide recommendations and guidance to the industry sponsors, academic laboratories, and regulatory agencies working on AAV-based GTx viral vector modalities with the goal of achieving a more consistent approach to anti-AAV cellular immune response assessment.
Collapse
Affiliation(s)
| | - Mitra Azadeh
- Ultragenyx Pharmaceutical Inc, Novato, California, USA
| | - George Buchlis
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Mike Havert
- Gene Therapy Partners, San Diego, California, USA
| | | | - Vibha Jawa
- Bristol Myers Squibb Pharmaceutical, Princeton, New Jersey, USA
| | - Brian Long
- BioMarin Pharmaceutical Inc, Novato, California, USA
| | | | - Andrew McDermott
- Labcorp Early Development Laboratories Inc, Indianapolis, Indiana, USA
| | - Mark Milton
- Lake Boon Pharmaceutical Consulting LLC, Hudson, New York, USA
| | | | | | - Bonnie Wu
- Janssen Pharmaceuticals, Raritan, New Jersey, USA
| |
Collapse
|
8
|
Enhanced In Vitro and In Vivo Potency of a T Cell Epitope in the Ebola Virus Glycoprotein Following Amino Acid Replacement at HLA-A*02:01 Binding Positions. J Virol 2022; 96:e0116621. [PMID: 36069549 PMCID: PMC9517714 DOI: 10.1128/jvi.01166-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Studies on Ebola virus disease (EVD) survivors and clinical studies on Ebola virus (EBOV) vaccine candidates have pinpointed the importance of a strong antibody response in protection and survival from EBOV infection. However, little is known about the T cell responses to EBOV or EBOV vaccines. We used HLA-A*02:01 (HLA-A2) transgenic mice to study HLA-A2-specific T cell responses elicited following vaccination with EBOV glycoprotein (EBOV-GP) presented with three different systems: (i) recombinant protein (rEBOV-GP), (ii) vesicular stomatitis replication-competent recombinant virus (VSV-EBOV-GP), and (iii) modified vaccinia Ankara virus recombinant (MVA-EBOV-GP). T cells from immunized animals were analyzed using peptide pools representing the entire GP region and individual peptides. Regardless of the vaccine formulation, we identified a minimal 9mer epitope containing an HLA-A2 motif (FLDPATTS), which was confirmed through HLA-A2 binding affinity and immunization studies. Using binding prediction software, we identified substitutions surrounding position 9 (S9V, P10V, and Q11V) that predicted enhanced binding to the HLA-A2 molecule. This enhanced binding was confirmed through in vitro binding studies and enhanced potency was shown with in vivo immunization studies using the enhanced sequences and the wild-type sequence. Of note, in silico studies predicted the enhanced 9mer epitope carrying the S9V substitution as the best overall HLA-A2 epitope for the full-length EBOV-GP. These results suggest that EBOV-GP-S9V and EBOV-GP-P10V represent more potent in vivo immunogens. Identification and enhancement of EBOV-specific human HLA epitopes could lead to the development of tools and reagents to induce more robust T cell responses in human subjects. IMPORTANCE Vaccine efficacy and immunity to viral infection are often measured by neutralizing antibody titers. T cells are specialized subsets of immune cells with antiviral activity, but this response is variable and difficult to track. We showed that the HLA-A2-specific T cell response to the Ebola virus glycoprotein can be enhanced significantly by a single residue substitution designed to improve an epitope binding affinity to one of the most frequent MHC alleles in the human population. This strategy could be applied to improve T cell responses to Ebola vaccines designed to elicit antibodies and adapted to target MHC alleles of populations in regions where endemic infections, like Ebola virus disease, are still causing outbreaks with concerning pandemic potential.
Collapse
|
9
|
Vavolizza RD, Petroni GR, Mauldin IS, Chianese-Bullock KA, Olson WC, Smith KT, Dengel LT, Haden K, Grosh WW, Kaur V, Varhegyi N, Gaughan EM, Slingluff CL. Phase I/II clinical trial of a helper peptide vaccine plus PD-1 blockade in PD-1 antibody-naïve and PD-1 antibody-experienced patients with melanoma (MEL64). J Immunother Cancer 2022; 10:e005424. [PMID: 36100309 PMCID: PMC9472210 DOI: 10.1136/jitc-2022-005424] [Citation(s) in RCA: 9] [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] [Accepted: 08/15/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND A vaccine containing 6 melanoma-associated peptides to stimulate helper T cells (6MHP) is safe, immunogenic, and clinically active. A phase I/II trial was designed to evaluate safety and immunogenicity of 6MHP vaccines plus programmed death 1 (PD-1) blockade. PARTICIPANTS AND METHODS Participants with advanced melanoma received 6MHP vaccines in an incomplete Freund's adjuvant (6 vaccines over 12 weeks). Pembrolizumab was administered intravenously every 3 weeks. Tumor biopsies at baseline and day 22 were analyzed by multiplex immunohistochemistry. Primary end points were safety (Common Terminology Criteria for Adverse Events V.4.03) and immunogenicity (ex vivo interferon-γ ELISpot assay). Additional end points included changes in the tumor microenvironment (TME) and clinical outcomes. RESULTS Twenty-two eligible participants were treated: 6 naïve to PD-1 antibody (Ab) and 16 PD-1 Ab-experienced. Median follow-up was 24.4 months. Most common treatment-related adverse events (any grade) included injection site reactions, fatigue, anemia, lymphopenia, fever, elevated aspartate aminotransferase, pruritus, and rash. Treatment-related dose-limiting toxicities were observed in 3 (14%) participants, which did not cross the study safety bound. A high durable T cell response (Rsp) to 6MHP was detected in only one participant, but twofold T cell Rsps to 6MHP were detected in 7/22 (32%; 90% CI (16% to 52%)) by week 13. Objective clinical responses were observed in 23% (1 complete response, 4 partial responses), including 4/6 PD-1 Ab-naïve (67%) and 1/16 PD-1 Ab-experienced (6%). Overall survival (OS) was longer for PD-1 Ab-naïve than Ab-experienced participants (HR 6.3 (90% CI (2.1 to 28.7)). In landmark analyses at 13 weeks, OS was also longer for those with T cell Rsps (HR 6.5 (90% CI (2.1 to 29.2)) and for those with objective clinical responses. TME evaluation revealed increased densities of CD8+ T cells, CD20+ B cells, and Tbet+ cells by day 22. CONCLUSIONS Treatment with the 6MHP vaccine plus pembrolizumab was safe, increased intratumoral lymphocytes, and induced T cell Rsps associated with prolonged OS. The low T cell Rsp rate in PD-1 Ab-experienced participants corroborates prior murine studies that caution against delaying cancer vaccines until after PD-1 blockade. The promising objective response rate and OS in PD-1 Ab-naïve participants support consideration of a larger study in that setting.
Collapse
Affiliation(s)
- Rick Daniel Vavolizza
- Department of Surgery, University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Gina R Petroni
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Ileana S Mauldin
- Department of Surgery, University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | | | - Walter C Olson
- Department of Surgery, University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Kelly T Smith
- Cancer Center and Office of Research Core Administration, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Lynn T Dengel
- Department of Surgery, University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Kathleen Haden
- Department of Surgery, University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - William W Grosh
- Department of Medicine, Division of Hematology/Oncology University of Virginia, Charlottesville, Virginia, USA
| | - Varinder Kaur
- Department of Medicine, Division of Hematology/Oncology University of Virginia, Charlottesville, Virginia, USA
| | - Nikole Varhegyi
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Elizabeth M Gaughan
- Department of Medicine, Division of Hematology/Oncology University of Virginia, Charlottesville, Virginia, USA
| | - Craig L Slingluff
- Department of Surgery, University of Virginia Cancer Center, Charlottesville, Virginia, USA
| |
Collapse
|
10
|
Kramer KJ, Wilfong EM, Voss K, Barone SM, Shiakolas AR, Raju N, Roe CE, Suryadevara N, Walker LM, Wall SC, Paulo A, Schaefer S, Dahunsi D, Westlake CS, Crowe JE, Carnahan RH, Rathmell JC, Bonami RH, Georgiev IS, Irish JM. Single-cell profiling of the antigen-specific response to BNT162b2 SARS-CoV-2 RNA vaccine. Nat Commun 2022; 13:3466. [PMID: 35710908 PMCID: PMC9201272 DOI: 10.1038/s41467-022-31142-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 05/26/2022] [Indexed: 12/15/2022] Open
Abstract
RNA-based vaccines against SARS-CoV-2 have proven critical to limiting COVID-19 disease severity and spread. Cellular mechanisms driving antigen-specific responses to these vaccines, however, remain uncertain. Here we identify and characterize antigen-specific cells and antibody responses to the RNA vaccine BNT162b2 using multiple single-cell technologies for in depth analysis of longitudinal samples from a cohort of healthy participants. Mass cytometry and unbiased machine learning pinpoint an expanding, population of antigen-specific memory CD4+ and CD8+ T cells with characteristics of follicular or peripheral helper cells. B cell receptor sequencing suggest progression from IgM, with apparent cross-reactivity to endemic coronaviruses, to SARS-CoV-2-specific IgA and IgG memory B cells and plasmablasts. Responding lymphocyte populations correlate with eventual SARS-CoV-2 IgG, and a participant lacking these cell populations failed to sustain SARS-CoV-2-specific antibodies and experienced breakthrough infection. These integrated proteomic and genomic platforms identify an antigen-specific cellular basis of RNA vaccine-based immunity.
Collapse
Affiliation(s)
- Kevin J Kramer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Vaccine Center, Nashville, TN, 37232, USA
| | - Erin M Wilfong
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Human Immunology Discovery Initiative of the Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, 37232, USA
| | - Kelsey Voss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sierra M Barone
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Andrea R Shiakolas
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Vaccine Center, Nashville, TN, 37232, USA
| | - Nagarajan Raju
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Vaccine Center, Nashville, TN, 37232, USA
| | - Caroline E Roe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | | | - Lauren M Walker
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Vaccine Center, Nashville, TN, 37232, USA
| | - Steven C Wall
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Vaccine Center, Nashville, TN, 37232, USA
| | - Ariana Paulo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Vaccine Center, Nashville, TN, 37232, USA
| | - Samuel Schaefer
- Human Immunology Discovery Initiative of the Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA
| | - Debolanle Dahunsi
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Human Immunology Discovery Initiative of the Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA
| | - Camille S Westlake
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Nashville, TN, 37232, USA
- Human Immunology Discovery Initiative of the Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Program in Computational Microbiology and Immunology, Nashville, TN, 37232, USA
| | | | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Human Immunology Discovery Initiative of the Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA.
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, 37232, USA.
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Program in Computational Microbiology and Immunology, Nashville, TN, 37232, USA.
| | - Rachel H Bonami
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Human Immunology Discovery Initiative of the Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA.
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, 37232, USA.
- Vanderbilt Program in Computational Microbiology and Immunology, Nashville, TN, 37232, USA.
| | - Ivelin S Georgiev
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Vaccine Center, Nashville, TN, 37232, USA.
- Human Immunology Discovery Initiative of the Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA.
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, 37232, USA.
- Vanderbilt Program in Computational Microbiology and Immunology, Nashville, TN, 37232, USA.
| | - Jonathan M Irish
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Human Immunology Discovery Initiative of the Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA.
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, 37232, USA.
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Program in Computational Microbiology and Immunology, Nashville, TN, 37232, USA.
| |
Collapse
|
11
|
Control of Tumors by Antigen-Specific CD8 + T Cells through PDL1-Targeted Delivery of Antigenic Peptide. J Immunol Res 2022; 2022:9054569. [PMID: 35028321 PMCID: PMC8752305 DOI: 10.1155/2022/9054569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
Tumor antigen-specific T cell function is limited by immune tolerance in the tumor microenvironment. In the tumor microenvironment, tumor cells upregulate PD-L1 expression to promote T cell exhaustion by PD-1/PD-L1 interactions and undergo mutations to avoid being targeted by tumor antigen-specific T cells. Thus, tumor cells escape the immune surveillance by causing immune tolerance. We reason that a chimeric molecule made of a PD-L1-specific antibody linked to a cleavable antigenic peptide can target the antigenic peptide to the tumor microenvironment, resulting in the blockade of the PD-1/PD-L1 pathway and killing tumor cells through the coating of antigenic peptide. Here, we have generated a therapeutic chimeric protein containing the PD-L1 single-chain variable fragment (scFv) linked to a cleavable model cytotoxic T lymphocyte (CTL) epitope: E7 CTL peptide. Our study demonstrated that our chimeric protein (named PDL1-scFv-Fc-RE7) can target PD-L1-expressing tumor cells and enable E7 presentation by releasing cleavable E7 CTL peptide to coat tumor cells, resulting in tumor clearance by E7-specific CD8+ T cells. The presentation of the E7 peptide by cancer cells can then render tumor cells susceptible to the killing of preexisting E7-specific CD8+ T cells and contribute to tumor clearance. Our finding suggests a synergistic approach to not only enhance antigen-specific tumor clearance but also bypass immune tolerance.
Collapse
|
12
|
Dynamics of spike-and nucleocapsid specific immunity during long-term follow-up and vaccination of SARS-CoV-2 convalescents. Nat Commun 2022; 13:153. [PMID: 35013191 PMCID: PMC8748966 DOI: 10.1038/s41467-021-27649-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/02/2021] [Indexed: 12/23/2022] Open
Abstract
Anti-viral immunity continuously declines over time after SARS-CoV-2 infection. Here, we characterize the dynamics of anti-viral immunity during long-term follow-up and after BNT162b2 mRNA-vaccination in convalescents after asymptomatic or mild SARS-CoV-2 infection. Virus-specific and virus-neutralizing antibody titers rapidly declined in convalescents over 9 months after infection, whereas virus-specific cytokine-producing polyfunctional T cells persisted, among which IL-2-producing T cells correlated with virus-neutralizing antibody titers. Among convalescents, 5% of individuals failed to mount long-lasting immunity after infection and showed a delayed response to vaccination compared to 1% of naïve vaccinees, but successfully responded to prime/boost vaccination. During the follow-up period, 8% of convalescents showed a selective increase in virus-neutralizing antibody titers without accompanying increased frequencies of circulating SARS-CoV-2-specific T cells. The same convalescents, however, responded to vaccination with simultaneous increase in antibody and T cell immunity revealing the strength of mRNA-vaccination to increase virus-specific immunity in convalescents. Waning immunity to SARS-CoV-2 is of concern. Here the authors follow spike- and nucleocapsid specific immunity in convalescent individuals for 9 months observing a decline in antibody levels but persisting T cell response. Vaccination approximately 11 months after infection boosts antibody and T cell immunity.
Collapse
|
13
|
Abstract
The enzyme-linked immunospot (ELISpot) is a highly sensitive immunoassay that measures the frequency of cytokine-secreting cells at the single-cell level. The secreted molecules are detected by using a detection antibody system similar to that used in the enzyme-linked immunosorbent assay (ELISA). The ELISpot assay is carried out in a 96-well plate and an automated ELISpot reader is used for analysis. The assay is easy to perform, robust and allows rapid analysis of a large number of samples and is not limited to measurement of cytokines; it is suitable for almost any secreted protein where single-cell analysis is of interest.
Collapse
|
14
|
Ducret A, Ackaert C, Bessa J, Bunce C, Hickling T, Jawa V, Kroenke MA, Lamberth K, Manin A, Penny HL, Smith N, Terszowski G, Tourdot S, Spindeldreher S. Assay format diversity in pre-clinical immunogenicity risk assessment: Toward a possible harmonization of antigenicity assays. MAbs 2021; 14:1993522. [PMID: 34923896 PMCID: PMC8726688 DOI: 10.1080/19420862.2021.1993522] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
A major impediment to successful use of therapeutic protein drugs is their ability to induce anti-drug antibodies (ADA) that can alter treatment efficacy and safety in a significant number of patients. To this aim, in silico, in vitro, and in vivo tools have been developed to assess sequence and other liabilities contributing to ADA development at different stages of the immune response. However, variability exists between similar assays developed by different investigators due to the complexity of assays, a degree of uncertainty about the underlying science, and their intended use. The impact of protocol variations on the outcome of the assays, i.e., on the immunogenicity risk assigned to a given drug candidate, cannot always be precisely assessed. Here, the Non-Clinical Immunogenicity Risk Assessment working group of the European Immunogenicity Platform (EIP) reviews currently used assays and protocols and discusses feasibility and next steps toward harmonization and standardization.
Collapse
Affiliation(s)
- Axel Ducret
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Chloé Ackaert
- ImmunXperts SA (A Nexelis Group Company), Gosselies, Belgium
| | - Juliana Bessa
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | - Timothy Hickling
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Vibha Jawa
- Biotherapeutics and Bioanalysis Non-Clinical Development, Bristol Myers Squibb, Princeton, NJ, USA
| | - Mark A Kroenke
- Clinical Immunology-Translational Medicine, Amgen Inc, Thousand Oaks, CA, USA
| | - Kasper Lamberth
- Analysis & Characterisation, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | - Anaïs Manin
- Abzena, Babraham Research Campus, Cambridge, UK
| | - Hweixian L Penny
- Clinical Immunology-Translational Medicine, Amgen Inc, Thousand Oaks, CA, USA
| | - Noel Smith
- Lonza Biologics, Chesterford Research Park, Saffron Walden, UK
| | - Grzegorz Terszowski
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | |
Collapse
|
15
|
Abstract
The FluoroSpot assay is a development of the highly sensitive enzyme-linked immunospot (ELISpot) assay which enables functional measurement of immunity at the single-cell level. Both assays are performed in a 96-well format and measures the frequency of analyte-secreting cells, in ELISpot usually limited to one analyte per well due to the use of enzymes and precipitating substrates for detection. FluoroSpot, performed in a similar way as ELISpot, overcomes this limitation by detecting each analyte with an assigned fluorophore instead of an enzyme. By using readers equipped with fluorophore-specific filters, cells producing single or multiple cytokines can be identified simultaneously in the same well. This greatly facilitates the analysis of functionally distinct subpopulations in heterogenous cell samples, for example, the frequency of polyfunctional T cells, suggested to be of importance in various disease states. FluoroSpot maintains the simplicity and sensitivity of the ELISpot while taking the assay a step further towards a multiplex analysis and an in-depth understanding of the quality of an immune response. We describe here a 96-well plate method to analyze cells that have secreted up to four different cytokines simultaneously (Four-color Fluorospot).
Collapse
|
16
|
Patel SP, Petroni GR, Roszik J, Olson WC, Wages NA, Chianese-Bullock KA, Smolkin M, Varhegyi N, Gaughan E, Smith KT, Haden K, Hall EH, Gnjatic S, Hwu P, Slingluff CL. Phase I/II trial of a long peptide vaccine (LPV7) plus toll-like receptor (TLR) agonists with or without incomplete Freund's adjuvant (IFA) for resected high-risk melanoma. J Immunother Cancer 2021; 9:e003220. [PMID: 34413169 PMCID: PMC8378357 DOI: 10.1136/jitc-2021-003220] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND We performed a clinical trial to evaluate safety and immunogenicity of a novel long peptide vaccine administered in combinations of incomplete Freund's adjuvant (IFA) and agonists for TLR3 (polyICLC) and TLR7/8 (resiquimod). We hypothesized that T cell responses to minimal epitope peptides (MEPs) within the long peptides would be enhanced compared with prior vaccines with MEP themselves and that T cell responses would be enhanced with TLR agonists, compared with IFA alone. METHODS Participants with resected stage IIB-IV melanoma were vaccinated with seven long melanoma peptides (LPV7) from tyrosinase, gp100, MAGE-A1, MAGE-A10, and NY-ESO-1, each containing a known MEP for CD8+ T cells, plus a tetanus helper peptide (Tet) restricted by Class II MHC. Enrollment was guided by an adaptive design to one of seven adjuvant combinations. Vaccines were administered at weeks 1, 2, 3, 6, 9, 12 at rotating injection sites. T cell and IgG antibody (Ab) responses were measured with IFN-gamma ELIspot assay ex vivo and ELISA, respectively. RESULTS Fifty eligible participants were assigned to seven study groups, with highest enrollment on arm E (LPV7+Tet+IFA+polyICLC). There was one dose-limiting toxicity (DLT) in Group E (grade 3 injection site reaction, 6% DLT rate). All other treatment-related adverse events were grades 1-2. The CD8+ T cell immune response rate (IRR) to MEPs was 18%, less than in prior studies using MEP vaccines in IFA. The CD8+ T cell IRR trended higher for IFA-containing adjuvants (24%) than adjuvants containing only TLR agonists (6%). Overall T cell IRR to full-length LPV7 was 30%; CD4+ T cell IRR to Tet was 40%, and serum Ab IRR to LPV7 was 84%. These IRRs also trended higher for IFA-containing adjuvants (36% vs 18%, 48% vs 24%, and 97% vs 60%, respectively). CONCLUSIONS The LPV7 vaccine is safe with each of seven adjuvant strategies and induced T cell responses to CD8 MEPs ex vivo in a subset of patients but did not enhance IRRs compared with prior vaccines using short peptides. Immunogenicity was supported more by IFA than by TLR agonists alone and may be enhanced by polyICLC plus IFA. TRIAL REGISTRATION NUMBER NCT02126579.
Collapse
Affiliation(s)
- Sapna P Patel
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gina R Petroni
- University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jason Roszik
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Walter C Olson
- University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Nolan A Wages
- Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | - Mark Smolkin
- Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Nikole Varhegyi
- Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Elizabeth Gaughan
- University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kelly T Smith
- University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kathleen Haden
- University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Emily H Hall
- University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Craig L Slingluff
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
17
|
Kramer KJ, Wilfong EM, Voss K, Barone SM, Shiakolas AR, Raju N, Roe CE, Suryadevara N, Walker L, Wall SC, Paulo A, Schaefer S, Dahunsi D, Westlake CS, Crowe JE, Carnahan RH, Rathmell JC, Bonami RH, Georgiev IS, Irish JM. Single-Cell Profiling of the Antigen-Specific Response to BNT162b2 SARS-CoV-2 RNA Vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.07.28.453981. [PMID: 34341788 PMCID: PMC8328055 DOI: 10.1101/2021.07.28.453981] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RNA-based vaccines against SARS-CoV-2 are critical to limiting COVID-19 severity and spread. Cellular mechanisms driving antigen-specific responses to these vaccines, however, remain uncertain. We used single-cell technologies to identify and characterized antigen-specific cells and antibody responses to the RNA vaccine BNT162b2 in longitudinal samples from a cohort of healthy donors. Mass cytometry and machine learning pinpointed a novel expanding, population of antigen-specific non-canonical memory CD4 + and CD8 + T cells. B cell sequencing suggested progression from IgM, with apparent cross-reactivity to endemic coronaviruses, to SARS-CoV-2-specific IgA and IgG memory B cells and plasmablasts. Responding lymphocyte populations correlated with eventual SARS-CoV-2 IgG and a donor lacking these cell populations failed to sustain SARS-CoV-2-specific antibodies and experienced breakthrough infection. These integrated proteomic and genomic platforms reveal an antigen-specific cellular basis of RNA vaccine-based immunity. ONE SENTENCE SUMMARY Single-cell profiling reveals the cellular basis of the antigen-specific response to the BNT162b2 SARS-CoV-2 RNA vaccine.
Collapse
|
18
|
Kang TH, Yang A, Tsai Y, Ferrall L, Hung CF. Targeted tumor coating with antigenic, CTL-recognizable peptides via Annexin A5 chimeric constructs following chemotherapy redirects adaptive CD8+ T cell immunity for tumor clearance. Cell Mol Immunol 2021; 18:1578-1580. [PMID: 33139904 PMCID: PMC8166831 DOI: 10.1038/s41423-020-00563-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- Tae Heung Kang
- Institute of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Andrew Yang
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Yachea Tsai
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Louise Ferrall
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
19
|
Taborska P, Strizova Z, Stakheev D, Sojka L, Bartunkova J, Smrz D. CD4 + T Cells of Prostate Cancer Patients Have Decreased Immune Responses to Antigens Derived From SARS-CoV-2 Spike Glycoprotein. Front Immunol 2021; 12:629102. [PMID: 34012431 PMCID: PMC8128251 DOI: 10.3389/fimmu.2021.629102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
The adaptive immune response to severe acute respiratory coronavirus 2 (SARS-CoV-2) is important for vaccine development and in the recovery from coronavirus disease 2019 (COVID-19). Men and cancer patients have been reported to be at higher risks of contracting the virus and developing the more severe forms of COVID-19. Prostate cancer (PCa) may be associated with both of these risks. We show that CD4+ T cells of SARS-CoV-2-unexposed patients with hormone-refractory (HR) metastatic PCa had decreased CD4+ T cell immune responses to antigens from SARS-CoV-2 spike glycoprotein but not from the spiked glycoprotein of the ‘common cold’-associated human coronavirus 229E (HCoV-229E) as compared with healthy male volunteers who responded comparably to both HCoV-229E- and SARS-CoV-2-derived antigens. Moreover, the HCoV-229E spike glycoprotein antigen-elicited CD4+ T cell immune responses cross-reacted with the SARS-CoV-2 spiked glycoprotein antigens. PCa patients may have impaired responses to the vaccination, and the cross-reactivity can mediate antibody-dependent enhancement (ADE) of COVID-19. These findings highlight the potential for increased vulnerability of PCa patients to COVID-19.
Collapse
Affiliation(s)
- Pavla Taborska
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Dmitry Stakheev
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Ludek Sojka
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia.,Department of Technical Operations, SOTIO, a.s., Prague, Czechia
| | - Jirina Bartunkova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Daniel Smrz
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| |
Collapse
|
20
|
Slingluff CL, Zarour HM, Tawbi HAH, Kirkwood JM, Postow MA, Friedlander P, Devoe CE, Gaughan EM, Mauldin IS, Olson WC, Smith KT, Macri MJ, Ricciardi T, Ryan A, Venhaus R, Wolchok JD. A phase 1 study of NY-ESO-1 vaccine + anti-CTLA4 antibody Ipilimumab (IPI) in patients with unresectable or metastatic melanoma. Oncoimmunology 2021; 10:1898105. [PMID: 33796406 PMCID: PMC8007150 DOI: 10.1080/2162402x.2021.1898105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Ipilimumab (IPI) can enhance immunity to the cancer-testis antigen NY-ESO-1. A clinical trial was designed to assess safety, immunogenicity, and clinical responses with IPI + NY-ESO-1 vaccines and effects on the tumor microenvironment (TME). Patients with measurable NY-ESO-1+ tumors were enrolled among three arms: A) IPI + NY-ESO-1 protein + poly-ICLC (pICLC) + incomplete Freund’s adjuvant (IFA); B) IPI + NY-ESO-1 overlapping long peptides (OLP) + pICLC + IFA; and C) IPI + NY-ESO-1 OLP + pICLC. Clinical responses were assessed by irRC. T cell and Ab responses were assessed by ex vivo IFN-gamma ELIspot and ELISA. Tumor biopsies pre- and post-treatment were evaluated for immune infiltrates. Eight patients were enrolled: 5, 2, and 1 in Arms A-C, respectively. There were no DLTs. Best clinical responses were SD (4) and PD (4). T-cell and antibody (Ab) responses to NY-ESO-1 were detected in 6 (75%) and 7 (88%) patients, respectively, and were associated with SD. The breadth of Ab responses was greater for patients with SD than PD (p = .036). For five patients evaluable in the TME, treatment was associated with increases in proliferating (Ki67+) CD8+ T cells and decreases in RORγt+ CD4+ T cells. T cell densities increased for those with SD. Detection of T cell responses to NY-ESO-1 ex vivo in most patients suggests that IPI may have enhanced those responses. Proliferating intratumoral CD8+ T cells increased after vaccination plus IPI suggesting favorable impact of IPI plus NY-ESO-1 vaccines on the TME. List of Abbreviations: Ab = antibody; CTCAE = NCI Common Terminology Criteria for Adverse Events; DHFR/DHRP = dihydrofolate reductase; DLT = Dose-limiting toxicity; ELISA = enzyme-linked immunosorbent assay; IFA = incomplete Freund’s adjuvant (Montanide ISA-51); IFNγ = Interferon gamma; IPI = Ipilimumab; irRC = immune-related response criteria; mIFH = multispectral immunofluorescence histology; OLP = NY-ESO-1 overlapping long peptides; PBMC = peripheral blood mononuclear cells; PD = Progressive disease; pICLC = poly-ICLC (Hiltonol), a TLR3/MDA-5 agonist; RLT = Regimen-limiting Toxicity; ROI = regions of interest; RT = room temperature; SAE = serious adverse event; SD = stable disease; TEAE = treatment-emergent adverse events; TLR = toll-like receptor; TME = tumor microenvironment; TRAE = treatment-related adverse events.
Collapse
Affiliation(s)
- Craig L Slingluff
- Department of Surgery/Division of Surgical Oncology, University of Virginia, Charlottesville, VA, USA
| | - Hassane M Zarour
- Division of Medical Oncology, Dept of Medicine and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hussein Abdul-Hassan Tawbi
- Division of Medical Oncology, Dept of Medicine and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX
| | - John M Kirkwood
- Division of Medical Oncology, Dept of Medicine and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael A Postow
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Philip Friedlander
- Department of Medicine, Hematology, and Medical Oncology, Mount Sinai Medical Center, New York, NY, USA
| | - Craig E Devoe
- Northwell Health Cancer Institute, Lake Success, NY, USA
| | - Elizabeth M Gaughan
- Department of Medicine/Division of Hematology Oncology, University of Virginia, Charlottesville, VA, USA
| | - Ileana S Mauldin
- Department of Surgery/Division of Surgical Oncology, University of Virginia, Charlottesville, VA, USA
| | - Walter C Olson
- Department of Surgery/Division of Surgical Oncology, University of Virginia, Charlottesville, VA, USA
| | - Kelly T Smith
- Department of Surgery/Division of Surgical Oncology, University of Virginia, Charlottesville, VA, USA
| | - Mary J Macri
- Ludwig Institute for Cancer Research, New York, NY, USA
| | | | - Aileen Ryan
- Ludwig Institute for Cancer Research, New York, NY, USA
| | - Ralph Venhaus
- Ludwig Institute for Cancer Research, New York, NY, USA
| | - Jedd D Wolchok
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center.,Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| |
Collapse
|
21
|
Macrophage Plasticity and Function in the Lung Tumour Microenvironment Revealed in 3D Heterotypic Spheroid and Explant Models. Biomedicines 2021; 9:biomedicines9030302. [PMID: 33804204 PMCID: PMC7999110 DOI: 10.3390/biomedicines9030302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 11/16/2022] Open
Abstract
In non-small cell lung cancer (NSCLC), stroma-resident and tumour-infiltrating macrophages may facilitate an immunosuppressive tumour microenvironment (TME) and hamper immunotherapeutic responses. Analysis of tumour-associated macrophage (TAM) plasticity in NSCLC is largely lacking. We established a novel, multi-marker, dual analysis approach for assessing monocyte-derived macrophage (Mφ) polarisation and M1/M2 phenotypic plasticity. We developed a flow cytometry-based, two-marker analysis (CD64 and CD206) of CD14+ cells. The phenotype and immune function of in vitro-induced TAMs was studied in a heterotypic spheroid and tumour-derived explant model of NSCLC. Heterotypic spheroids and NSCLC explants skewed Mφs from an M1- (CD206loCD64hi) to M2-like (CD206hiCD64lo) phenotype. Lipopolysaccharide (LPS) and IFNγ treatment reversed M2-like Mφ polarisation, indicating the plasticity of Mφs. Importantly, antigen-specific CD8+ T cell responses were reduced in the presence of tumour explant-conditioned Mφs, but not spheroid-conditioned Mφs, suggesting explants are likely a more relevant model of the immune TME than cell line-derived spheroids. Our data indicates the importance of multi-marker, functional analyses within Mφ subsets and the advantages of the ex vivo NSCLC explant model in immunomodulation studies. We highlight the plasticity of the M1/M2 phenotype using the explant model and provide a tool for studying therapeutic interventions designed to reprogram M2-like Mφ-induced immunosuppression.
Collapse
|
22
|
Langat RK, Farah B, Indangasi J, Ogola S, Omosa-Manyonyi G, Anzala O, Bizimana J, Tekirya E, Ngetsa C, Silwamba M, Muyanja E, Chetty P, Jangano M, Hills N, Gilmour J, Dally L, Cox JH, Hayes P. Performance of International AIDS Vaccine Initiative African clinical research laboratories in standardised ELISpot and peripheral blood mononuclear cell processing in support of HIV vaccine clinical trials. Afr J Lab Med 2021; 10:1056. [PMID: 33833946 PMCID: PMC8014752 DOI: 10.4102/ajlm.v10i1.1056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/23/2020] [Indexed: 11/28/2022] Open
Abstract
Background Standardisation of procedures for performing cellular functional assays across laboratories participating in multicentre clinical trials is key for generating comparable and reliable data. Objective This article describes the performance of accredited laboratories in Africa and Europe on testing done in support of clinical trials. Methods For enzyme-linked immunospot assay (ELISpot) proficiency, characterised peripheral blood mononuclear cells (PBMCs) obtained from 48 HIV-negative blood donors in Johannesburg, South Africa, were sent to participating laboratories between February 2010 and February 2014. The PBMCs were tested for responses against cytomegalovirus, Epstein Barr and influenza peptide pools in a total of 1751 assays. In a separate study, a total of 1297 PBMC samples isolated from healthy HIV-negative participants in clinical trials of two prophylactic HIV vaccine candidates in Kenya, Uganda, Rwanda and Zambia were analysed for cell viability, cell yield and cell recovery from frozen PBMCs. Results Most (99%) of the 1751 ELISpot proficiency assays had data within acceptable ranges with low responses to mock stimuli. No significant statistical difference were observed in ELISpot responses at the five laboratories actively conducting immunological analyses. Of the 1297 clinical trial PBMCs processed, 94% had cell viability above 90% and 96% had cell yield above 0.7 million per mL of blood in freshly isolated cells. All parameters were within the predefined acceptance criteria. Conclusion We demonstrate that multiple laboratories can generate reliable, accurate and comparable data by using standardised procedures, having regular training, having regular equipment maintenance and using centrally sourced reagents.
Collapse
Affiliation(s)
- Robert K Langat
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya.,International AIDS Vaccine Initiative (IAVI), Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Bashir Farah
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Jackton Indangasi
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Simon Ogola
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Gloria Omosa-Manyonyi
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Omu Anzala
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | | | | | - Caroline Ngetsa
- Kenya Medical Research Institute Centre for Geographical Medicine Research Coast, Mombasa, Kenya
| | | | - Enoch Muyanja
- Ugandan Virus Research Institute-IAVI, Entebbe, Uganda
| | - Paramesh Chetty
- International AIDS Vaccine Initiative, Johannesburg, South Africa
| | | | - Nancy Hills
- School of Medicine, University of California, San Francisco, California, United States
| | - Jill Gilmour
- International AIDS Vaccine Initiative (IAVI), Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Len Dally
- Emmes Corporation, Rockville, Maryland, United States
| | - Josephine H Cox
- Clinical Trials Program, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Peter Hayes
- International AIDS Vaccine Initiative (IAVI), Human Immunology Laboratory, Imperial College, London, United Kingdom
| |
Collapse
|
23
|
CERI, CEFX, and CPI: Largely Improved Positive Controls for Testing Antigen-Specific T Cell Function in PBMC Compared to CEF. Cells 2021; 10:cells10020248. [PMID: 33514016 PMCID: PMC7911306 DOI: 10.3390/cells10020248] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/21/2020] [Accepted: 01/14/2021] [Indexed: 01/17/2023] Open
Abstract
Monitoring antigen-specific T cell immunity relies on functional tests that require T cells and antigen presenting cells to be uncompromised. Drawing of blood, its storage and shipment from the clinical site to the test laboratory, and the subsequent isolation, cryopreservation and thawing of peripheral blood mononuclear cells (PBMCs) before the actual test is performed can introduce numerous variables that may jeopardize the results. Therefore, no T cell test is valid without assessing the functional fitness of the PBMC being utilized. This can only be accomplished through the inclusion of positive controls that actually evaluate the performance of the antigen-specific T cell and antigen presenting cell (APC) compartments. For Caucasians, CEF peptides have been commonly used to this extent. Moreover, CEF peptides only measure CD8 cell functionality. We introduce here universal CD8+ T cell positive controls without any racial bias, as well as positive controls for the CD4+ T cell and APC compartments. In summary, we offer new tools and strategies for the assessment of PBMC functional fitness required for reliable T cell immune monitoring.
Collapse
|
24
|
Lee MN, Meyerson M. Antigen identification for HLA class I- and HLA class II-restricted T cell receptors using cytokine-capturing antigen-presenting cells. Sci Immunol 2021; 6:6/55/eabf4001. [PMID: 33483338 DOI: 10.1126/sciimmunol.abf4001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/17/2020] [Indexed: 12/17/2022]
Abstract
A major limitation to understanding the associations of human leukocyte antigen (HLA) and CD8+ and CD4+ T cell receptor (TCR) genes with disease pathophysiology is the technological barrier of identifying which HLA molecules, epitopes, and TCRs form functional complexes. Here, we present a high-throughput epitope identification system that combines capture of T cell-secreted cytokines by barcoded antigen-presenting cells (APCs), cell sorting, and next-generation sequencing to identify class I- and class II-restricted epitopes starting from highly complex peptide-encoding oligonucleotide pools. We engineered APCs to express anti-cytokine antibodies, a library of DNA-encoded peptides, and multiple HLA class I or II molecules. We demonstrate that these engineered APCs link T cell activation-dependent cytokines with the DNA that encodes the presented peptide. We validated this technology by showing that we could select known targets of viral epitope-, neoepitope-, and autoimmune epitope-specific TCRs, starting from mixtures of peptide-encoding oligonucleotides. Then, starting from 10 TCRβ sequences that are found commonly in humans but lack known targets, we identified seven CD8+ or CD4+ TCR-targeted epitopes encoded by the human cytomegalovirus (CMV) genome. These included known epitopes, as well as a class I and a class II CMV epitope that have not been previously described. Thus, our cytokine capture-based assay makes use of a signal secreted by both CD8+ and CD4+ T cells and allows pooled screening of thousands of encoded peptides to enable epitope discovery for orphan TCRs. Our technology may enable identification of HLA-epitope-TCR complexes relevant to disease control, etiology, or treatment.
Collapse
Affiliation(s)
- Mark N Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. .,Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA.,Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. .,Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA.,Departments of Genetics and Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
25
|
Slingluff CL, Petroni GR, Chianese-Bullock KA, Wages NA, Olson WC, Smith KT, Haden K, Dengel LT, Dickinson A, Reed C, Gaughan EM, Grosh WW, Kaur V, Varhegyi N, Smolkin M, Galeassi NV, Deacon D, Hall EH. Trial to evaluate the immunogenicity and safety of a melanoma helper peptide vaccine plus incomplete Freund's adjuvant, cyclophosphamide, and polyICLC (Mel63). J Immunother Cancer 2021; 9:jitc-2020-000934. [PMID: 33479025 PMCID: PMC7825263 DOI: 10.1136/jitc-2020-000934] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2020] [Indexed: 12/17/2022] Open
Abstract
Background Peptide vaccines designed to stimulate melanoma-reactive CD4+ T cells can induce T cell and antibody (Ab) responses, associated with enhanced overall survival. We hypothesized that adding toll-like receptor 3 agonist polyICLC to an incomplete Freund’s adjuvant (IFA) would be safe and would support strong, durable CD4+ T cell and Ab responses. We also hypothesized that oral low-dose metronomic cyclophosphamide (mCy) would be safe, would reduce circulating regulatory T cells (T-regs) and would further enhance immunogenicity. Participants and methods An adaptive design based on toxicity and durable CD4+ T cell immune response (dRsp) was used to assign participants with resected stage IIA-IV melanoma to one of four study regimens. The regimens included a vaccine comprising six melanoma peptides restricted by Class II MHC (6MHP) in an emulsion with IFA alone (Arm A), with IFA plus systemic mCy (Arm B), with IFA+ local polyICLC (Arm C), or with IFA+ polyICLC+ mCy (Arm D). Toxicities were recorded (CTCAE V.4.03). T cell responses were measured by interferon γ ELIspot assay ex vivo. Serum Ab responses to 6MHP were measured by ELISA. Circulating T-regs were assessed by flow cytometry. Results Forty-eight eligible participants were enrolled and treated. Early data on safety and dRsp favored enrollment on arm D. Total enrollment on Arms A-D were 3, 7, 6, and 32, respectively. Treatment-related dose-limiting toxicities (DLTs) were observed in 1/7 (14%) participants on arm B and 2/32 (6%) on arm D. None exceeded the 25% DLT threshold for early closure to enrollment for any arm. Strong durable T cell responses to 6MHP were detected ex vivo in 0%, 29%, 67%, and 47% of participants on arms A-D, respectively. IgG Ab responses were greatest for arms C and D. Circulating T-regs frequencies were not altered by mCy. Conclusions 6MHP vaccines administered with IFA, polyICLC, and mCy were well tolerated. The dRsp rate for arm D of 47% (90% CI 32 to 63) exceeded the 18% (90% CI 11 to 26) rate previously observed with 6MHP in IFA alone. Vaccination with IFA+ polyICLC (arm C) also showed promise for enhancing T cell and Ab responses.
Collapse
Affiliation(s)
- Craig L Slingluff
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA .,University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Gina R Petroni
- University of Virginia Cancer Center, Charlottesville, Virginia, USA.,Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kimberly A Chianese-Bullock
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Nolan A Wages
- University of Virginia Cancer Center, Charlottesville, Virginia, USA.,Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Walter C Olson
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kelly T Smith
- Office of Research Cores Administration, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kathleen Haden
- University of Virginia Cancer Center, Charlottesville, Virginia, USA.,University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Lynn T Dengel
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Anna Dickinson
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Caroline Reed
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Elizabeth M Gaughan
- Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - William W Grosh
- Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Varinder Kaur
- Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Nikole Varhegyi
- Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mark Smolkin
- Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Nadejda V Galeassi
- Cardiovascular Imaging Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Donna Deacon
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Emily H Hall
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,University of Virginia Cancer Center, Charlottesville, Virginia, USA
| |
Collapse
|
26
|
Przybyla A, Lehmann AA, Zhang T, Mackiewicz J, Galus Ł, Kirchenbaum GA, Mackiewicz A, Lehmann PV. Functional T Cell Reactivity to Melanocyte Antigens Is Lost during the Progression of Malignant Melanoma, but Is Restored by Immunization. Cancers (Basel) 2021; 13:cancers13020223. [PMID: 33435427 PMCID: PMC7827050 DOI: 10.3390/cancers13020223] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Healthy humans develop spontaneous CD8+ T cell responses to melanoma associated antigens (MA) expressed by normal melanocytes. This natural autoimmunity directed against melanocytes might confer protection against the development of malignant melanoma (MM), where MA are overexpressed tumor-associated antigens. We report that functional T cell reactivity to MA is diminished in untreated MM patients. Three lines of evidence suggest that the MA-reactive T cells present in healthy subjects undergo exhaustion once MM establishes itself. First, only the MA-specific T cell reactivity was affected in the MM patients. Second, in these patients, the residual MA-specific T cells were functionally impaired, showing a diminished per cell IFN-γ productivity. Third, immunizations with allogeneic melanoma cells restored natural CD8+ T cell autoimmunity to MA. Abstract Healthy human subjects develop spontaneous CD8+ T cell responses to melanoma associated antigens (MA) expressed by normal melanocytes, such as Tyrosinase, MAGE-A3, Melan/Mart-1, gp100, and NY-ESO-1. This natural autoimmunity directed against melanocytes might confer protection against the development of malignant melanoma (MM), where MA are present as overexpressed tumor-associated antigens. Consistent with this notion we report here that functional T cell reactivity to MA was found to be significantly diminished to MAGE-A3, Melan-A/Mart-1, and gp100 in untreated MM patients. Three lines of evidence suggest that the MA-reactive T cells present in healthy subjects undergo exhaustion once MM establishes itself. First, only the MA-specific T cell reactivity was affected in the MM patients; that to third party recall antigens was not. Second, in these patients, the residual MA-specific T cells, unlike third party antigen reactive T cells, were functionally impaired, showing a diminished per cell IFN-γ productivity. Third, we show that immunization with MA restored natural CD8+ T cell autoimmunity to MA in 85% of the MM patients. The role of natural T cell autoimmunity to tumor-associated MA is discussed based on discrete levels of T cell activation thresholds.
Collapse
Affiliation(s)
- Anna Przybyla
- Research and Development Department, Cellular Technology Limited (CTL), Shaker Heights, OH 44122, USA; (A.P.); (A.A.L.); (T.Z.); (G.A.K.)
- Department of Cancer Immunology, Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznan, Poland;
| | - Alexander A. Lehmann
- Research and Development Department, Cellular Technology Limited (CTL), Shaker Heights, OH 44122, USA; (A.P.); (A.A.L.); (T.Z.); (G.A.K.)
| | - Ting Zhang
- Research and Development Department, Cellular Technology Limited (CTL), Shaker Heights, OH 44122, USA; (A.P.); (A.A.L.); (T.Z.); (G.A.K.)
| | - Jacek Mackiewicz
- Department of Medical and Experimental Oncology, University of Medical Sciences, 60-355 Poznan, Poland; (J.M.); (Ł.G.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Center, 61-866 Poznan, Poland
| | - Łukasz Galus
- Department of Medical and Experimental Oncology, University of Medical Sciences, 60-355 Poznan, Poland; (J.M.); (Ł.G.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Center, 61-866 Poznan, Poland
- Chemotherapy Department, Greater Poland Cancer Center, 61-866 Poznan, Poland
| | - Greg A. Kirchenbaum
- Research and Development Department, Cellular Technology Limited (CTL), Shaker Heights, OH 44122, USA; (A.P.); (A.A.L.); (T.Z.); (G.A.K.)
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznan, Poland;
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Center, 61-866 Poznan, Poland
| | - Paul V. Lehmann
- Research and Development Department, Cellular Technology Limited (CTL), Shaker Heights, OH 44122, USA; (A.P.); (A.A.L.); (T.Z.); (G.A.K.)
- Correspondence: ; Tel.: +1-216-965-6311
| |
Collapse
|
27
|
Abstract
The enzyme-linked immune absorbent spot (ELISpot) assay allows for the quantification of the number of cells producing a particular secreted analyte. As T lymphocytes secrete cytokines such as interferon (IFN)-γ upon binding of the T cell receptor with its cognate antigen epitope, IFN-γ ELISpot allows for the measurement of antigen-specific T cells in an immune sample. Immune cells are isolated from the vaccinated subject and incubated with the epitope/antigen of interest on polyvinylidene difluoride (PVDF)-lined microplates precoated with a capture antibody to IFN-γ. Cytokine spots are then detected utilizing an IFN-γ-specific detection antibody and an enzyme-linked conjugate. Here, we describe the quantification of OVA-specific CD8 and CD4 T cells from mouse splenocytes to measure vaccine-induced cellular responses.
Collapse
Affiliation(s)
- Bassel Akache
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Michael J McCluskie
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada.
| |
Collapse
|
28
|
Ferretti AP, Kula T, Wang Y, Nguyen DMV, Weinheimer A, Dunlap GS, Xu Q, Nabilsi N, Perullo CR, Cristofaro AW, Whitton HJ, Virbasius A, Olivier KJ, Buckner LR, Alistar AT, Whitman ED, Bertino SA, Chattopadhyay S, MacBeath G. Unbiased Screens Show CD8 + T Cells of COVID-19 Patients Recognize Shared Epitopes in SARS-CoV-2 that Largely Reside outside the Spike Protein. Immunity 2020; 53:1095-1107.e3. [PMID: 33128877 PMCID: PMC7574860 DOI: 10.1016/j.immuni.2020.10.006] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/03/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022]
Abstract
Developing effective strategies to prevent or treat coronavirus disease 2019 (COVID-19) requires understanding the natural immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We used an unbiased, genome-wide screening technology to determine the precise peptide sequences in SARS-CoV-2 that are recognized by the memory CD8+ T cells of COVID-19 patients. In total, we identified 3-8 epitopes for each of the 6 most prevalent human leukocyte antigen (HLA) types. These epitopes were broadly shared across patients and located in regions of the virus that are not subject to mutational variation. Notably, only 3 of the 29 shared epitopes were located in the spike protein, whereas most epitopes were located in ORF1ab or the nucleocapsid protein. We also found that CD8+ T cells generally do not cross-react with epitopes in the four seasonal coronaviruses that cause the common cold. Overall, these findings can inform development of next-generation vaccines that better recapitulate natural CD8+ T cell immunity to SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Yifan Wang
- TScan Therapeutics, Waltham, MA 02451, USA
| | | | | | | | - Qikai Xu
- TScan Therapeutics, Waltham, MA 02451, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Cebon JS, Gore M, Thompson JF, Davis ID, McArthur GA, Walpole E, Smithers M, Cerundolo V, Dunbar PR, MacGregor D, Fisher C, Millward M, Nathan P, Findlay MPN, Hersey P, Evans TRJ, Ottensmeier CH, Marsden J, Dalgleish AG, Corrie PG, Maria M, Brimble M, Williams G, Winkler S, Jackson HM, Endo-Munoz L, Tutuka CSA, Venhaus R, Old LJ, Haack D, Maraskovsky E, Behren A, Chen W. Results of a randomized, double-blind phase II clinical trial of NY-ESO-1 vaccine with ISCOMATRIX adjuvant versus ISCOMATRIX alone in participants with high-risk resected melanoma. J Immunother Cancer 2020; 8:e000410. [PMID: 32317292 PMCID: PMC7204806 DOI: 10.1136/jitc-2019-000410] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND To compare the clinical efficacy of New York Esophageal squamous cell carcinoma-1 (NY-ESO-1) vaccine with ISCOMATRIX adjuvant versus ISCOMATRIX alone in a randomized, double-blind phase II study in participants with fully resected melanoma at high risk of recurrence. METHODS Participants with resected stage IIc, IIIb, IIIc and IV melanoma expressing NY-ESO-1 were randomized to treatment with three doses of NY-ESO-1/ISCOMATRIX or ISCOMATRIX adjuvant administered intramuscularly at 4-week intervals, followed by a further dose at 6 months. Primary endpoint was the proportion free of relapse at 18 months in the intention-to-treat (ITT) population and two per-protocol populations. Secondary endpoints included relapse-free survival (RFS) and overall survival (OS), safety and NY-ESO-1 immunity. RESULTS The ITT population comprised 110 participants, with 56 randomized to NY-ESO-1/ISCOMATRIX and 54 to ISCOMATRIX alone. No significant toxicities were observed. There were no differences between the study arms in relapses at 18 months or for median time to relapse; 139 vs 176 days (p=0.296), or relapse rate, 27 (48.2%) vs 26 (48.1%) (HR 0.913; 95% CI 0.402 to 2.231), respectively. RFS and OS were similar between the study arms. Vaccine recipients developed strong positive antibody responses to NY-ESO-1 (p≤0.0001) and NY-ESO-1-specific CD4+ and CD8+ responses. Biopsies following relapse did not demonstrate differences in NY-ESO-1 expression between the study populations although an exploratory study demonstrated reduced (NY-ESO-1)+/Human Leukocyte Antigen (HLA) class I+ double-positive cells in biopsies from vaccine recipients performed on relapse in 19 participants. CONCLUSIONS The vaccine was well tolerated, however, despite inducing antigen-specific immunity, it did not affect survival endpoints. Immune escape through the downregulation of NY-ESO-1 and/or HLA class I molecules on tumor may have contributed to relapse.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/adverse effects
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Biopsy
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/adverse effects
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Chemotherapy, Adjuvant/adverse effects
- Chemotherapy, Adjuvant/methods
- Cholesterol/administration & dosage
- Cholesterol/adverse effects
- Dermatologic Surgical Procedures
- Disease-Free Survival
- Double-Blind Method
- Drug Combinations
- Female
- Follow-Up Studies
- Humans
- Immunogenicity, Vaccine
- Male
- Melanoma/diagnosis
- Melanoma/immunology
- Melanoma/mortality
- Melanoma/therapy
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Middle Aged
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/prevention & control
- Neoplasm Staging
- Phospholipids/administration & dosage
- Phospholipids/adverse effects
- Saponins/administration & dosage
- Saponins/adverse effects
- Skin/pathology
- Skin Neoplasms/diagnosis
- Skin Neoplasms/immunology
- Skin Neoplasms/mortality
- Skin Neoplasms/therapy
Collapse
Affiliation(s)
- Jonathan S Cebon
- Cancer Immunobiology Programme, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University at Austin Health, Heidelberg, Victoria, Australia
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
| | - Martin Gore
- Oncology, Royal Marsden Hospital NHS Trust, London, UK
| | - John F Thompson
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
| | - Ian D Davis
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
- Monash University Eastern Health Clinical School, Box Hill, Victoria, Australia
| | - Grant A McArthur
- Melanona and Skin Service, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Euan Walpole
- Cancer Services Division, Princess Alexandra Hospital Health Service District, Woolloongabba, Queensland, Australia
| | - Mark Smithers
- Oncology Services Unit, Princess Alexandra Hospital Health Service District, Woolloongabba, Queensland, Australia
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, Oxfordshire, UK
| | - P Rod Dunbar
- School of Biological Sciences and Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Duncan MacGregor
- Department of Anatomical Pathology, Austin Health, Heidelberg, Victoria, Australia
| | - Cyril Fisher
- Oncology, Royal Marsden Hospital NHS Trust, London, UK
| | - Michael Millward
- School of Medicine and Pharmacology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Paul Nathan
- Mount Vernon Cancer Centre, Mount Vernon Hospital, Northwood, London, UK
| | - Michael P N Findlay
- School of Medicine and Health Science, The University of Auckland, Auckland, New Zealand
| | - Peter Hersey
- Melanoma Immunology and Oncology Group, Centenary Institute, Newtown, New South Wales, Australia
| | - T R Jeffry Evans
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Jeremy Marsden
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Angus G Dalgleish
- Cell and Molecular Sciences, Division of Oncology, St Georges Hospital Medical School, London, UK
| | - Pippa G Corrie
- West Anglia Cancer Research Network Oncology Centre, Addenbrooke's Hospital, Cambridge, Cambridgeshire, UK
| | - Marples Maria
- The Cancer Research Centre, Weston Park Hospital, Sheffield, UK
| | - Margaret Brimble
- School of Biological Sciences and Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Geoff Williams
- School of Biological Sciences and Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Sintia Winkler
- School of Biological Sciences and Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Heather M Jackson
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
| | - Liliana Endo-Munoz
- Cancer Immunobiology Programme, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University at Austin Health, Heidelberg, Victoria, Australia
| | - Candani S A Tutuka
- Cancer Immunobiology Programme, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University at Austin Health, Heidelberg, Victoria, Australia
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
| | - Ralph Venhaus
- Ludwig Institute for Cancer Research, New York, New York, USA
| | - Lloyd J Old
- Ludwig Institute for Cancer Research, New York, New York, USA
| | - Dennis Haack
- Versagenics Inc, Morrisville, North Carolina, USA
| | | | - Andreas Behren
- Cancer Immunobiology Programme, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University at Austin Health, Heidelberg, Victoria, Australia
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
| | - Weisan Chen
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
- Biochemistry and Genetics, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
30
|
Chen H, Schürch CM, Noble K, Kim K, Krutzik PO, O'Donnell E, Vander Tuig J, Nolan GP, McIlwain DR. Functional comparison of PBMCs isolated by Cell Preparation Tubes (CPT) vs. Lymphoprep Tubes. BMC Immunol 2020; 21:15. [PMID: 32228458 PMCID: PMC7106580 DOI: 10.1186/s12865-020-00345-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/12/2020] [Indexed: 11/10/2022] Open
Abstract
Background Cryopreserved human peripheral blood mononuclear cells (PBMCs) are a commonly used sample type for a variety of immunological assays. Many factors can affect the quality of PBMCs, and careful consideration and validation of an appropriate PBMC isolation and cryopreservation method is important for well-designed clinical studies. A major point of divergence in PBMC isolation protocols is the collection of blood, either directly into vacutainers pre-filled with density gradient medium or the use of conical tubes containing a porous barrier to separate the density gradient medium from blood. To address potential differences in sample outcome, we isolated, cryopreserved, and compared PBMCs using parallel protocols differing only in the use of one of two common tube types for isolation. Methods Whole blood was processed in parallel using both Cell Preparation Tubes™ (CPT, BD Biosciences) and Lymphoprep™ Tubes (Axis-Shield) and assessed for yield and viability prior to cryopreservation. After thawing, samples were further examined by flow cytometry for cell yield, cell viability, frequency of 10 cell subsets, and capacity for stimulation-dependent CD4+ and CD8+ T cell intracellular cytokine production. Results No significant differences in cell recovery, viability, frequency of immune cell subsets, or T cell functionality between PBMC samples isolated using CPT or Lymphoprep tubes were identified. Conclusion CPT and Lymphoprep tubes are effective and comparable methods for PBMC isolation for immunological studies.
Collapse
Affiliation(s)
- Han Chen
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian M Schürch
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | - Garry P Nolan
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - David R McIlwain
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA. .,WCCT Global Inc., Cypress, CA, USA.
| |
Collapse
|
31
|
Jin HS, Choi DS, Ko M, Kim D, Lee DH, Lee S, Lee AY, Kang SG, Kim SH, Jung Y, Jeong Y, Chung JJ, Park Y. Extracellular pH modulating injectable gel for enhancing immune checkpoint inhibitor therapy. J Control Release 2019; 315:65-75. [PMID: 31669264 DOI: 10.1016/j.jconrel.2019.10.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/11/2019] [Accepted: 10/21/2019] [Indexed: 12/29/2022]
Abstract
Clinical data from diverse cancer types shows that the increased T cell infiltration in tumors correlates with improved patient prognosis. Acidic extracellular pH is a major attribute of the tumor microenvironment (TME) that promotes immune evasion and tumor progression. Therefore, antagonizing tumor acidity can be a powerful approach in cancer immunotherapy. Here, Pluronic F-127 is used as a NaHCO3 releasing carrier to focally alleviate extracellular tumor acidity. In a mouse tumor model, intratumoral treatment with pH modulating injectable gel (pHe-MIG) generates immune-favorable TME, as evidenced by the decrease of immune-suppressive cells and increase of tumor infiltrating CD8+T cells. The combination of pHe-MIG with immune checkpoint inhibitors, anti-PD-1 and anti-TIGIT antibodies, increases intratumoral T cell function, which leads to tumor clearance. Mechanistically, extracellular acidity was shown to upregulate co-inhibitory immune checkpoint receptors and inhibit mTOR signaling pathways in memory CD8+T cells, which impaired effector functions. Furthermore, an acidic pH environment increased the expression and engagement of TIGIT and its ligand CD155, which suggested that the extracellular pH can regulate the suppressive function of TIGIT pathway. Collectively, these findings suggest that pHe-MIG holds potential as a new TME modulator for effective immune checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Hyung-Seung Jin
- ASAN Institute for Life Sciences, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Da-Som Choi
- ASAN Institute for Life Sciences, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Minkyung Ko
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Dongkap Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea
| | - Dong-Hee Lee
- ASAN Institute for Life Sciences, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Soojin Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Ah Young Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seung Goo Kang
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Soo Hyun Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 136-705, Republic of Korea
| | - Youngmee Jung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Youngdo Jeong
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Justin J Chung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| | - Yoon Park
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
32
|
Dinh T, Oh J, Cameron DW, Lee SH, Cowan J. Differential immunomodulation of T-cells by immunoglobulin replacement therapy in primary and secondary antibody deficiency. PLoS One 2019; 14:e0223861. [PMID: 31613907 PMCID: PMC6793872 DOI: 10.1371/journal.pone.0223861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/30/2019] [Indexed: 11/18/2022] Open
Abstract
Patients with primary or secondary antibody deficiency (PAD or SAD) are at increased risk of recurrent infections that can be alleviated by immunoglobulin replacement therapy (IRT). In addition to replenishing antibody levels, IRT has been suggested to modulate immune response in patients with antibody deficiency. Although both commonly treated with IRT, the underlying causes of PAD and SAD vary greatly, suggesting differential modulation of T-cell function that may lead to different responses to IRT. To explore this, peripheral blood mononuclear cells (PBMCs) were sampled from 17 PAD and 14 SAD patients before and 2–10 months after initiation of IRT, and analyzed for changes in T-cell phenotype and function. Proportions of CD4, CD8, Treg, or memory T-cells did not significantly change post-IRT compared to pre-IRT. However, we report distinct modulation in T-cell function between PAD and SAD patients post-IRT. Upon α-CD3/CD28 stimulation, proportion of IFN-γ+ CD4 and CD8 T-cells increased in SAD (p = 0.005) but not PAD patients post-IRT compared to baseline. Interestingly, total T-cell proliferation was reduced post-IRT in both PAD and SAD patients, although the reduction in proliferation was primarily due to reduced CD4 T-cell proliferation in PAD (p = 0.025) in contrast to CD8 T-cells in SAD (p = 0.042). In summary, even though IRT provides patients with passive humoral immunity-mediated protection in PAD and SAD, our findings suggest that IRT immunomodulation of T-cells is different in T-cell subsets depending on underlying immunodeficiency.
Collapse
Affiliation(s)
- Tri Dinh
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jun Oh
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Donald William Cameron
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail: (SHL); (JC)
| | - Juthaporn Cowan
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- * E-mail: (SHL); (JC)
| |
Collapse
|
33
|
He J, Zhang R, Shao M, Zhao X, Miao M, Chen J, Liu J, Zhang X, Zhang X, Jin Y, Wang Y, Zhang S, Zhu L, Jacob A, Jia R, You X, Li X, Li C, Zhou Y, Yang Y, Ye H, Liu Y, Su Y, Shen N, Alexander J, Guo J, Ambrus J, Lin X, Yu D, Sun X, Li Z. Efficacy and safety of low-dose IL-2 in the treatment of systemic lupus erythematosus: a randomised, double-blind, placebo-controlled trial. Ann Rheum Dis 2019; 79:141-149. [PMID: 31537547 PMCID: PMC6937406 DOI: 10.1136/annrheumdis-2019-215396] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 08/03/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
Objectives Open-labelled clinical trials suggested that low-dose IL-2 might be effective in treatment of systemic lupus erythematosus (SLE). A double-blind and placebo-controlled trial is required to formally evaluate the safety and efficacy of low-dose IL-2 therapy. Methods A randomised, double-blind and placebo-controlled clinical trial was designed to treat 60 patients with active SLE. These patients received either IL-2 (n=30) or placebo (n=30) with standard treatment for 12 weeks, and were followed up for additional 12 weeks. IL-2 at a dose of 1 million IU or placebo was administered subcutaneously every other day for 2 weeks and followed by a 2-week break as one treatment cycle. The primary endpoint was the SLE Responder Index-4 (SRI-4) at week 12. The secondary endpoints were other clinical responses, safety and dynamics of immune cell subsets. Results At week 12, the SRI-4 response rates were 55.17% and 30.00% for IL-2 and placebo, respectively (p=0.052). At week 24, the SRI-4 response rate of IL-2 group was 65.52%, compared with 36.67% of the placebo group (p=0.027). The primary endpoint was not met at week 12. Low-dose IL-2 treatment resulted in 53.85% (7/13) complete remission in patients with lupus nephritis, compared with 16.67% (2/12) in the placebo group (p=0.036). No serious infection was observed in the IL-2 group, but two in placebo group. Besides expansion of regulatory T cells, low-dose IL-2 may also sustain cellular immunity with enhanced natural killer cells. Conclusions Low-dose IL-2 might be effective and tolerated in treatment of SLE. Trial registration number ClinicalTrials.gov Registries (NCT02465580 and NCT02932137).
Collapse
Affiliation(s)
- Jing He
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Beijing, China
| | - Ruijun Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Miao Shao
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xiaozhen Zhao
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Miao Miao
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Jiali Chen
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Jiajia Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Xiaoying Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Xia Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yuebo Jin
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yu Wang
- Center for Applied Statistics and School of Statistics, Renmin University of China, Beijing, China
| | - Shilei Zhang
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, China
| | - Lei Zhu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Alexander Jacob
- Department of Medicine, SUNY at Buffalo School of Medicine, Buffalo, New York, USA
| | - Rulin Jia
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Xujie You
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Xue Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Chun Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yunshan Zhou
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yue Yang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Hua Ye
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yanying Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yin Su
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Nan Shen
- Department of Rheumatology and Immunology,China-Australia Centre for Personalised Immunology, Shanghai Renji Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jessy Alexander
- Department of Medicine, SUNY at Buffalo School of Medicine, Buffalo, New York, USA
| | - Jianping Guo
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Beijing, China
| | - Julian Ambrus
- Department of Medicine, SUNY at Buffalo School of Medicine, Buffalo, New York, USA
| | - Xin Lin
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, China
| | - Di Yu
- Department of Rheumatology and Immunology,China-Australia Centre for Personalised Immunology, Shanghai Renji Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Shanghai, China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| |
Collapse
|
34
|
Whole Recombinant Saccharomyces cerevisiae Yeast Expressing Ras Mutations as Treatment for Patients With Solid Tumors Bearing Ras Mutations: Results From a Phase 1 Trial. J Immunother 2019. [PMID: 29528991 PMCID: PMC5895167 DOI: 10.1097/cji.0000000000000219] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We are developing whole, heat-killed, recombinant Saccharomyces cerevisiae yeast, engineered to encode target proteins, which stimulate immune responses against malignant cells expressing those targets. This phase 1 trial, enrolling patients with advanced colorectal or pancreas cancer, was designed to evaluate safety, immunogenicity, response, and overall survival of ascending doses of the GI-4000 series of products, which express 3 different forms of mutated Ras proteins. The study enrolled 33 heavily pretreated subjects (14 with pancreas and 19 with colorectal cancer), whose tumors were genotyped before enrollment to identify the specific ras mutation and thereby to identify which GI-4000 product to administer. No dose limiting toxicities were observed and no subject discontinued treatment due to a GI-4000 related adverse event (AE). The majority of AEs and all fatal events were due to underlying disease progression and AE frequencies were not significantly different among dose groups. GI-4000 was immunogenic, as Ras mutation-specific immune responses were detected on treatment in ∼60% of subjects. No objective tumor responses were observed but based on imaging, clinical status and/or biochemical markers, stable disease was observed in 6 subjects (18%) on day 29, while 1 subject had stable disease at days 57 and 85 follow-up visits. The median overall survival was 3.3 months (95% confidence interval, 2.3–5.3 mo), and 5 subjects survived past the 48-week follow-up period. No significant dose-dependent trends for survival were observed. This first clinical trial in humans with GI-4000 demonstrated a favorable safety profile and immunogenicity in the majority of subjects.
Collapse
|
35
|
Przybyla A, Zhang T, Li R, Roen DR, Mackiewicz A, Lehmann PV. Natural T cell autoreactivity to melanoma antigens: clonally expanded melanoma-antigen specific CD8 + memory T cells can be detected in healthy humans. Cancer Immunol Immunother 2019; 68:709-720. [PMID: 30783693 PMCID: PMC11028361 DOI: 10.1007/s00262-018-02292-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 12/24/2018] [Indexed: 12/30/2022]
Abstract
We used four-color ImmunoSpot® assays, in conjunction with peptide pools that cover the sequence of tyrosinase (Tyr), melanoma-associated antigen A3 (MAGE-A3), melanocyte antigen/melanoma antigen recognized by T cells 1 (Melan-A/MART-1), glycoprotein 100 (gp100), and New York esophageal squamous cell carcinoma-1 (NY-ESO-1) to characterize the melanoma antigen (MA)-specific CD8 + cell repertoire in PBMC of 40 healthy human donors (HD). Tyr triggered interferon gamma (IFN-γ)-secreting CD8 + T cells in 25% of HD within 24 h of antigen stimulation ex vivo. MAGE-A3, Melan-A/MART-1, and gp100 also induced recall responses in 10%, 7.5%, and 2.5% of HD, respectively. At this time point, these CD8 + T cells did not yet produce GzB (granzyme B). However, they engaged in GzB production after 72 h of antigen stimulation. By this 72-h time point, 57.5% of the HD responded to at least one, and typically several, of the MA. A closer characterization of the Tyr-specific CD8 + T cell repertoire indicated that it was low-affinity, and to primarily entail a stem cell-like subpopulation. Collectively, our data reveal pre-existing endogenous T cell immunity against melanoma antigens in healthy donors, and analogous to natural autoantibodies, we have termed this "natural T cell autoreactivity".
Collapse
Affiliation(s)
- Anna Przybyla
- Research and Development Department, Cellular Technology Limited (CTL), 20521 Chagrin Boulevard, Shaker Heights, Cleveland, OH, 44122-5350, USA
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Ting Zhang
- Research and Development Department, Cellular Technology Limited (CTL), 20521 Chagrin Boulevard, Shaker Heights, Cleveland, OH, 44122-5350, USA
| | - Ruliang Li
- Research and Development Department, Cellular Technology Limited (CTL), 20521 Chagrin Boulevard, Shaker Heights, Cleveland, OH, 44122-5350, USA
| | - Diana R Roen
- Research and Development Department, Cellular Technology Limited (CTL), 20521 Chagrin Boulevard, Shaker Heights, Cleveland, OH, 44122-5350, USA
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| | - Paul V Lehmann
- Research and Development Department, Cellular Technology Limited (CTL), 20521 Chagrin Boulevard, Shaker Heights, Cleveland, OH, 44122-5350, USA.
| |
Collapse
|
36
|
Khanniche A, Zhou L, Jiang B, Song J, Jin Y, Yin J, Wang S, Ji P, Shen H, Wang Y, Xu H. Restored and Enhanced Memory T Cell Immunity in Rheumatoid Arthritis After TNFα Blocker Treatment. Front Immunol 2019; 10:887. [PMID: 31105703 PMCID: PMC6499160 DOI: 10.3389/fimmu.2019.00887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 04/05/2019] [Indexed: 12/29/2022] Open
Abstract
TNFα inhibitors have shaped the landscape of rheumatoid arthritis (RA) therapy with high clinical efficiency. However, their impact on T cell recall responses is not well-elucidated. We aimed to analyze the immune profiles of memory T cells in RA patients undergoing TNFα inhibitor Golimumab (GM) treatment. Frequencies of peripheral T cell subsets and cytokine expression profiles in memory T cells (TM) upon PMA/Ionomycine stimulation were determined by flow cytometry. Antigen-specific CD8 T cell immunity was analyzed through stimulating PBMCs with CMV-EBV-Flu (CEF) viral peptide pool and subsequent intracellular IFNγ staining. Both peripheral CD8 and CD4 T cells from GM treated patients had a shift pattern characterized by an enlarged effector TM and a reduced central TM cell population when compared to GM untreated group. An increase in the frequencies of TNFα+, IL-2+, and IL-17+ CD8 TM cells was observed whereas only TNFα+CD4 TM cells increased in GM treated patients. Moreover, GM treated patients contained more peripheral IFNγ-producing CD8 T cells specific to CEF viral peptides. Together, these results show a distinct T cell subset pattern and enhanced memory T cell immunity upon GM treatment, suggesting an immunoregulatory effect of TNF inhibitor Golimumab on peripheral memory T cell responses.
Collapse
Affiliation(s)
- Asma Khanniche
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ling Zhou
- Department of Rheumatology and Immunology, Shanghai Chang Zheng Hospital, Second Military Medical University, Shanghai, China
| | - Bin Jiang
- Department of Rheumatology, Renji Hospital, Shanghai, China
| | - Jing Song
- Department of Rheumatology and Immunology, Shanghai Chang Zheng Hospital, Second Military Medical University, Shanghai, China
| | - Yanhua Jin
- Department of Rheumatology and Immunology, Shanghai Chang Zheng Hospital, Second Military Medical University, Shanghai, China
| | - Jian Yin
- Department of Rheumatology and Immunology, Shanghai Chang Zheng Hospital, Second Military Medical University, Shanghai, China
| | - Shujun Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Ji
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Shen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ying Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Chang Zheng Hospital, Second Military Medical University, Shanghai, China.,Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
37
|
Hu Z, Anandappa AJ, Sun J, Kim J, Leet DE, Bozym DJ, Chen C, Williams L, Shukla SA, Zhang W, Tabbaa D, Steelman S, Olive O, Livak KJ, Kishi H, Muraguchi A, Guleria I, Stevens J, Lane WJ, Burkhardt UE, Fritsch EF, Neuberg D, Ott PA, Keskin DB, Hacohen N, Wu CJ. A cloning and expression system to probe T-cell receptor specificity and assess functional avidity to neoantigens. Blood 2018; 132:1911-1921. [PMID: 30150207 PMCID: PMC6213317 DOI: 10.1182/blood-2018-04-843763] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/19/2018] [Indexed: 12/23/2022] Open
Abstract
Recent studies have highlighted the promise of targeting tumor neoantigens to generate potent antitumor immune responses and provide strong motivation for improving our understanding of antigen-T-cell receptor (TCR) interactions. Advances in single-cell sequencing technologies have opened the door for detailed investigation of the TCR repertoire, providing paired information from TCRα and TCRβ, which together determine specificity. However, a need remains for efficient methods to assess the specificity of discovered TCRs. We developed a streamlined approach for matching TCR sequences with cognate antigen through on-demand cloning and expression of TCRs and screening against candidate antigens. Here, we first demonstrate the system's capacity to identify viral-antigen-specific TCRs and compare the functional avidity of TCRs specific for a given antigen target. We then apply this system to identify neoantigen-specific TCR sequences from patients with melanoma treated with personalized neoantigen vaccines and characterize functional avidity of neoantigen-specific TCRs. Furthermore, we use a neoantigen-prediction pipeline to show that an insertion-deletion mutation in a putative chronic lymphocytic leukemia (CLL) driver gives rise to an immunogenic neoantigen mut-MGA, and use this approach to identify the mut-MGA-specific TCR sequence. This approach provides a means to identify and express TCRs, and then rapidly assess antigen specificity and functional avidity of a reconstructed TCR, which can be applied for monitoring antigen-specific T-cell responses, and potentially for guiding the design of effective T-cell-based immunotherapies.
Collapse
MESH Headings
- Antigens, Neoplasm/immunology
- Cancer Vaccines/therapeutic use
- Cells, Cultured
- Cloning, Molecular/methods
- HEK293 Cells
- Humans
- Jurkat Cells
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Melanoma/immunology
- Melanoma/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- T-Cell Antigen Receptor Specificity
Collapse
Affiliation(s)
- Zhuting Hu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Annabelle J Anandappa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Jing Sun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Jintaek Kim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Donna E Leet
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - David J Bozym
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Christina Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Sachet A Shukla
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA
| | - Wandi Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Diana Tabbaa
- Broad Institute of MIT and Harvard, Cambridge, MA
| | | | - Oriol Olive
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Kenneth J Livak
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA
| | - Hiroyuki Kishi
- Department of Immunology, University of Toyama, Toyama, Japan
| | | | - Indira Guleria
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | - Jonathan Stevens
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | - William J Lane
- Harvard Medical School, Boston, MA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | - Ute E Burkhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Edward F Fritsch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Donna Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA; and
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA; and
| | - Nir Hacohen
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Massachusetts General Hospital, Boston, MA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA; and
| |
Collapse
|
38
|
Salido J, Ruiz MJ, Trifone C, Figueroa MI, Caruso MP, Gherardi MM, Sued O, Salomón H, Laufer N, Ghiglione Y, Turk G. Phenotype, Polyfunctionality, and Antiviral Activity of in vitro Stimulated CD8 + T-Cells From HIV + Subjects Who Initiated cART at Different Time-Points After Acute Infection. Front Immunol 2018; 9:2443. [PMID: 30405632 PMCID: PMC6205955 DOI: 10.3389/fimmu.2018.02443] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/02/2018] [Indexed: 12/16/2022] Open
Abstract
Since anti-HIV treatment cannot cure the infection, many strategies have been proposed to eradicate the viral reservoir, which still remains as a major challenge. The success of some of these strategies will rely on the ability of HIV-specific CD8+ T-cells (CD8TC) to clear reactivated infected cells. Here, we aimed to investigate the phenotype and function of in vitro expanded CD8TC obtained from HIV+ subjects on combination antiretroviral therapy (cART), either initiated earlier (median = 3 months postinfection, ET: Early treatment) or later (median = 20 months postinfection, DT: Delayed treatment) after infection. Peripheral blood mononuclear cells from 12 DT and 13 ET subjects were obtained and stimulated with Nef and Gag peptide pools plus IL-2 for 14 days. ELISPOT was performed pre- and post-expansion. CD8TC memory/effector phenotype, PD-1 expression, polyfunctionality (CD107a/b, IFN-γ, IL-2, CCL4 (MIP-1β), and/or TNF-α production) and antiviral activity were evaluated post-expansion. Magnitude of ELISPOT responses increased after expansion by 103 times, in both groups. Expanded cells were highly polyfunctional, regardless of time of cART initiation. The memory/effector phenotype distribution was sharply skewed toward an effector phenotype after expansion in both groups although ET subjects showed significantly higher proportions of stem-cell and central memory CD8TCs. PD-1 expression was clustered in HIV-specific effector memory CD8TCs, subset that also showed the highest proportion of cytokine-producing cells. Moreover, PD-1 expression directly correlated with CD8TC functionality. Expanded CD8TCs from DT and ET subjects were highly capable of mediating antiviral activity, measured by two different assays. Antiviral function directly correlated with the proportion of fully differentiated effector cells (viral inhibition assay) as well as with CD8TC polyfunctionality and PD-1 expression (VITAL assay). In sum, we show that, despite being dampened in subjects on cART, the HIV-specific CD8TC response could be selectively stimulated and expanded in vitro, presenting a high proportion of cells able to carry-out multiple effector functions. Timing of cART initiation had an impact on the memory/effector differentiation phenotype, most likely reflecting how different periods of antigen persistence affected immune function. Overall, these results have important implications for the design and evaluation of strategies aimed at modulating CD8TCs to achieve the HIV functional cure.
Collapse
Affiliation(s)
- Jimena Salido
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - María Julia Ruiz
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - César Trifone
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | | | - María Paula Caruso
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - María Magdalena Gherardi
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Omar Sued
- Fundación Huésped, Buenos Aires, Argentina
| | - Horacio Salomón
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Natalia Laufer
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
- Hospital General de Agudos “Dr. JA Fernández”, Buenos Aires, Argentina
| | - Yanina Ghiglione
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Gabriela Turk
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| |
Collapse
|
39
|
Zhang X, Lu X, Moog C, Yuan L, Liu Z, Li Z, Xia W, Zhou Y, Wu H, Zhang T, Su B. KIR3DL1-Negative CD8 T Cells and KIR3DL1-Negative Natural Killer Cells Contribute to the Advantageous Control of Early Human Immunodeficiency Virus Type 1 Infection in HLA-B Bw4 Homozygous Individuals. Front Immunol 2018; 9:1855. [PMID: 30147699 PMCID: PMC6096002 DOI: 10.3389/fimmu.2018.01855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/27/2018] [Indexed: 12/20/2022] Open
Abstract
Bw4 homozygosity in human leukocyte antigen class B alleles has been associated with a delayed acquired immunodeficiency syndrome (AIDS) development and better control of human immunodeficiency virus type 1 (HIV-1) viral load (VL) than Bw6 homozygosity. Efficient CD8 T cell and natural killer (NK) cell functions have been described to restrain HIV-1 replication. However, the role of KIR3DL1 expression on these cells was not assessed in Bw4-homozygous participants infected with HIV-1 CRF01_A/E subtype, currently the most prevalent subtype in China. Here, we found that the frequency of KIR3DL1-expressing CD8 T cells of individuals homozygous for Bw6 [1.53% (0–4.56%)] was associated with a higher VL set point (Spearman rs = 0.59, P = 0.019), but this frequency of KIR3DL1+CD8+ T cells [1.37% (0.04–6.14%)] was inversely correlated with CD4 T-cell count in individuals homozygous for Bw4 (rs = −0.59, P = 0.011). Moreover, CD69 and Ki67 were more frequently expressed in KIR3DL1−CD8+ T cells in individuals homozygous for Bw4 than Bw6 (P = 0.046 for CD69; P = 0.044 for Ki67), although these molecules were less frequently expressed in KIR3DL1+CD8+ T cells than in KIR3DL1−CD8+ T cells in both groups (all P < 0.05). KIR3DL1−CD8+ T cells have stronger p24-specific CD8+ T-cell responses secreting IFN-γ and CD107a than KIR3DL1+CD8+ T cells in both groups (all P < 0.05). Thus, KIR3DL1 expression on CD8 T cells were associated with the loss of multiple functions. Interestingly, CD69+NK cells lacking KIR3DL1 expression were inversely correlated with HIV-1 VL set point in Bw4-homozygous individuals (rs = −0.52, P = 0.035). Therefore, KIR3DL1−CD8+ T cells with strong early activation and proliferation may, together with KIR3DL1−CD69+NK cells, play a protective role during acute/early HIV infection in individuals homozygous for Bw4. These findings highlight the superior functions of KIR3DL1−CD8+ T cells and KIR3DL1−CD69+NK cells being a potential factor contributing to delayed disease progression in the early stages of HIV-1 infection.
Collapse
Affiliation(s)
- Xin Zhang
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Xiaofan Lu
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Christiane Moog
- INSERM U1109, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Vaccine Research Institute (VRI), Créteil, France
| | - Lin Yuan
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Zhiying Liu
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Zhen Li
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Wei Xia
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Yuefang Zhou
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Hao Wu
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Tong Zhang
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Bin Su
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| |
Collapse
|
40
|
Direct Detection of T- and B-Memory Lymphocytes by ImmunoSpot® Assays Reveals HCMV Exposure that Serum Antibodies Fail to Identify. Cells 2018; 7:cells7050045. [PMID: 29783767 PMCID: PMC5981269 DOI: 10.3390/cells7050045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/13/2018] [Accepted: 05/15/2018] [Indexed: 01/04/2023] Open
Abstract
It is essential to identify donors who have not been infected with human cytomegalovirus (HCMV) in order to avoid transmission of HCMV to recipients of blood transfusions or organ transplants. In the present study, we tested the reliability of seronegativity as an indicator for the lack of HCMV exposure in healthy human blood donors. Eighty-two HCMV seronegative individuals were identified, and their peripheral blood mononuclear cells (PBMC) were tested in ImmunoSpot® assays for the presence of HCMV-specific T- and B-memory lymphocytes. Eighty-two percent (67 of 82) of these HCMV seronegative individuals featured at least one memory cell that was lineage specific for HCMV, with the majority of these subjects possessing CD4+ and CD8+ T cells, as well as B cells, providing three independent lines of evidence for having developed immunity to HCMV. Only 15 of these 82 donors (18%) showed neither T- nor B-cell memory to HCMV, consistent with immunological naïveté to the virus. The data suggest that measurements of serum antibodies frequently fail to reveal HCMV exposure in humans, which may be better identified by direct detection of HCMV-specific memory lymphocytes.
Collapse
|
41
|
Welter A, Sundararaman S, Li R, Zhang T, Karulin AY, Lehmann A, Naeem V, Roen DR, Kuerten S, Lehmann PV. High-Throughput GLP-Capable Target Cell Visualization Assay for Measuring Cell-Mediated Cytotoxicity. Cells 2018; 7:cells7050035. [PMID: 29695103 PMCID: PMC5981259 DOI: 10.3390/cells7050035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 11/17/2022] Open
Abstract
One of the primary effector functions of immune cells is the killing of virus-infected or malignant cells in the body. Natural killer (NK) and CD8 effector T cells are specialized for this function. The gold standard for measuring such cell-mediated cytolysis has been the chromium release assay, in which the leakage of the radioactive isotope from damaged target cells is being detected. Flow cytometry-based single cell analysis of target cells has recently been established as a non-radioactive alternative. Here we introduce a target cell visualization assay (TVA) that applies similar target cell staining approaches as used in flow cytometry but based on single cell computer image analysis. Two versions of TVA are described here. In one, the decrease in numbers of calcein-stained, i.e., viable, target cells is assessed. In the other, the CFSE/PI TVA, the increase in numbers of dead target cells is established in addition. TVA assays are shown to operate with the same sensitivity as standard chromium release assays, and, leaving data audit trails in form of scanned (raw), analyzed, and quality-controlled images, thus meeting requirements for measuring cell-mediated cytolysis in a regulated environment.
Collapse
Affiliation(s)
- Anna Welter
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Srividya Sundararaman
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Ruliang Li
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Ting Zhang
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Alexey Y Karulin
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Alexander Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Villian Naeem
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Diana R Roen
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Paul V Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| |
Collapse
|
42
|
Diederich JM, Staudt M, Meisel C, Hahn K, Meinl E, Meisel A, Klehmet J. Neurofascin and Compact Myelin Antigen-Specific T Cell Response Pattern in Chronic Inflammatory Demyelinating Polyneuropathy Subtypes. Front Neurol 2018; 9:171. [PMID: 29615965 PMCID: PMC5868132 DOI: 10.3389/fneur.2018.00171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/06/2018] [Indexed: 12/20/2022] Open
Abstract
Objective The objective of this study is to investigate whether chronic inflammatory demyelinating polyneuropathy (CIDP) and its subtypes differ in their type 1 T-helper (TH1) cell response against nodal/paranodal neurofascin (NF186, NF155) as well as myelin protein zero (P0 180–199) and myelin basic protein (MBP 82–100). Methods Interferon-gamma (IFN-γ) enzyme-linked immunospot assay was used to detect antigen-specific T cell responses in 48 patients suffering typical CIDP (n = 18), distal acquired demyelinating polyneuropathy (n = 8), multifocal acquired demyelinating sensory and motor polyneuropathy (MADSAM; n = 9), and sensory CIDP (n = 13) compared to other non-immune polyneuropathy (ON; n = 19) and healthy controls (n = 9). Results Compared to controls, MADSAM and sensory CIDP patients showed broadest IFN-γ T cell responses to all four antigens. Positive IFN-γ responses against two or more antigens were highly predictive for CIDP (positive predictive value = 0.95) and were found in 77% of CIDP patients. Patients with limited antigen-specific response were females, more severely affected with neuropathic pain and proximal paresis. The area under the receiver operating characteristics curve (AUC) of NF186 in MADSAM was 0.94 [95% confidential interval (CI) 0.82–1.00] compared to ON. For sensory CIDP, AUC of P0 180–199 was 0.94 (95% CI 0.86–1.00) and for MBP 82–100 0.95 (95% CI 0.88–1.00) compared to ON. Conclusion Cell-mediated immune responses to (para)nodal and myelin-derived antigens are common in CIDP. TH1 response against NF186 may be used as a biomarker for MADSAM and TH1 responses against P0 180–199 and MBP 82–100 as biomarkers for sensory CIDP. Larger multicenter studies study are warranted in order to establish these immunological markers as a diagnostic tools.
Collapse
Affiliation(s)
| | - Maximilian Staudt
- Neurocure Research Center Berlin, Charité University Medicine, Berlin, Germany
| | - Christian Meisel
- Department of Medical Immunology, Charité University Medicine, Berlin, Germany
| | - Katrin Hahn
- Department of Neurology, Charité University Medicine, Berlin, Germany
| | - Edgar Meinl
- Clinical Neuroimmunology, Ludwigs-Maximilians University, Munich, Germany
| | - Andreas Meisel
- Neurocure Research Center Berlin, Charité University Medicine, Berlin, Germany.,Department of Neurology, Charité University Medicine, Berlin, Germany
| | - Juliane Klehmet
- Neurocure Research Center Berlin, Charité University Medicine, Berlin, Germany
| |
Collapse
|
43
|
Jeewandara C, Ogg GS, Malavige GN. Cultured ELISpot Assay to Investigate Dengue Virus Specific T-Cell Responses. Methods Mol Biol 2018; 1808:165-171. [PMID: 29956182 DOI: 10.1007/978-1-4939-8567-8_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The cultured Enzyme-Linked ImmunoSpot (ELISpot) assay is a functional T cell assay, which is commonly used to assess virus-specific T cell responses. The use of an in vitro expansion step before the ELISpot distinguishes such "cultured" ELISpots from "ex vivo" ELISpots. Cultured ELISpots have the advantage that lower frequency responses can be analyzed compared to ex vivo ELISpots, but do carry the associated potential distortions of the expansion phase. Cultured ELISpot assays are of value to determine silent and symptomatic transmission of the Dengue virus (DENV) in the community and to identify the correlates of a DENV-specific protective immune response. We have evaluated T cell responses to the DENV using cultured ELISpot assays with serotype-specific T cell epitopes to determine past infecting dengue virus (DENV) serotypes. The peptides used in this assay do not cross react with the Japanese encephalitis virus nor other flaviviruses. Therefore, this assay is likely to be useful in determining the past infecting DENV serotype in immune-epidemiological studies and in dengue vaccine trials.
Collapse
Affiliation(s)
- Chandima Jeewandara
- Centre for Dengue Research, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Graham S Ogg
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Gathsaurie Neelika Malavige
- Centre for Dengue Research, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka. .,MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
44
|
Klehmet J, Staudt M, Diederich JM, Siebert E, Meinl E, Harms L, Meisel A. Neurofascin (NF)155- and NF186-Specific T Cell Response in a Patient Developing a Central Pontocerebellar Demyelination after 10 Years of CIDP. Front Neurol 2017; 8:724. [PMID: 29312139 PMCID: PMC5744188 DOI: 10.3389/fneur.2017.00724] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/13/2017] [Indexed: 01/29/2023] Open
Abstract
Background Information and pathobiological understanding about central demyelinating manifestation in patients, who primarily suffer from chronic inflammatory demyelinating polyneuropathy (CIDP), are scarce. Methods IFN-γ-response as well as antibodies against the (para)nodal antigens neurofascin (NF)155 and NF 186 had been tested by Elispot assay and ELISA before clinical manifestation and at follow-up. Case description and results The patient described here developed a subacute brainstem syndrome more than 10 years after diagnosis of CIDP under low-dose maintenance treatment of intravenous immunoglobulins (IVIG). MRI revealed enhancing right-sided pontocerebellar lesion. CSF examination showed mild pleocytosis and elevated protein, and negative oligoclonal bands. Further diagnostics exclude differential diagnoses such as tuberculoma, sarcoidosis, or metastasis. Specific IFN-γ response against NF155 and NF186 as measured by Elispot assay was elevated before clinical manifestation. NF155 and NF186 antibodies were negative. Escalation of IVIG treatment at 2 g/kg BW followed by 1.4 g/kg BW led to clinical remission albeit to a new asymptomatic central lesion. Follow-up NF155 and NF186-Elispot turned negative. Conclusion The case reported here with a delayed central manifestation after an initially typical CIDP and NF155 and NF186 T cell responses does not resemble described cases of combined central and peripheral demyelination but may reflect a novel subtype within the great clinical heterogeneity of CIDP.
Collapse
Affiliation(s)
- Juliane Klehmet
- Charité University Medicine Berlin, NeuroCure Clinical Research Center (NCRC), Berlin, Germany.,Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Max Staudt
- Charité University Medicine Berlin, NeuroCure Clinical Research Center (NCRC), Berlin, Germany
| | - Jan-Markus Diederich
- Charité University Medicine Berlin, NeuroCure Clinical Research Center (NCRC), Berlin, Germany
| | - Eberhard Siebert
- Department of Neuroradiology, Charité University Medicine Berlin, Berlin, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Lutz Harms
- Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Andreas Meisel
- Charité University Medicine Berlin, NeuroCure Clinical Research Center (NCRC), Berlin, Germany.,Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
45
|
A Positive Control for Detection of Functional CD4 T Cells in PBMC: The CPI Pool. Cells 2017; 6:cells6040047. [PMID: 29215584 PMCID: PMC5753071 DOI: 10.3390/cells6040047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 12/29/2022] Open
Abstract
Testing of peripheral blood mononuclear cells (PBMC) for immune monitoring purposes requires verification of their functionality. This is of particular concern when the PBMC have been shipped or stored for prolonged periods of time. While the CEF (Cytomegalo-, Epstein-Barr and Flu-virus) peptide pool has become the gold standard for testing CD8 cell functionality, a positive control for CD4 cells is so far lacking. The latter ideally consists of proteins so as to control for the functionality of the antigen processing and presentation compartments, as well. Aiming to generate a positive control for CD4 cells, we first selected 12 protein antigens from infectious/environmental organisms that are ubiquitous: Varicella, Influenza, Parainfluenza, Mumps, Cytomegalovirus, Streptococcus, Mycoplasma, Lactobacillus, Neisseria, Candida, Rubella, and Measles. Of these antigens, three were found to elicited interferon (IFN)-γ-producing CD4 cells in the majority of human test subjects: inactivated cytomegalo-, parainfluenza-, and influenza virions (CPI). While individually none of these three antigens triggered a recall response in all donors, the pool of the three (the ‘CPI pool’), did. One hundred percent of 245 human donors tested were found to be CPI positive, including Caucasians, Asians, and African-Americans. Therefore, the CPI pool appears to be suitable to serve as universal positive control for verifying the functionality of CD4 and of antigen presenting cells.
Collapse
|
46
|
Ruiz MJ, Salido J, Abusamra L, Ghiglione Y, Cevallos C, Damilano G, Rodriguez AM, Trifone C, Laufer N, Giavedoni LD, Sued O, Salomón H, Gherardi MM, Turk G. Evaluation of Different Parameters of Humoral and Cellular Immune Responses in HIV Serodiscordant Heterosexual Couples: Humoral Response Potentially Implicated in Modulating Transmission Rates. EBioMedicine 2017; 26:25-37. [PMID: 29129698 PMCID: PMC5832641 DOI: 10.1016/j.ebiom.2017.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/24/2017] [Accepted: 11/01/2017] [Indexed: 02/05/2023] Open
Abstract
As the HIV/AIDS pandemic still progresses, understanding the mechanisms governing viral transmission as well as protection from HIV acquisition is fundamental. In this context, cohorts of HIV serodiscordant heterosexual couples (SDC) represent a unique tool. The present study was aimed to evaluate specific parameters of innate, cellular and humoral immune responses in SDC. Specifically, plasma levels of cytokines and chemokines, HIV-specific T-cell responses, gp120-specific IgG and IgA antibodies, and HIV-specific antibody-dependent cellular cytotoxicity (ADCC) activity were assessed in nine HIV-exposed seronegative individuals (ESN) and their corresponding HIV seropositive partners (HIV+-P), in eighteen chronically infected HIV subjects (C), nine chronically infected subjects known to be HIV transmitters (CT) and ten healthy HIV− donors (HD). Very low magnitude HIV-specific cellular responses were found in two out of six ESN. Interestingly, HIV+-P had the highest ADCC magnitude, the lowest IgA levels and the highest IgG/IgA ratio, all compared to CT. Positive correlations between CD4+ T-cell counts and both IgG/IgA ratios and %ADCC killing uniquely distinguished HIV+-P. Additionally, evidence of IgA interference with ADCC responses from HIV+-P and CT is provided. These data suggest for the first time a potential role of ADCC and/or gp120-specific IgG/IgA balance in modulating heterosexual transmission. In sum, this study provides key information to understand the host factors that influence viral transmission, which should be considered in both the development of prophylactic vaccines and novel immunotherapies for HIV-1 infection. The evaluation of different immune parameters in HIV serodiscordant couples helped identify factors shaping transmission. Innate and cellular immune responses were apparently not involved in this scenario. HIV-specific ADCC, IgA titer and IgG/IgA balance were identified as factors involved in modulating viral transmission.
The existence of individuals that remain HIV negative despite being repeatedly exposed to the virus has long been described. To date, only homozygosis for a 32-base pair deletion in the ccr5 gene has been consistently shown to be a determinant of HIV resistance. Still, subjects bearing the WT ccr5 gene have also been described as resistant or less susceptible to HIV. Thus, other mechanisms must be involved in this phenomenon. The results presented here postulate ADCC and IgG/IgA ratio as potential mechanisms involved in modulating HIV transmission in the context of serodiscordant couples and inspire further investigations.
Collapse
Affiliation(s)
- María Julia Ruiz
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Jimena Salido
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | | | - Yanina Ghiglione
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Cintia Cevallos
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Gabriel Damilano
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Ana María Rodriguez
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - César Trifone
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Natalia Laufer
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina; Hospital Juan A. Fernández, Unidad Enfermedades Infecciosas, Buenos Aires, Argentina
| | - Luis D Giavedoni
- Department of Virology and Immunology, Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Omar Sued
- Fundación Huésped, Buenos Aires, Argentina; Hospital Juan A. Fernández, Unidad Enfermedades Infecciosas, Buenos Aires, Argentina
| | - Horacio Salomón
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - María Magdalena Gherardi
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Gabriela Turk
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina.
| |
Collapse
|
47
|
Nitschke NJ, Bjoern J, Iversen TZ, Andersen MH, Svane IM. Indoleamine 2,3-dioxygenase and survivin peptide vaccine combined with temozolomide in metastatic melanoma. Stem Cell Investig 2017; 4:77. [PMID: 29057249 DOI: 10.21037/sci.2017.08.06] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/25/2017] [Indexed: 11/06/2022]
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase (IDO) and survivin have been identified as potential targets for cancer vaccination. In this phase II study a vaccine using the peptides Sur1M2 and IDO5 was combined with the chemotherapy temozolomide (TMZ) for treatment of metastatic melanoma patients. The aim was to simultaneously target several immune inhibiting mechanisms and the highly malignant cells expressing survivin. METHODS HLA-A2 positive patients with advanced malignant melanoma were treated biweekly with 150 mg/m2 TMZ daily for 7 days followed by subcutaneous vaccination with 250 µg of each peptide in 500 µL Montanide solution at day 8. Granulocyte-macrophage colony-stimulating factor was used as an adjuvant and topical imiquimod was applied prior to vaccination. Treatment was continued until disease progression. Clinical response was evaluated by PET-CT and immunological outcome was assessed by ELISPOT and flow cytometry. RESULTS In total, 17 patients were treated with a clinical benefit rate of 18% including one patient with partial tumor regression. Immune analyses revealed a vaccine specific response in 8 (67%) of 12 patients tested, a significant decrease in the frequency of CD4+ T-cells during treatment, a tendency towards decreasing frequencies of naïve CD4+ and CD8+ T-cells, and increasing frequencies of memory CD4+ and CD8+ T-cells. CONCLUSIONS These results demonstrate that vaccine-induced immunity towards survivin and IDO-derived peptides can be achieved in combination with TMZ in patients mainly suffering from grade M1c melanoma including patients with brain metastases. A significant clinical activity could not be proven in this small study and a larger setup is needed to properly assess clinical efficacy.
Collapse
Affiliation(s)
- Nikolaj Juul Nitschke
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Jon Bjoern
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Herlev, Denmark.,Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | | | - Mads Hald Andersen
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Herlev, Denmark.,Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| |
Collapse
|
48
|
Delayed Activation Kinetics of Th2- and Th17 Cells Compared to Th1 Cells. Cells 2017; 6:cells6030029. [PMID: 28895901 PMCID: PMC5617975 DOI: 10.3390/cells6030029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 08/29/2017] [Accepted: 09/05/2017] [Indexed: 01/12/2023] Open
Abstract
During immune responses, different classes of T cells arise: Th1, Th2, and Th17. Mobilizing the right class plays a critical role in successful host defense and therefore defining the ratios of Th1/Th2/Th17 cells within the antigen-specific T cell repertoire is critical for immune monitoring purposes. Antigen-specific Th1, Th2, and Th17 cells can be detected by challenging peripheral blood mononuclear cells (PBMC) with antigen, and establishing the numbers of T cells producing the respective lead cytokine, IFN-γ and IL-2 for Th1 cells, IL-4 and IL-5 for Th2, and IL-17 for Th-17 cells, respectively. Traditionally, these cytokines are measured within 6 h in flow cytometry. We show here that 6 h of stimulation is sufficient to detect peptide-induced production of IFN-γ, but 24 h are required to reveal the full frequency of protein antigen-specific Th1 cells. Also the detection of IL-2 producing Th1 cells requires 24 h stimulation cultures. Measurements of IL-4 producing Th2 cells requires 48-h cultures and 96 h are required for frequency measurements of IL-5 and IL-17 secreting T cells. Therefore, accounting for the differential secretion kinetics of these cytokines is critical for the accurate determination of the frequencies and ratios of antigen-specific Th1, Th2, and Th17 cells.
Collapse
|
49
|
Comte D, Karampetsou MP, Yoshida N, Kis-Toth K, Kyttaris VC, Tsokos GC. Signaling Lymphocytic Activation Molecule Family Member 7 Engagement Restores Defective Effector CD8+ T Cell Function in Systemic Lupus Erythematosus. Arthritis Rheumatol 2017; 69:1035-1044. [PMID: 28076903 DOI: 10.1002/art.40038] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/05/2017] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Effector CD8+ T cell function is impaired in systemic lupus erythematosus (SLE) and is associated with a compromised ability to fight infections. Signaling lymphocytic activation molecule family member 7 (SLAMF7) engagement has been shown to enhance natural killer cell degranulation. This study was undertaken to characterize the expression and function of SLAMF7 on CD8+ T cell subsets isolated from the peripheral blood of SLE patients and healthy subjects. METHODS CD8+ T cell subset distribution, SLAMF7 expression, and expression of cytolytic enzymes (perforin, granzyme A [GzmA], and GzmB) on cells isolated from SLE patients and healthy controls were analyzed by flow cytometry. CD107a expression and interferon-γ (IFNγ) production in response to viral antigenic stimulation in the presence or absence of an anti-SLAMF7 antibody were assessed by flow cytometry. Antiviral cytotoxic activity in response to SLAMF7 engagement was determined using a flow cytometry-based assay. RESULTS The distribution of CD8+ T cell subsets was altered in the peripheral blood of SLE patients, with a decreased effector cell subpopulation. Memory CD8+ T cells from SLE patients displayed decreased amounts of SLAMF7, a surface receptor that characterizes effector CD8+ T cells. Ligation of SLAMF7 increased CD8+ T cell degranulation capacity and the percentage of IFNγ-producing cells in response to antigen challenge in SLE patients and healthy controls. Moreover, SLAMF7 engagement promoted cytotoxic lysis of target cells in response to stimulation with viral antigens. CONCLUSION CD8+ T cell activation in response to viral antigens is defective in SLE patients. Activation of SLAMF7 through a specific monoclonal antibody restores CD8+ T cell antiviral effector function to normal levels and thus represents a potential therapeutic option in SLE.
Collapse
Affiliation(s)
- Denis Comte
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, and Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Maria P Karampetsou
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Nobuya Yoshida
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Katalin Kis-Toth
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Vasileios C Kyttaris
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - George C Tsokos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
50
|
Archin NM, Kirchherr JL, Sung JA, Clutton G, Sholtis K, Xu Y, Allard B, Stuelke E, Kashuba AD, Kuruc JD, Eron J, Gay CL, Goonetilleke N, Margolis DM. Interval dosing with the HDAC inhibitor vorinostat effectively reverses HIV latency. J Clin Invest 2017; 127:3126-3135. [PMID: 28714868 DOI: 10.1172/jci92684] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 06/01/2017] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The histone deacetylase (HDAC) inhibitor vorinostat (VOR) can increase HIV RNA expression in vivo within resting CD4+ T cells of aviremic HIV+ individuals. However, while studies of VOR or other HDAC inhibitors have reported reversal of latency, none has demonstrated clearance of latent infection. We sought to identify the optimal dosing of VOR for effective serial reversal of HIV latency. METHODS In a study of 16 HIV-infected, aviremic individuals, we measured resting CD4+ T cell-associated HIV RNA ex vivo and in vivo following a single exposure to VOR, and then in vivo after a pair of doses separated by 48 or 72 hours, and finally following a series of 10 doses given at 72-hour intervals. RESULTS Serial VOR exposures separated by 72 hours most often resulted in an increase in cell-associated HIV RNA within circulating resting CD4+ T cells. VOR was well tolerated by all participants. However, despite serial reversal of latency over 1 month of VOR dosing, we did not observe a measurable decrease (>0.3 log10) in the frequency of latent infection within resting CD4+ T cells. CONCLUSIONS These findings outline parameters for the experimental use of VOR to clear latent infection. Latency reversal can be achieved by VOR safely and repeatedly, but effective depletion of persistent HIV infection will require additional advances. In addition to improvements in latency reversal, these advances may include the sustained induction of potent antiviral immune responses capable of recognizing and clearing the rare cells in which HIV latency has been reversed. TRIAL REGISTRATION Clinicaltrials.gov NCT01319383. FUNDING NIH grants U01 AI095052, AI50410, and P30 CA016086 and National Center for Advancing Translational Sciences grant KL2 TR001109.
Collapse
Affiliation(s)
- Nancie M Archin
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and
| | - Jennifer L Kirchherr
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases
| | - Julia Am Sung
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and
| | - Genevieve Clutton
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Microbiology and Immunology, UNC Chapel Hill School of Medicine
| | - Katherine Sholtis
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases
| | - Yinyan Xu
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases
| | - Brigitte Allard
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases
| | - Erin Stuelke
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases
| | - Angela D Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy
| | - Joann D Kuruc
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and
| | - Joseph Eron
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and.,Department of Epidemiology, UNC Chapel Hill School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Cynthia L Gay
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and
| | - Nilu Goonetilleke
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Microbiology and Immunology, UNC Chapel Hill School of Medicine
| | - David M Margolis
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine and.,Microbiology and Immunology, UNC Chapel Hill School of Medicine.,Department of Epidemiology, UNC Chapel Hill School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|