1
|
Krishna BA, Lim EY, Metaxaki M, Jackson S, Mactavous L, Lyons PA, Doffinger R, Bradley JR, Smith KGC, Sinclair J, Matheson NJ, Lehner PJ, Sithole N, Wills MR. Spontaneous, persistent, T cell-dependent IFN-γ release in patients who progress to Long Covid. SCIENCE ADVANCES 2024; 10:eadi9379. [PMID: 38381822 PMCID: PMC10881041 DOI: 10.1126/sciadv.adi9379] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
After acute infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), a proportion of patients experience persistent symptoms beyond 12 weeks, termed Long Covid. Understanding the mechanisms that cause this debilitating disease and identifying biomarkers for diagnostic, therapeutic, and monitoring purposes are urgently required. We detected persistently high levels of interferon-γ (IFN-γ) from peripheral blood mononuclear cells of patients with Long Covid using highly sensitive FluoroSpot assays. This IFN-γ release was seen in the absence of ex vivo peptide stimulation and remains persistently elevated in patients with Long Covid, unlike the resolution seen in patients recovering from acute SARS-CoV-2 infection. The IFN-γ release was CD8+ T cell-mediated and dependent on antigen presentation by CD14+ cells. Longitudinal follow-up of our study cohort showed that symptom improvement and resolution correlated with a decrease in IFN-γ production to baseline levels. Our study highlights a potential mechanism underlying Long Covid, enabling the search for biomarkers and therapeutics in patients with Long Covid.
Collapse
Affiliation(s)
- Benjamin A. Krishna
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Eleanor Y. Lim
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Marina Metaxaki
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge CB2 0AW, UK
| | - Sarah Jackson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Lenette Mactavous
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - NIHR BioResource
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Paul A. Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - John R. Bradley
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- National Institute for Health Research (NIHR) Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
- Department of Renal Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Kenneth G. C. Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - John Sinclair
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Nicholas J. Matheson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
- NHS Blood and Transplant, Cambridge CB2 0PT, UK
| | - Paul J. Lehner
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Nyaradzai Sithole
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Mark R. Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
2
|
Raineri D, Abreu H, Vilardo B, Kustrimovic N, Venegoni C, Cappellano G, Chiocchetti A. Deep Flow Cytometry Unveils Distinct Immune Cell Subsets in Inducible T Cell Co-Stimulator Ligand (ICOSL)- and ICOS-Knockout Mice during Experimental Autoimmune Encephalomyelitis. Int J Mol Sci 2024; 25:2509. [PMID: 38473756 DOI: 10.3390/ijms25052509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
The inducible T cell co-stimulator ligand (ICOSL), expressed by antigen presenting cells, binds to the inducible T cell co-stimulator (ICOS) on activated T cells. Improper function of the ICOS/ICOSL pathway has been implicated in several autoimmune diseases, including multiple sclerosis (MS). Previous studies showed that ICOS-knockout (KO) mice exhibit severe experimental autoimmune encephalomyelitis (EAE), the animal model of MS, but data on ICOSL deficiency are not available. In our study, we explored the impact of both ICOS and ICOSL deficiencies on MOG35-55 -induced EAE and its associated immune cell dynamics by employing ICOSL-KO and ICOS-KO mice with a C57BL/6J background. During EAE resolution, MOG-driven cytokine levels and the immunophenotype of splenocytes were evaluated by ELISA and multiparametric flow cytometry, respectively. We found that both KO mice exhibited an overlapping and more severe EAE compared to C57BL/6J mice, corroborated by a reduction in memory/regulatory T cell subsets and interleukin (IL-)17 levels. It is noteworthy that an unsupervised analysis showed that ICOSL deficiency modifies the immune response in an original way, by affecting T central and effector memory (TCM, TEM), long-lived CD4+ TEM cells, and macrophages, compared to ICOS-KO and C57BL/6J mice, suggesting a role for other binding partners to ICOSL in EAE development, which deserves further study.
Collapse
Affiliation(s)
- Davide Raineri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Hugo Abreu
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Beatrice Vilardo
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Natasa Kustrimovic
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Chiara Venegoni
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Giuseppe Cappellano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| |
Collapse
|
3
|
Imbratta C, Gela A, Bilek N, Mabwe S, Cloete Y, Mortensen R, Borges ÁH, Maenetje P, Mlotshwa M, Churchyard G, Sudi L, Sabi I, Meewes P, Wallis CL, Hatherill M, Scriba TJ, Nemes E. Qualification of the differential leukocyte count and immunophenotyping in cryopreserved ex vivo whole blood assay. Cytometry A 2023; 103:992-1003. [PMID: 37675607 DOI: 10.1002/cyto.a.24793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/25/2023] [Accepted: 09/04/2023] [Indexed: 09/08/2023]
Abstract
We developed a flow cytometry-based assay, termed Differential Leukocyte Counting and Immunophenotyping in Cryopreserved Ex vivo whole blood (DLC-ICE), that allows quantification of absolute counts and frequencies of leukocyte subsets and measures expression of activation, phenotypic and functional markers. We evaluated the performance of the DLC-ICE assay by determining inter-operator variability for processing fresh whole blood (WB) from healthy donors collected at multiple clinical sites. In addition, we assessed inter-operator variability for staining of fixed cells and robustness across different anticoagulants. Accuracy was evaluated by comparing DLC-ICE measurements to real-time cell enumeration using an accredited hematology analyzer. Finally, we developed and tested the performance of a 27-colour immunophenotyping panel on cryopreserved fixed WB and compared results to matched fresh WB. Overall, we observed <20% variability in absolute counts and frequencies of granulocytes, monocytes and lymphocytes (T, B and NK cells) when fresh WB was collected in different anti-coagulant tubes, processed or stained by independent operators. Absolute cell counts measured across operators and anti-coagulants using the DLC-ICE method exhibited excellent correlation with the reference method, complete blood count (CBC) with differential, measured using a hematology analyzer (r2 > 0.9 for majority of measurements). A comparison of leukocyte immunophenotyping on fresh WB versus DLC-ICE processed blood yielded equivalent and linear results over a wide dynamic range (r2 = 0.94 over 10-104 cells/μL). These results demonstrate low variability across trained operators, high robustness, linearity and accuracy, supporting utility of the DLC-ICE assay for large cohort studies involving multiple clinical research sites.
Collapse
Affiliation(s)
- Claire Imbratta
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Anele Gela
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Nicole Bilek
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Simbarashe Mabwe
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Yolundi Cloete
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Rasmus Mortensen
- Department of Infectious Diseases Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Álvaro H Borges
- Department of Infectious Diseases Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Pholo Maenetje
- Aurum Institute, Parktown, South Africa
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Mandla Mlotshwa
- Aurum Institute, Parktown, South Africa
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Gavin Churchyard
- Aurum Institute, Parktown, South Africa
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Lwitiho Sudi
- Mbeya Medical Research Centre, National Institute for Medical Research (NIMR), Mbeya, Tanzania
| | - Issa Sabi
- Mbeya Medical Research Centre, National Institute for Medical Research (NIMR), Mbeya, Tanzania
| | | | - Carole L Wallis
- BARC, South Africa
- Lancet Laboratories, Johannesburg, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
4
|
Davies EL, Noor M, Lim EY, Houldcroft CJ, Okecha G, Atkinson C, Reeves MB, Jackson SE, Wills MR. HCMV carriage in the elderly diminishes anti-viral functionality of the adaptive immune response resulting in virus replication at peripheral sites. Front Immunol 2022; 13:1083230. [PMID: 36591233 PMCID: PMC9797693 DOI: 10.3389/fimmu.2022.1083230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection and periodic reactivation is, generally, well controlled by adaptative immune responses in the healthy. In older people, overt HCMV disease is rarely seen despite the association of HCMV with increased risk of mortality; evidence from studies of unwell aged populations suggest that HCMV seropositivity is an important co-morbidity factor. HCMV genomes have been detected in urine from older donors, suggesting that the immune response prevents systemic disease but possibly immunomodulation due to lifelong viral carriage may alter its efficacy at peripheral tissue sites. Previously we have demonstrated that there were no age-related expansions of T cell responses to HCMV or increase in latent viral carriage with age and these T cells produced anti-viral cytokines and viremia was very rarely detected. To investigate the efficacy of anti-HCMV responses with increasing age, we used an in vitro Viral Dissemination Assay (VDA) using autologous dermal fibroblasts to determine the anti-viral effector capacity of total PBMC, as well as important subsets (T cells, NK cells). In parallel we assessed components of the humoral response (antibody neutralization) and combined this with qPCR detection of HCMV in blood, saliva and urine in a cohort of young and old donors. Consistent with previous studies, we again show HCMV specific cIL-10, IFNγ and TNFα T cell responses to peptides did not show an age-related defect. However, assessment of direct anti-viral cellular and antibody-mediated adaptive immune responses using the VDA shows that older donors are significantly less able to control viral dissemination in an in vitro assay compared to young donors. Corroborating this observation, we detected viral genomes in saliva samples only from older donors, these donors had a defect in cellular control of viral spread in our in vitro assay. Phenotyping of fibroblasts used in this study shows expression of a number of checkpoint inhibitor ligands which may contribute to the defects observed. The potential to therapeutically intervene in checkpoint inhibitor pathways to prevent HCMV reactivation in the unwell aged is an exciting avenue to explore.
Collapse
Affiliation(s)
- Emma L. Davies
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Mahlaqua Noor
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Eleanor Y. Lim
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Charlotte J. Houldcroft
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Georgina Okecha
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Claire Atkinson
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Matthew B. Reeves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Sarah E. Jackson
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Mark R. Wills
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| |
Collapse
|
5
|
Harari A, Sarivalasis A, de Jonge K, Thierry AC, Huber F, Boudousquie C, Rossier L, Orcurto A, Imbimbo M, Baumgaertner P, Bassani-Sternberg M, Kandalaft LE. A Personalized Neoantigen Vaccine in Combination with Platinum-Based Chemotherapy Induces a T-Cell Response Coinciding with a Complete Response in Endometrial Carcinoma. Cancers (Basel) 2021; 13:5801. [PMID: 34830955 PMCID: PMC8616532 DOI: 10.3390/cancers13225801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 11/17/2022] Open
Abstract
Endometrial cancer (EC) is a common gynecological malignancy and the fourth most common malignancy in European and North American women. Amongst EC, the advanced serous, p53-mutated, and pMMR subtypes have the highest risk of relapse despite optimal standard of care therapy. At present, there is no standard of care maintenance treatment to prevent relapse among these high-risk patients. Vaccines are a form of immunotherapy that can potentially increase the immunogenicity of pMMR, serous, and p53-mutated tumors to render them responsive to check point inhibitor-based immunotherapy. We demonstrate, for the first time, the feasibility of generating a personalized dendritic cell vaccine pulsed with peptide neoantigens in a patient with pMMR, p53-mutated, and serous endometrial adenocarcinoma (SEC). The personalized vaccine was administered in combination with systemic chemotherapy to treat an inoperable metastatic recurrence. This treatment association demonstrated the safety and immunogenicity of the personalized dendritic cell vaccine. Interestingly, a complete oncological response was obtained with respect to both radiological assessment and the tumor marker CA-125.
Collapse
Affiliation(s)
- Alexandre Harari
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (K.d.J.); (A.-C.T.); (F.H.); (C.B.); (L.R.); (P.B.); (M.B.-S.)
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), 1005 Lausanne, Switzerland
| | - Apostolos Sarivalasis
- Department of Oncology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland; (A.S.); (A.O.); (M.I.)
| | - Kaat de Jonge
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (K.d.J.); (A.-C.T.); (F.H.); (C.B.); (L.R.); (P.B.); (M.B.-S.)
| | - Anne-Christine Thierry
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (K.d.J.); (A.-C.T.); (F.H.); (C.B.); (L.R.); (P.B.); (M.B.-S.)
| | - Florian Huber
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (K.d.J.); (A.-C.T.); (F.H.); (C.B.); (L.R.); (P.B.); (M.B.-S.)
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), 1005 Lausanne, Switzerland
| | - Caroline Boudousquie
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (K.d.J.); (A.-C.T.); (F.H.); (C.B.); (L.R.); (P.B.); (M.B.-S.)
| | - Laetitia Rossier
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (K.d.J.); (A.-C.T.); (F.H.); (C.B.); (L.R.); (P.B.); (M.B.-S.)
| | - Angela Orcurto
- Department of Oncology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland; (A.S.); (A.O.); (M.I.)
| | - Martina Imbimbo
- Department of Oncology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland; (A.S.); (A.O.); (M.I.)
| | - Petra Baumgaertner
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (K.d.J.); (A.-C.T.); (F.H.); (C.B.); (L.R.); (P.B.); (M.B.-S.)
| | - Michal Bassani-Sternberg
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (K.d.J.); (A.-C.T.); (F.H.); (C.B.); (L.R.); (P.B.); (M.B.-S.)
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), 1005 Lausanne, Switzerland
| | - Lana E. Kandalaft
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (K.d.J.); (A.-C.T.); (F.H.); (C.B.); (L.R.); (P.B.); (M.B.-S.)
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), 1005 Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland; (A.S.); (A.O.); (M.I.)
| |
Collapse
|
6
|
Hao Y, Hao S, Andersen-Nissen E, Mauck WM, Zheng S, Butler A, Lee MJ, Wilk AJ, Darby C, Zager M, Hoffman P, Stoeckius M, Papalexi E, Mimitou EP, Jain J, Srivastava A, Stuart T, Fleming LM, Yeung B, Rogers AJ, McElrath JM, Blish CA, Gottardo R, Smibert P, Satija R. Integrated analysis of multimodal single-cell data. Cell 2021; 184:3573-3587.e29. [PMID: 34062119 PMCID: PMC8238499 DOI: 10.1016/j.cell.2021.04.048] [Citation(s) in RCA: 7712] [Impact Index Per Article: 1928.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/03/2021] [Accepted: 04/28/2021] [Indexed: 02/08/2023]
Abstract
The simultaneous measurement of multiple modalities represents an exciting frontier for single-cell genomics and necessitates computational methods that can define cellular states based on multimodal data. Here, we introduce "weighted-nearest neighbor" analysis, an unsupervised framework to learn the relative utility of each data type in each cell, enabling an integrative analysis of multiple modalities. We apply our procedure to a CITE-seq dataset of 211,000 human peripheral blood mononuclear cells (PBMCs) with panels extending to 228 antibodies to construct a multimodal reference atlas of the circulating immune system. Multimodal analysis substantially improves our ability to resolve cell states, allowing us to identify and validate previously unreported lymphoid subpopulations. Moreover, we demonstrate how to leverage this reference to rapidly map new datasets and to interpret immune responses to vaccination and coronavirus disease 2019 (COVID-19). Our approach represents a broadly applicable strategy to analyze single-cell multimodal datasets and to look beyond the transcriptome toward a unified and multimodal definition of cellular identity.
Collapse
Affiliation(s)
- Yuhan Hao
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; New York Genome Center, New York, NY 10013, USA
| | - Stephanie Hao
- Technology Innovation Lab, New York Genome Center, New York, NY 10013, USA
| | - Erica Andersen-Nissen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Cape Town HVTN Immunology Lab, Hutchinson Cancer Research Institute of South Africa, Cape Town 8001, South Africa
| | - William M Mauck
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Shiwei Zheng
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; New York Genome Center, New York, NY 10013, USA
| | - Andrew Butler
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; New York Genome Center, New York, NY 10013, USA
| | - Maddie J Lee
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aaron J Wilk
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Charlotte Darby
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Michael Zager
- Center for Data Visualization, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Paul Hoffman
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Marlon Stoeckius
- Technology Innovation Lab, New York Genome Center, New York, NY 10013, USA
| | - Efthymia Papalexi
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; New York Genome Center, New York, NY 10013, USA
| | - Eleni P Mimitou
- Technology Innovation Lab, New York Genome Center, New York, NY 10013, USA
| | - Jaison Jain
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Avi Srivastava
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Tim Stuart
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Lamar M Fleming
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Angela J Rogers
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Juliana M McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94063, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Peter Smibert
- Technology Innovation Lab, New York Genome Center, New York, NY 10013, USA.
| | - Rahul Satija
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; New York Genome Center, New York, NY 10013, USA.
| |
Collapse
|
7
|
Baumgaertner P, Sankar M, Herrera F, Benedetti F, Barras D, Thierry AC, Dangaj D, Kandalaft LE, Coukos G, Xenarios I, Guex N, Harari A. Unsupervised Analysis of Flow Cytometry Data in a Clinical Setting Captures Cell Diversity and Allows Population Discovery. Front Immunol 2021; 12:633910. [PMID: 33995353 PMCID: PMC8119773 DOI: 10.3389/fimmu.2021.633910] [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: 11/26/2020] [Accepted: 04/12/2021] [Indexed: 11/13/2022] Open
Abstract
Data obtained with cytometry are increasingly complex and their interrogation impacts the type and quality of knowledge gained. Conventional supervised analyses are limited to pre-defined cell populations and do not exploit the full potential of data. Here, in the context of a clinical trial of cancer patients treated with radiotherapy, we performed longitudinal flow cytometry analyses to identify multiple distinct cell populations in circulating whole blood. We cross-compared the results from state-of-the-art recommended supervised analyses with results from MegaClust, a high-performance data-driven clustering algorithm allowing fast and robust identification of cell-type populations. Ten distinct cell populations were accurately identified by supervised analyses, including main T, B, dendritic cell (DC), natural killer (NK) and monocytes subsets. While all ten subsets were also identified with MegaClust, additional cell populations were revealed (e.g. CD4+HLA-DR+ and NKT-like subsets), and DC profiling was enriched by the assignment of additional subset-specific markers. Comparison between transcriptomic profiles of purified DC populations and publicly available datasets confirmed the accuracy of the unsupervised clustering algorithm and demonstrated its potential to identify rare and scarcely described cell subsets. Our observations show that data-driven analyses of cytometry data significantly enrich the amount and quality of knowledge gained, representing an important step in refining the characterization of immune responses.
Collapse
Affiliation(s)
- Petra Baumgaertner
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Martial Sankar
- Vital-IT, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Fernanda Herrera
- Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Fabrizio Benedetti
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - David Barras
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Anne-Christine Thierry
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Denarda Dangaj
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Lana E Kandalaft
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Ioannis Xenarios
- Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Nicolas Guex
- Vital-IT, Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Bioinformatics Competence Center (BICC), University of Lausanne, Lausanne, Switzerland
| | - Alexandre Harari
- Centre of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
8
|
Braudeau C, Salabert-Le Guen N, Chevreuil J, Rimbert M, Martin JC, Josien R. An easy and reliable whole blood freezing method for flow cytometry immuno-phenotyping and functional analyses. CYTOMETRY PART B-CLINICAL CYTOMETRY 2021; 100:652-665. [PMID: 33544978 DOI: 10.1002/cyto.b.21994] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/01/2020] [Accepted: 01/26/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Immune profiling by flow cytometry is not always possible on fresh blood samples due to time and/or transport constraints. Furthermore, the cryopreservation of peripheral blood mononuclear cells (PBMC) requires on-site specialized lab facilities, thus severely restricting the extent to which blood immune monitoring can be applied to multicenter clinical studies. These major limitations can be addressed through the development of simplified whole blood freezing methods. METHODS In this report, we describe an optimized easy protocol for rapid whole blood freezing with the CryoStor® CS10 solution. Using flow cytometry, we compared cellular viability and composition on cryopreserved whole blood samples to matched fresh blood, as well as fresh and frozen PBMC. RESULTS Though partial loss of neutrophils was observed, leucocyte viability was routinely >75% and we verified the preservation of viable T cells, NK cells, monocytes, dendritic cells, and eosinophils in frequencies similar to those observed in fresh samples. A moderate decrease in B cell frequencies was observed. Importantly, we validated the possibility to analyze major intracellular markers, such as FOXP3 and Helios in regulatory T cells. Finally, we demonstrated good functional preservation of CS10-cryopreserved cells through the analysis of intracellular cytokine production in ex vivo stimulated T cells (IFNg, IL-4, IL-17A,) and monocytes (IL-1b, IL-6, TNFa). CONCLUSIONS In conclusion, our protocol provides a robust method to apply reliable immune monitoring studies to cryopreserved whole blood samples, hence offering new important opportunities for the design of future multicenter clinical trials.
Collapse
Affiliation(s)
- Cecile Braudeau
- Laboratoire d'Immunologie, CIMNA, LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Nina Salabert-Le Guen
- Laboratoire d'Immunologie, CIMNA, LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Justine Chevreuil
- Laboratoire d'Immunologie, CIMNA, LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Marie Rimbert
- Laboratoire d'Immunologie, CIMNA, LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Jerome C Martin
- Laboratoire d'Immunologie, CIMNA, LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Regis Josien
- Laboratoire d'Immunologie, CIMNA, LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| |
Collapse
|
9
|
Staats J. Immunophenotyping of Human Regulatory T Cells. Methods Mol Biol 2019; 2032:141-177. [PMID: 31522418 DOI: 10.1007/978-1-4939-9650-6_9] [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: 05/13/2023]
Abstract
Regulatory T cells, also known as Tregs, play a pivotal role in maintaining homeostasis of the immune system and self-tolerance. Tregs express CD3, CD4, CD25, and FOXP3 but lack CD127. CD4 and CD3 identify helper T lymphocytes, of which Tregs are a subset. CD25 is IL-2Rα, an essential activation marker that is expressed in high levels on Tregs. FOXP3 is the canonical transcription factor, important in the development, maintenance, and identification of Tregs. CD127 is IL-7 receptor, expressed inversely with suppression, and is therefore downregulated on Tregs. Flow cytometry is a powerful tool that is capable of simultaneously measuring Tregs along with several markers associated with subpopulations of Tregs, activation, maturation, proliferation, and surrogates of functional suppression. This chapter describes a multicolor flow cytometry-based approach to measure human Tregs, including details for surface staining, fixation/permeabilization, intracellular/intranuclear staining, acquisition of samples on a flow cytometer, plus analysis and interpretation of resulting FCS files.
Collapse
Affiliation(s)
- Janet Staats
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
- Duke Immune Profiling Core, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
10
|
Elizaga ML, Li SS, Kochar NK, Wilson GJ, Allen MA, Tieu HVN, Frank I, Sobieszczyk ME, Cohen KW, Sanchez B, Latham TE, Clarke DK, Egan MA, Eldridge JH, Hannaman D, Xu R, Ota-Setlik A, McElrath MJ, Hay CM. Safety and tolerability of HIV-1 multiantigen pDNA vaccine given with IL-12 plasmid DNA via electroporation, boosted with a recombinant vesicular stomatitis virus HIV Gag vaccine in healthy volunteers in a randomized, controlled clinical trial. PLoS One 2018; 13:e0202753. [PMID: 30235286 PMCID: PMC6147413 DOI: 10.1371/journal.pone.0202753] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/03/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The addition of plasmid cytokine adjuvants, electroporation, and live attenuated viral vectors may further optimize immune responses to DNA vaccines in heterologous prime-boost combinations. The objective of this study was to test the safety and tolerability of a novel prime-boost vaccine regimen incorporating these strategies with different doses of IL-12 plasmid DNA adjuvant. METHODS In a phase 1 study, 88 participants received an HIV-1 multiantigen (gag/pol, env, nef/tat/vif) DNA vaccine (HIV-MAG, 3000 μg) co-administered with IL-12 plasmid DNA adjuvant at 0, 250, 1000, or 1500 μg (N = 22/group) given intramuscularly with electroporation (Ichor TriGrid™ Delivery System device) at 0, 1 and 3 months; followed by attenuated recombinant vesicular stomatitis virus, serotype Indiana, expressing HIV-1 Gag (VSV-Gag), 3.4 ⊆ 107 plaque-forming units (PFU), at 6 months; 12 others received placebo. Injections were in both deltoids at each timepoint. Participants were monitored for safety and tolerability for 15 months. RESULTS The dose of IL-12 pDNA did not increase pain scores, reactogenicity, or adverse events with the co-administered DNA vaccine, or following the VSV-Gag boost. Injection site pain and reactogenicity were common with intramuscular injections with electroporation, but acceptable to most participants. VSV-Gag vaccine often caused systemic reactogenicity symptoms, including a viral syndrome (in 41%) of fever, chills, malaise/fatigue, myalgia, and headache; and decreased lymphocyte counts 1 day after vaccination. CONCLUSIONS HIV-MAG DNA vaccine given by intramuscular injection with electroporation was safe at all doses of IL-12 pDNA. The VSV-Gag vaccine at this dose was associated with fever and viral symptoms in some participants, but the vaccine regimens were safe and generally well-tolerated. TRIAL REGISTRATION Clinical Trials.gov NCT01578889.
Collapse
Affiliation(s)
- Marnie L. Elizaga
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Shuying S. Li
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Nidhi K. Kochar
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Gregory J. Wilson
- Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Mary A. Allen
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hong Van N. Tieu
- Laboratory of Infectious Disease Prevention, New York Blood Center, New York, New York, United States of America
| | - Ian Frank
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Magdalena E. Sobieszczyk
- Division of Infectious Diseases, Columbia University Medical Center, New York, New York, United States of America
| | - Kristen W. Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Brittany Sanchez
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Theresa E. Latham
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - David K. Clarke
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - Michael A. Egan
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - John H. Eldridge
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - Drew Hannaman
- Ichor Medical Systems, Incorporated, San Diego, California, United States of America
| | - Rong Xu
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - Ayuko Ota-Setlik
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Christine Mhorag Hay
- Infectious Diseases Division, University of Rochester Medical Center, Rochester, New York, United States of America
| | | |
Collapse
|
11
|
Baumgaertner P, Ioannidou K, Speiser DE. Immune Monitoring of Blood and Tumor Microenvironment. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
12
|
Optimizing recovery of frozen human peripheral blood mononuclear cells for flow cytometry. PLoS One 2017; 12:e0187440. [PMID: 29091947 PMCID: PMC5665600 DOI: 10.1371/journal.pone.0187440] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/19/2017] [Indexed: 11/28/2022] Open
Abstract
Introduction Live peripheral blood mononuclear cells (PBMCs) can be frozen and thawed for later analyses by adding and removing a cryoprotectant, such as dimethyl sulfoxide (DMSO). Laboratories across the world use various procedures, but published evidence of optimal thawing procedures is scarce. Materials and methods PBMCs were separated from blood collected from healthy Danish blood donors, and stored at -80°C after adding of DMSO. The essential steps in the thawing procedure were modified and performance was evaluated by flow cytometry with respect to the percentage and total yield of viable PMBCs. Results The best-performing washing medium was Roswell Park Memorial Institute (RPMI) 1640 at 37°C with 20% fetal bovine serum. When using 10 mL washing medium in a 15-mL Falcon tube, samples should be centrifuged for at least 10 minutes at 500 g. We failed to detect any differences between the tested methods of mixing PBMCs with washing medium. Likewise, neither the thawing duration nor centrifugation temperature (20°C and 37°C) had any effect. PBMCs could be incubated (rested) for up to eight hours in a 37°C 5% CO2 incubator without affecting cell counts, but incubating PBMCs for 16 hours significantly decreased viability and recovery. In general, high viability was not necessarily associated with high recovery. Conclusion Changing the thawing procedure significantly impacted PBMC viability and live cell recovery. Evaluating both viability and live PBMC recovery are necessary to evaluate method performance. Investigation of differential loss of PBMC subtypes and phenotypic changes during thawing and incubation requires further evaluation.
Collapse
|
13
|
Agashe C, Chiang D, Grishin A, Masilamani M, Jones SM, Wood RA, Sicherer SH, Burks AW, Leung DYM, Dawson P, Sampson HA, Berin MC. Impact of granulocyte contamination on PBMC integrity of shipped blood samples: Implications for multi-center studies monitoring regulatory T cells. J Immunol Methods 2017; 449:23-27. [PMID: 28629732 PMCID: PMC5573627 DOI: 10.1016/j.jim.2017.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/29/2017] [Accepted: 06/15/2017] [Indexed: 11/15/2022]
Abstract
In centralized immune monitoring for a multi-center allergen immunotherapy trial, we observed frequent loss of CD4+ T cell integrity following staining of cultured PBMCs with our regulatory T cell flow cytometry panel. Samples were marked by a loss of total cellular events, altered scatter properties, and reduced CD3+CD4+ events. This occurred only in samples that were stained with Foxp3 and were therefore treated with Foxp3 fixation-permeabilization buffer. We identified granulocyte contamination in samples associated with a loss of integrity, and went on to test the impact of granulocyte depletion on day-old blood samples. Granulocyte depletion prevented loss of cell integrity and CD3+CD4+ events, and reduced variability in detection of Foxp3+ cells. Addition of purified neutrophils back to PBMCs altered scatter properties and detection of CD4+ T cells. Implementation of a granulocyte depletion step in our standard operating protocols has reduced assay failure due to loss of sample integrity from 31% to 0%. Routine incorporation of a granulocyte depletion step during PBMC isolation is recommended prior to downstream immune monitoring in blood with next-day processing.
Collapse
Affiliation(s)
- Charuta Agashe
- Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Chiang
- Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Grishin
- Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madhan Masilamani
- Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stacie M Jones
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, AR, USA
| | - Robert A Wood
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Scott H Sicherer
- Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A Wesley Burks
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | | | - Hugh A Sampson
- Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M Cecilia Berin
- Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
14
|
Jackson SE, Sedikides GX, Okecha G, Poole EL, Sinclair JH, Wills MR. Latent Cytomegalovirus (CMV) Infection Does Not Detrimentally Alter T Cell Responses in the Healthy Old, But Increased Latent CMV Carriage Is Related to Expanded CMV-Specific T Cells. Front Immunol 2017; 8:733. [PMID: 28694811 PMCID: PMC5483450 DOI: 10.3389/fimmu.2017.00733] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/09/2017] [Indexed: 01/22/2023] Open
Abstract
Human cytomegalovirus (HCMV) primary infection and periodic reactivation of latent virus is generally well controlled by T-cell responses in healthy people. In older donors, overt HCMV disease is not generally seen despite the association of HCMV infection with increased risk of mortality. However, increases in HCMV DNA in urine of older people suggest that, although the immune response retains functionality, immunomodulation of the immune response due to lifelong viral carriage may alter its efficacy. Viral transcription is limited during latency to a handful of viral genes and there is both an IFNγ and cellular IL-10 CD4+ T-cell response to HCMV latency-associated proteins. Production of cIL-10 by HCMV-specific CD4+ T-cells is a candidate for aging-related immunomodulation. To address whether long-term carriage of HCMV changes the balance of cIL-10 and IFNγ-secreting T-cell populations, we recruited a large donor cohort aged 23–78 years and correlated T-cell responses to 11 HCMV proteins with age, HCMV IgG levels, latent HCMV load in CD14+ monocytes, and T-cell numbers in the blood. IFNγ responses by CD4+ and CD8+ T-cells to all HCMV proteins were detected, with no age-related increase in this cohort. IL-10-secreting CD4+ T cell responses were predominant to latency-associated proteins but did not increase with age. Quantification of HCMV genomes in CD14+ monocytes, a known site of latent HCMV carriage, did not reveal any increase in viral genome copies in older donors. Importantly, there was a significant positive correlation between the latent viral genome copy number and the breadth and magnitude of the IFNγ T-cell response to HCMV proteins. This study suggests in healthy aged donors that HCMV-specific changes in the T cell compartment were not affected by age and were effective, as viremia was a very rare event. Evidence from studies of unwell aged has shown HCMV to be an important comorbidity factor, surveillance of latent HCMV load and low-level viremia in blood and body fluids, alongside typical immunological measures and assessment of the antiviral capacity of the HCMV-specific immune cell function would be informative in determining if antiviral treatment of HCMV replication in the old maybe beneficial.
Collapse
Affiliation(s)
- Sarah E Jackson
- Division of Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - George X Sedikides
- Division of Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Georgina Okecha
- Division of Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Emma L Poole
- Division of Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - John H Sinclair
- Division of Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Mark R Wills
- Division of Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
15
|
Ford T, Wenden C, Mbekeani A, Dally L, Cox JH, Morin M, Winstone N, Hill AVS, Gilmour J, Ewer KJ. Cryopreservation-related loss of antigen-specific IFNγ producing CD4 + T-cells can skew immunogenicity data in vaccine trials: Lessons from a malaria vaccine trial substudy. Vaccine 2017; 35:1898-1906. [PMID: 28285985 PMCID: PMC5387668 DOI: 10.1016/j.vaccine.2017.02.038] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/31/2017] [Accepted: 02/18/2017] [Indexed: 12/19/2022]
Abstract
Ex vivo functional immunoassays such as ELISpot and intracellular cytokine staining (ICS) by flow cytometry are crucial tools in vaccine development both in the identification of novel immunogenic targets and in the immunological assessment of samples from clinical trials. Cryopreservation and subsequent thawing of PBMCs via validated processes has become a mainstay of clinical trials due to processing restrictions inherent in the disparate location and capacity of trial centres, and also in the need to standardize biological assays at central testing facilities. Logistical and financial requirement to batch process samples from multiple study timepoints are also key. We used ELISpot and ICS assays to assess antigen-specific immunogenicity in blood samples taken from subjects enrolled in a phase II malaria heterologous prime-boost vaccine trial and showed that the freeze thaw process can result in a 3–5-fold reduction of malaria antigen-specific IFNγ-producing CD3+CD4+ effector populations from PBMC samples taken post vaccination. We have also demonstrated that peptide responsive CD8+ T cells are relatively unaffected, as well as CD4+ T cell populations that do not produce IFNγ. These findings contribute to a growing body of data that could be consolidated and synthesised as guidelines for clinical trials with the aim of increasing the efficiency of vaccine development pipelines.
Collapse
Affiliation(s)
- Tom Ford
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK.
| | - Claire Wenden
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Alison Mbekeani
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Len Dally
- EMMES Corporation, Rockville, MD, USA
| | - Josephine H Cox
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK
| | | | - Nicola Winstone
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Adrian V S Hill
- Centre for Clinical Vaccinology and Tropical Medicine and the Jenner Institute Laboratories, University of Oxford, UK
| | - Jill Gilmour
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Katie J Ewer
- Centre for Clinical Vaccinology and Tropical Medicine and the Jenner Institute Laboratories, University of Oxford, UK
| |
Collapse
|
16
|
Draxler DF, Madondo MT, Hanafi G, Plebanski M, Medcalf RL. A flowcytometric analysis to efficiently quantify multiple innate immune cells and T Cell subsets in human blood. Cytometry A 2017; 91:336-350. [PMID: 28264143 DOI: 10.1002/cyto.a.23080] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 12/20/2016] [Accepted: 02/16/2017] [Indexed: 01/28/2023]
Abstract
The balance of inflammation and immunosuppression driven by changed ratios in diverse myeloid and T cell subsets, as well as their state of activation and ability to migrate to lymphoid compartments or inflammatory sites, has emerged as a highly active area of study across clinical trials of vaccines and therapies against cancer, trauma, as well as autoimmune and infectious diseases. There is a need for effective protocols which maximally use the possibilities offered by modern flow cytometers to characterize such immune cell changes in peripheral blood using small volumes of human blood. Additionally, longitudinal clinical studies often use cryopreserved samples, which can impact flow cytometric results. To efficiently gauge both the innate and the adaptive immune response, two novel 15-color antibody panels to identify key myeloid and T cell subsets and their functional potential were established. This approach was used to compare cellular immune profiles in fresh whole blood and in matched cryopreserved peripheral blood mononuclear cells (PBMCs). Cocktail I was designed to identify and characterize myeloid cell populations including dendritic cells (DCs), monocytic monocyte-derived suppressor cells (MO-MDSC), and monocytes, determining further core aspects of their state of maturity, T cell stimulatory (or inhibitory) potential, and migration capability. Cocktail II was used for phenotyping diverse T cells subsets, and their key migration and functional regulatory capabilities. The two 15-color antibody panels for the evaluation of both immune-stimulating and immunosuppressive processes presented herein allowed for efficient evaluation of the balance of immune activation versus immunosuppression across key blood cells, with good resolution for all 15 markers stained for in each panel. Gating strategies for the myeloid and T cells are presented to further support specific subset identification. This protocol was shown to be reproducible across donors and useful to study both RBC-lysed whole blood and cryopreserved PBMCs. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- D F Draxler
- Molecular Neurotrauma and Haemostasis, Australian Center for Blood Diseases, Central Clinical School, Monash University, Clayton, Australia
| | - M T Madondo
- Vaccine and Infectious Diseases Laboratory, Department of Immunology and Pathology, Monash University, Clayton, Australia
| | - G Hanafi
- Molecular Neurotrauma and Haemostasis, Australian Center for Blood Diseases, Central Clinical School, Monash University, Clayton, Australia
| | - M Plebanski
- Vaccine and Infectious Diseases Laboratory, Department of Immunology and Pathology, Monash University, Clayton, Australia
| | - R L Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Center for Blood Diseases, Central Clinical School, Monash University, Clayton, Australia
| |
Collapse
|
17
|
Koguchi Y, Gonzalez IL, Meeuwsen TL, Miller WL, Haley DP, Tanibata-Branham AN, Bahjat KS. A Semi-automated Approach to Preparing Antibody Cocktails for Immunophenotypic Analysis of Human Peripheral Blood. J Vis Exp 2016:e53485. [PMID: 26890325 PMCID: PMC4781742 DOI: 10.3791/53485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Immunophenotyping of peripheral blood by flow cytometry determines changes in the frequency and activation status of peripheral leukocytes during disease and treatment. It has the potential to predict therapeutic efficacy and identify novel therapeutic targets. Whole blood staining utilizes unmanipulated blood, which minimizes artifacts that can occur during sample preparation. However, whole blood staining must also be done on freshly collected blood to ensure the integrity of the sample. Additionally, it is best to prepare antibody cocktails on the same day to avoid potential instability of tandem-dyes and prevent reagent interaction between brilliant violet dyes. Therefore, whole blood staining requires careful standardization to control for intra and inter-experimental variability. Here, we report deployment of an automated liquid handler equipped with a two-dimensional (2D) barcode reader into a standard process of making antibody cocktails for flow cytometry. Antibodies were transferred into 2D barcoded tubes arranged in a 96 well format and their contents compiled in a database. The liquid handler could then locate the source antibody vials by referencing antibody names within the database. Our method eliminated tedious coordination for positioning of source antibody tubes. It provided versatility allowing the user to easily change any number of details in the antibody dispensing process such as specific antibody to use, volume, and destination by modifying the database without rewriting the scripting in the software method for each assay. A proof of concept experiment achieved outstanding inter and intra- assay precision, demonstrated by replicate preparation of an 11-color, 17-antibody flow cytometry assay. These methodologies increased overall throughput for flow cytometry assays and facilitated daily preparation of the complex antibody cocktails required for the detailed phenotypic characterization of freshly collected anticoagulated peripheral blood.
Collapse
Affiliation(s)
- Yoshinobu Koguchi
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center;
| | - Iliana L Gonzalez
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center
| | - Tanisha L Meeuwsen
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center
| | - William L Miller
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center
| | - Daniel P Haley
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center; Sony Biotechnology
| | | | - Keith S Bahjat
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center; Bristol-Myers Squibb;
| |
Collapse
|
18
|
Implementation of highly sophisticated flow cytometry assays in multicenter clinical studies: considerations and guidance. Bioanalysis 2015; 7:1299-311. [DOI: 10.4155/bio.15.61] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Flow cytometry is increasingly becoming an important technology for biomarkers used in drug discovery and development. Within clinical development flow cytometry is used for the determination of PD biomarkers, disease or efficacy biomarkers or patient stratification biomarkers. Significant differences exist between flow cytometry methodology and other widely used technologies measuring soluble biomarkers including ligand binding and mass spectrometry. These differences include the very heavy reliance on aspects of sample processing techniques as well as sample stabilization to ensure viable samples. These differences also require exploration of new approaches and wider discussion regarding method validation requirements. This paper provides a review of the current challenges, solutions, regulatory environment and recommendations for the application of flow cytometry to measure biomarkers in clinical development.
Collapse
|
19
|
Nemes E, Kagina BMN, Smit E, Africa H, Steyn M, Hanekom WA, Scriba TJ. Differential leukocyte counting and immunophenotyping in cryopreservedex vivowhole blood. Cytometry A 2014; 87:157-65. [DOI: 10.1002/cyto.a.22610] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/24/2014] [Accepted: 11/28/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Elisa Nemes
- South African Tuberculosis Vaccine Initiative (SATVI); Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town; Cape Town Western Cape South Africa
| | - Benjamin M. N. Kagina
- South African Tuberculosis Vaccine Initiative (SATVI); Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town; Cape Town Western Cape South Africa
| | - Erica Smit
- South African Tuberculosis Vaccine Initiative (SATVI); Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town; Cape Town Western Cape South Africa
| | - Hadn Africa
- South African Tuberculosis Vaccine Initiative (SATVI); Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town; Cape Town Western Cape South Africa
| | - Marcia Steyn
- South African Tuberculosis Vaccine Initiative (SATVI); Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town; Cape Town Western Cape South Africa
| | - Willem A. Hanekom
- South African Tuberculosis Vaccine Initiative (SATVI); Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town; Cape Town Western Cape South Africa
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative (SATVI); Institute of Infectious Disease and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town; Cape Town Western Cape South Africa
| |
Collapse
|