1
|
Tursi AR, Lages CS, Quayle K, Koenig ZT, Loni R, Eswar S, Cobeña-Reyes J, Thornton S, Tilburgs T, Andorf S. CytoPheno: Automated descriptive cell type naming in flow and mass cytometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.639902. [PMID: 40161808 PMCID: PMC11952469 DOI: 10.1101/2025.03.11.639902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Advances in cytometry have led to increases in the number of cellular markers that are routinely measured. The resulting complexity of the data has prompted a shift from manual to automated analysis methods. Currently, numerous unsupervised methods are available to cluster cells based on marker expression values. However, phenotyping the resulting clusters is typically not part of the automated process. Manually identifying both marker definitions (e.g. CD4+, CCR7+, CD45RA+, CD19-) and descriptive cell type names (e.g. naïve CD4+ T cells) based on marker expression values can be time-consuming, subjective, and error-prone. In this work we propose an algorithm that addresses these problems through the creation of an automated tool, CytoPheno, that assigns marker definitions and cell type names to unidentified clusters. First, post-clustered expression data undergoes per-marker calculations to assign markers as positive or negative. Next, marker names undergo a standardization process to match to Protein Ontology identifier terms. Finally, marker descriptions are matched to cell type names within the Cell Ontology. Each part of the tool was tested with benchmark data to demonstrate performance. Additionally, the tool is encompassed in a graphical user interface (R Shiny) to increase user accessibility and interpretability. Overall, CytoPheno can aid researchers in timely and unbiased phenotyping of post-clustered cytometry data.
Collapse
Affiliation(s)
- Amanda R Tursi
- Department of Biomedical Informatics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Celine S Lages
- Division of Rheumatology, Research Flow Cytometry Core, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kenneth Quayle
- Division of Rheumatology, Research Flow Cytometry Core, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zachary T Koenig
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rashi Loni
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Shruti Eswar
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pharmacology, Physiology & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - José Cobeña-Reyes
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sherry Thornton
- Division of Rheumatology, Research Flow Cytometry Core, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tamara Tilburgs
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sandra Andorf
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
2
|
Ivison S, Boucher G, Zheng G, Garcia RV, Kohen R, Bitton A, Rioux JD, Levings MK. Improving Reliability of Immunological Assays by Defining Minimal Criteria for Cell Fitness. Immunohorizons 2024; 8:622-634. [PMID: 39248805 PMCID: PMC11447670 DOI: 10.4049/immunohorizons.2300095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
Human PBMC-based assays are often used as biomarkers for the diagnosis and prognosis of disease, as well as for the prediction and tracking of response to biological therapeutics. However, the development and use of PBMC-based biomarker assays is often limited by poor reproducibility. Complex immunological assays can be further complicated by variation in cell handling before analysis, especially when using cryopreserved cells. Variation in postthaw viability is further increased if PBMC isolation and cryopreservation are done more than a few hours after collection. There is currently a lack of evidence-based standards for the minimal PBMC viability or "fitness" required to ensure the integrity and reproducibility of immune cell-based assays. In this study, we use an "induced fail" approach to examine the effect of thawed human PBMC fitness on four flow cytometry-based assays. We found that cell permeability-based viability stains at the time of thawing did not accurately quantify cell fitness, whereas a combined measurement of metabolic activity and early apoptosis markers did. Investigation of the impact of different types and levels of damage on PBMC-based assays revealed that only when cells were >60-70% live and apoptosis negative did biomarker values cease to be determined by cell fitness rather than the inherent biology of the cells. These data show that, to reproducibly measure immunological biomarkers using cryopreserved PBMCs, minimal acceptable standards for cell fitness should be incorporated into the assay protocol.
Collapse
Affiliation(s)
- Sabine Ivison
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | | | - Grace Zheng
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Rosa V. Garcia
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Rita Kohen
- McGill University Health Centre, Montreal, Quebec, Canada
| | - Alain Bitton
- McGill University Health Centre, Montreal, Quebec, Canada
| | - John D. Rioux
- Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Megan K. Levings
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
3
|
Waerlop G, Leroux-Roels G, Pagnon A, Begue S, Salaun B, Janssens M, Medaglini D, Pettini E, Montomoli E, Gianchecchi E, Lambe T, Godfrey L, Bull M, Bellamy D, Amdam H, Bredholt G, Cox RJ, Clement F. Proficiency tests to evaluate the impact on assay outcomes of harmonized influenza-specific Intracellular Cytokine Staining (ICS) and IFN-ɣ Enzyme-Linked ImmunoSpot (ELISpot) protocols. J Immunol Methods 2023; 523:113584. [PMID: 37918618 DOI: 10.1016/j.jim.2023.113584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 09/30/2023] [Accepted: 10/28/2023] [Indexed: 11/04/2023]
Abstract
The magnitude and quality of cell-mediated immune responses elicited by natural infection or vaccination are commonly measured by Interferon-ɣ (IFN-ɣ) Enzyme-Linked ImmunoSpot (ELISpot) and Intracellular Cytokine Staining (ICS). To date, laboratories apply a variety of in-house procedures which leads to diverging results, complicates interlaboratory comparisons and hampers vaccine evaluations. During the FLUCOP project, efforts have been made to develop harmonized Standard Operating Procedures (SOPs) for influenza-specific IFN-ɣ ELISpot and ICS assays. Exploratory pilot studies provided information about the interlaboratory variation before harmonization efforts were initiated. Here we report the results of two proficiency tests organized to evaluate the impact of the harmonization effort on assay results and the performance of participating FLUCOP partners. The introduction of the IFN-ɣ ELISpot SOP reduced variation of both background and stimulated responses. Post-harmonization background responses were all lower than an arbitrary threshold of 50 SFU/million cells. When stimulated with A/California and B/Phuket, a statistically significant reduction in variation (p < 0.0001) was observed and CV values were strongly reduced, from 148% to 77% for A/California and from 126% to 73% for B/Phuket. The harmonizing effect of applying an ICS SOP was also confirmed by an increased homogeneity of data obtained by the individual labs. The application of acceptance criteria on cell viability and background responses further enhanced the data homogeneity. Finally, as the same set of samples was analyzed by both the IFN-ɣ ELISpot and the ICS assays, a method comparison was performed. A clear correlation between the two methods was observed, but they cannot be considered interchangeable. In conclusion, proficiency tests show that a limited harmonization effort consisting of the introduction of SOPs and the use of the same in vitro stimulating antigens leads to a reduction of the interlaboratory variation of IFN-ɣ ELISpot data and demonstrate that substantial improvements for the ICS assay are achieved as comparable laboratory datasets could be generated. Additional steps to further reduce the interlaboratory variation of ICS data can consist of standardized gating templates and detailed data reporting instructions as well as further efforts to harmonize reagent and instrument use.
Collapse
Affiliation(s)
- Gwenn Waerlop
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium.
| | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium
| | - Anke Pagnon
- Sanofi, Research Global Immunology, Marcy l'Etoile, France
| | - Sarah Begue
- Sanofi, Research Global Immunology, Marcy l'Etoile, France
| | | | | | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy; VisMederi srl, 53100 Siena, Italy
| | | | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, United Kingdom
| | - Leila Godfrey
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford, UK
| | - Maireid Bull
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, United Kingdom
| | - Duncan Bellamy
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Håkon Amdam
- Influenza Centre, Department of Clinical Science, University of Bergen, N5021 Bergen, Norway
| | - Geir Bredholt
- Influenza Centre, Department of Clinical Science, University of Bergen, N5021 Bergen, Norway
| | - Rebecca Jane Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, N5021 Bergen, Norway
| | - Frédéric Clement
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium
| |
Collapse
|
4
|
Joaquina S, Forcados C, Caulier B, Inderberg EM, Wälchli S. Determination of CAR T cell metabolism in an optimized protocol. Front Bioeng Biotechnol 2023; 11:1207576. [PMID: 37409169 PMCID: PMC10318902 DOI: 10.3389/fbioe.2023.1207576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/12/2023] [Indexed: 07/07/2023] Open
Abstract
Adoptive transfer of T cells modified to express chimeric antigenic receptors (CAR) has emerged as a solution to cure refractory malignancies. However, although CAR T cell treatment of haematological cancers has now shown impressive improvement in outcome, solid tumours have been more challenging to control. The latter type is protected by a strong tumour microenvironment (TME) which might impact cellular therapeutic treatments. Indeed, the milieu around the tumour can become particularly inhibitory to T cells by directly affecting their metabolism. Consequently, the therapeutic cells become physically impeded before being able to attack the tumour. It is therefore extremely important to understand the mechanism behind this metabolic break in order to develop TME-resistant CAR T cells. Historically, the measurement of cellular metabolism has been performed at a low throughput which only permitted a limited number of measurements. However, this has been changed by the introduction of real-time technologies which have lately become more popular to study CAR T cell quality. Unfortunately, the published protocols lack uniformity and their interpretation become confusing. We herein tested the essential parameters to perform a metabolic study on CAR T cells and propose a check list of factors that should be set in order to draw sound conclusion.
Collapse
Affiliation(s)
- Sandy Joaquina
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Christopher Forcados
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Benjamin Caulier
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
- Center for Cancer Cell Reprogramming (CanCell), Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
5
|
Schöllhorn A, Maia A, Kimmerle F, Born J, Rammensee HG, Dimitrov S, Gouttefangeas C. Staining of activated ß 2-integrins in combination with CD137 and CD154 for sensitive identification of functional antigen-specific CD4 + and CD8 + T cells. Front Immunol 2023; 13:1107366. [PMID: 36741378 PMCID: PMC9892897 DOI: 10.3389/fimmu.2022.1107366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
Common flow cytometry-based methods used for functional assessment of antigen-specific T cells rely on de novo expression of intracellular cytokines or cell surface activation induced markers. They come with some limitations such as complex experimental setting, loss of cell viability and often high unspecific background which impairs assay sensitivity. We have previously shown that staining of activated ß2-integrins either with multimers of their ligand ICAM-1 or with a monoclonal antibody can serve as a functional marker detectable on T cells after minutes (CD8+) or few hours (CD4+) of activation. Here, we present a simple method for detection of activated ß2-integrins in combination with established cell surface activation induced markers. We observed that activated ß2-integrins were still detectable after 14 hours of stimulation, allowing their detection together with CD137 and CD154. Combinatorial gating of cells expressing activated ß2-integrins and CD137 or CD154 reduced background in unstimulated samples, increasing the signal-to-noise ratio and allowing improved assessment of low-frequency T cell responses. Extracellular staining of these markers highly correlated with production of intracellular cytokines IL-2, TNF or IFNγ in CD4+ and CD8+ T cells. As an exemplary application, SARS-CoV-2 spike-specific T cell responses were assessed in individuals after COVID-19 vaccination. This method should be useful for epitope discovery projects and for the simultaneous monitoring of low-frequency antigen-specific CD4+ and CD8+ T cell responses in various physiological situations.
Collapse
Affiliation(s)
- Anna Schöllhorn
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Ana Maia
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Felix Kimmerle
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany,German Center for Diabetes Research (DZD), Tübingen, Germany,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen (IDM), Tübingen, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany,Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Stoyan Dimitrov
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany,*Correspondence: Stoyan Dimitrov, ; Cécile Gouttefangeas,
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany,Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany,*Correspondence: Stoyan Dimitrov, ; Cécile Gouttefangeas,
| |
Collapse
|
6
|
Telomerase-based GX301 cancer vaccine in patients with metastatic castration-resistant prostate cancer: a randomized phase II trial. Cancer Immunol Immunother 2021; 70:3679-3692. [PMID: 34351436 PMCID: PMC8571235 DOI: 10.1007/s00262-021-03024-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/25/2021] [Indexed: 12/19/2022]
Abstract
Debate is around the optimal immunization regimen for cancer vaccines since too intense vaccination schedules may exhaust reactive lymphocytes. GX301 is a telomerase-based cancer vaccine whose safety and immunological effects were tested in a phase I trial applying an eight administrations schedule. Main objective of this study was to comparatively analyse safety and immunological response to three GX301 regimens in metastatic castration-resistant prostate cancer patients with response/disease stability after docetaxel chemotherapy. This was a multicentre, randomized, parallel-group, open-label trial registered with EudraCT (2014-000095-26) and ClinicalTrials.gov (NCT02293707, 2014). Ninety-eight patients were randomized to receive either eight (regimen 1), four (regimen 2) or two (regimen 3) vaccine administrations. Sixty-three patients were assessable for the primary immunological end-point. Vaccine-specific immune responses were evaluated by intracellular staining for IFN, elispot and cytotoxic assay at 90 and 180 days from baseline. No major side effects were recorded. A 54% overall immune responder rate was observed with 95% of patients showing at least one vaccine-specific immune response. Rate of immunological responders and number of immunizations were proportionally related, suggesting superiority of regimens 1 and 2 over regimen 3. Overall survival did not differ among regimens in both immunological responders and non-responders and was inversely associated (P = 0.002) with increase in the number of circulating CD8 + T regulatory cells at 180 days. These data indicate that GX301 cancer vaccine is safe and immunogenic in metastatic castration-resistant prostate cancer patients. Schedules with high number of administrations should be preferred in future studies due to their better immunological outcome.
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
|
Price LS, Adamow M, Attig S, Fecci P, Norberg P, Reap E, Janetzki S, McNeil LK. Gating Harmonization Guidelines for Intracellular Cytokine Staining Validated in Second International Multiconsortia Proficiency Panel Conducted by Cancer Immunotherapy Consortium (CIC/CRI). Cytometry A 2020; 99:107-116. [PMID: 33090656 DOI: 10.1002/cyto.a.24244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/21/2020] [Accepted: 10/19/2020] [Indexed: 11/07/2022]
Abstract
Results from the first gating proficiency panel of intracellular cytokine staining (ICS) highlighted the value of using a consensus gating approach to reduce the variability across laboratories in reported %CD8+ or %CD4+ cytokine-positive cells. Based on the data analysis from the first proficiency panel, harmonization guidelines for a consensus gating protocol were proposed. To validate the recommendations from the first panel and to examine factors that were not included in the first panel, a second ICS gating proficiency panel was organized. All participants analyzed the same set of Flow Cytometry Standard (FCS) files using their own gating protocol. An optional learning module was provided to demonstrate how to apply the previously established gating recommendations and harmonization guidelines to actual ICS data files. Eighty-three participants took part in this proficiency panel. The results from this proficiency panel confirmed the harmonization guidelines from the first panel. These recommendations addressed the (1) placement of the cytokine-positive gate, (2) identification of CD4+ CD8+ double-positive T cells, (3) placement of lymphocyte gate, (4) inclusion of dim cells, (5) gate uniformity, and (6) proper adjustment of the biexponential scaling. In addition, based on the results of this proficiency gating panel, two new recommendations were added to expand the harmonization guidelines: (1) inclusion of dump channel marker to gate all live and dump negative cells and (2) backgating to confirm the correct placement of gates across all populations. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Leah S Price
- Bioforum, The Data Masters, CRO, Ness Ziona, Israel
| | - Matthew Adamow
- Memorial Sloan Kettering Cancer Center, New York, New York, 10065, USA
| | - Sebastian Attig
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Department for Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Peter Fecci
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Pamela Norberg
- Duke University Medical Center, Durham, North Carolina, 27710, USA
| | | | | | - Lisa K McNeil
- Elicio Therapeutics, Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
9
|
Generation of TCR-engineered reference cell samples to control T-cell assay performance. Methods Enzymol 2020. [PMID: 31948547 DOI: 10.1016/bs.mie.2019.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
In vitro cellular assays analyzing antigen-specific T cells are characterized by their high complexity and require controlled conditions to lower experimental variations. Without standard cellular reagents, it is difficult to compare results over time and across institutions. To overcome this problem, a simple and robust technology was developed to generate TCR-engineered reference samples (TERS) containing defined numbers of antigen-specific T cells. Utilization of TERS enables performance control of three main T-cell assays: MHC-peptide multimer staining, IFN-γ ELISpot and cytokine flow cytometry. TERS continuously deliver stable results and can be stored for longer periods of time. Here, an optimized manufacturing protocol, based on the electroporation of stable T-cell receptor in vitro-transcribed mRNA, is provided for versatile in-house production of TERS. Included are a guideline to optimize the electroporation settings on locally available electroporation devices and a step-by-step protocol for the production process.
Collapse
|
10
|
Gouttefangeas C, Schuhmacher J, Dimitrov S. Adhering to adhesion: assessing integrin conformation to monitor T cells. Cancer Immunol Immunother 2019; 68:1855-1863. [PMID: 31309255 PMCID: PMC11028104 DOI: 10.1007/s00262-019-02365-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/02/2019] [Indexed: 11/27/2022]
Abstract
Monitoring T cells is of major importance for the development of immunotherapies. Recent sophisticated assays can address particular aspects of the anti-tumor T-cell repertoire or support very large-scale immune screening for biomarker discovery. Robust methods for the routine assessment of the quantity and quality of antigen-specific T cells remain, however, essential. This review discusses selected methods that are commonly used for T-cell monitoring and summarizes the advantages and limitations of these assays. We also present a new functional assay, which specifically detects activated β2 integrins within a very short time following CD8+ T-cell stimulation. Because of its unique and favorable characteristics, this assay could be useful for implementation into our T-cell monitoring toolbox.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany.
| | - Juliane Schuhmacher
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Auf der Morgenstelle 15, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
| | - Stoyan Dimitrov
- Institute of Medical Psychology and Behavioral Neurobiology, Eberhard Karls University, Otfried-Müller Straße 25, 72076, Tübingen, Germany.
- German Center for Diabetes Research, 72076, Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich at the University of Tübingen (IDM), Otfried-Müller Straße 10, 72076, Tübingen, Germany.
| |
Collapse
|
11
|
Wang L, Gong W, Wang S, Neuber B, Sellner L, Schubert ML, Hückelhoven-Krauss A, Kunz A, Gern U, Michels B, Hinkelbein M, Mechler S, Richter P, Müller-Tidow C, Schmitt M, Schmitt A. Improvement of in vitro potency assays by a resting step for clinical-grade chimeric antigen receptor engineered T cells. Cytotherapy 2019; 21:566-578. [PMID: 30910382 DOI: 10.1016/j.jcyt.2019.02.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/01/2019] [Accepted: 02/25/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Chimeric antigen receptor engineered T (CAR-T) cell therapy is a promising approach currently revolutionizing the field of cancer immunotherapy. However, data concerning clinical-grade CAR-T cell stability and functionality after months of cryopreservation have not been released by companies so far. To investigate the effect of cryopreservation on CAR-T cells and to further optimize the potency assays, we performed this study. METHODS A third generation of CD19 CAR-T cells was manufactured according to Good Manufacturing Practice (GMP) requirements, which is applied to patients in an ongoing clinical phase 1 study. Quality control tests for sterility, endotoxin and mycoplasma were performed for each batch. Stability in terms of viability, recovery, transduction efficiency and functional capacity was determined using microscopy, multiparametric flow cytometry as well as chromium-51 release tests. RESULTS Up to 90days of cryopreservation had no influence on viability, recovery and transduction efficiency of CAR-T cells. However, higher cell concentration for cryopreservation could alter the cell viability and recovery but not the transduction efficiency. Moreover, directly after thawing, both the quantity and quality of the functionality of CAR-T cells were transiently hampered by the negative effects of cryopreservation. Notably, the impaired functionality could be fully restored and even strengthened after an overnight resting process. DISCUSSION Cryopreservation is a challenge for the functional activity of CAR-T cells. However, CAR-T cells regain their potency by overnight incubation at 37°C, which mimics the clinical application setting. Therefore, an overnight resting step should be included in in vitro potency assays.
Collapse
Affiliation(s)
- Lei Wang
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Wenjie Gong
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Sanmei Wang
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Leopold Sellner
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases, German Cancer Consortium, Heidelberg, Germany
| | - Maria-Luisa Schubert
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Alexander Kunz
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Ulrike Gern
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Birgit Michels
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Mandy Hinkelbein
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefanie Mechler
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Petra Richter
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases, German Cancer Consortium, Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases, German Cancer Consortium, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
12
|
Hejblum BP, Alkhassim C, Gottardo R, Caron F, Thiébaut R. Sequential Dirichlet process mixtures of multivariate skew $t$-distributions for model-based clustering of flow cytometry data. Ann Appl Stat 2019. [DOI: 10.1214/18-aoas1209] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
13
|
Hilf N, Kuttruff-Coqui S, Frenzel K, Bukur V, Stevanović S, Gouttefangeas C, Platten M, Tabatabai G, Dutoit V, van der Burg SH, Thor Straten P, Martínez-Ricarte F, Ponsati B, Okada H, Lassen U, Admon A, Ottensmeier CH, Ulges A, Kreiter S, von Deimling A, Skardelly M, Migliorini D, Kroep JR, Idorn M, Rodon J, Piró J, Poulsen HS, Shraibman B, McCann K, Mendrzyk R, Löwer M, Stieglbauer M, Britten CM, Capper D, Welters MJP, Sahuquillo J, Kiesel K, Derhovanessian E, Rusch E, Bunse L, Song C, Heesch S, Wagner C, Kemmer-Brück A, Ludwig J, Castle JC, Schoor O, Tadmor AD, Green E, Fritsche J, Meyer M, Pawlowski N, Dorner S, Hoffgaard F, Rössler B, Maurer D, Weinschenk T, Reinhardt C, Huber C, Rammensee HG, Singh-Jasuja H, Sahin U, Dietrich PY, Wick W. Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature 2019; 565:240-245. [PMID: 30568303 DOI: 10.1038/s41586-018-0810-y] [Citation(s) in RCA: 627] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022]
Abstract
Patients with glioblastoma currently do not sufficiently benefit from recent breakthroughs in cancer treatment that use checkpoint inhibitors1,2. For treatments using checkpoint inhibitors to be successful, a high mutational load and responses to neoepitopes are thought to be essential3. There is limited intratumoural infiltration of immune cells4 in glioblastoma and these tumours contain only 30-50 non-synonymous mutations5. Exploitation of the full repertoire of tumour antigens-that is, both unmutated antigens and neoepitopes-may offer more effective immunotherapies, especially for tumours with a low mutational load. Here, in the phase I trial GAPVAC-101 of the Glioma Actively Personalized Vaccine Consortium (GAPVAC), we integrated highly individualized vaccinations with both types of tumour antigens into standard care to optimally exploit the limited target space for patients with newly diagnosed glioblastoma. Fifteen patients with glioblastomas positive for human leukocyte antigen (HLA)-A*02:01 or HLA-A*24:02 were treated with a vaccine (APVAC1) derived from a premanufactured library of unmutated antigens followed by treatment with APVAC2, which preferentially targeted neoepitopes. Personalization was based on mutations and analyses of the transcriptomes and immunopeptidomes of the individual tumours. The GAPVAC approach was feasible and vaccines that had poly-ICLC (polyriboinosinic-polyribocytidylic acid-poly-L-lysine carboxymethylcellulose) and granulocyte-macrophage colony-stimulating factor as adjuvants displayed favourable safety and strong immunogenicity. Unmutated APVAC1 antigens elicited sustained responses of central memory CD8+ T cells. APVAC2 induced predominantly CD4+ T cell responses of T helper 1 type against predicted neoepitopes.
Collapse
Affiliation(s)
- Norbert Hilf
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | | | | | - Stefan Stevanović
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center Partner Site Tübingen, Tübingen, Germany
| | - Cécile Gouttefangeas
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center Partner Site Tübingen, Tübingen, Germany
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
| | - Michael Platten
- University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
- Medical Faculty Mannheim, Mannheim, Germany
| | - Ghazaleh Tabatabai
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center Partner Site Tübingen, Tübingen, Germany
- University Hospital Tübingen, Tübingen, Germany
| | | | - Sjoerd H van der Burg
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
- Leiden University Medical Center, Leiden, The Netherlands
| | - Per Thor Straten
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, University Hospital Herlev, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Hideho Okada
- University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Arie Admon
- Technion - Israel Institute of Technology, Haifa, Israel
| | | | | | - Sebastian Kreiter
- BioNTech AG, Mainz, Germany
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
| | - Andreas von Deimling
- University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | | | | | - Judith R Kroep
- Leiden University Medical Center, Leiden, The Netherlands
| | - Manja Idorn
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, University Hospital Herlev, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jordi Rodon
- Vall d'Hebron University Hospital, Barcelona, Spain
- M. D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | | | | | | | | | | | | | - Monika Stieglbauer
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
| | - Cedrik M Britten
- BioNTech AG, Mainz, Germany
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
- Oncology R&D, GlaxoSmithKline, Stevenage, UK
| | - David Capper
- University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
- Charité, University Medicine Berlin, Berlin, Germany
| | - Marij J P Welters
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
- Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | - Elisa Rusch
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
| | - Lukas Bunse
- University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Colette Song
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | | | | | - Jörg Ludwig
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | - John C Castle
- BioNTech AG, Mainz, Germany
- Agenus Inc., Lexington, KY, USA
| | | | - Arbel D Tadmor
- TRON GmbH - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Mainz, Germany
| | - Edward Green
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
- Medical Faculty Mannheim, Mannheim, Germany
| | | | - Miriam Meyer
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | - Sonja Dorner
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | | | | | | | | | | | - Hans-Georg Rammensee
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center Partner Site Tübingen, Tübingen, Germany
| | | | | | | | - Wolfgang Wick
- University Hospital Heidelberg, Heidelberg, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
14
|
Wang L, Ni M, Hückelhoven-Krauss A, Sellner L, Hoffmann JM, Neuber B, Luft T, Hegenbart U, Schönland S, Kleist C, Sill M, Chen BA, Wuchter P, Eckstein V, Krüger W, Hilgendorf I, Yerushalmi R, Nagler A, Müller-Tidow C, Ho AD, Dreger P, Schmitt M, Schmitt A. Modulation of B Cells and Homing Marker on NK Cells Through Extracorporeal Photopheresis in Patients With Steroid-Refractory/Resistant Graft-Vs.-Host Disease Without Hampering Anti-viral/Anti-leukemic Effects. Front Immunol 2018; 9:2207. [PMID: 30349527 PMCID: PMC6186805 DOI: 10.3389/fimmu.2018.02207] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/05/2018] [Indexed: 11/13/2022] Open
Abstract
Graft-vs.-host disease (GvHD), a severe complication of allogeneic hematopoietic stem cell transplantation, significantly affects the post-transplant morbidity and mortality. Systemic steroids remain the gold standard for the initial management of GvHD. However, up to 60% of patients will not sufficiently respond to steroids. Extracorporeal photopheresis (ECP), a cell-based immunotherapy, has shown good clinical results in such steroid-refractory/resistant GvHD patients. Given its immunomodulatory, but not global immunosuppressive and steroid-sparing capacity, ECP constitutes an attractive option. In the case of GvHD, the balance of immune cells is destroyed: effector cells are not any longer efficiently controlled by regulatory cells. ECP therapy may restore this balance. However, the precise mechanism and the impact of ECP on anti-viral/anti-leukemic function remain unclear. In this study, 839 ECP treatments were performed on patients with acute GvHD (aGvHD) and chronic GvHD (cGvHD). A comprehensive analysis of effector and regulatory cells in patients under ECP therapy included multi-parametric flow cytometry and tetramer staining, LuminexTM-based cytokine, interferon-γ enzyme-linked immunospot, and chromium-51 release assays. Gene profiling of myeloid-derived suppressor cells (MDSCs) was performed by microarray analysis. Immunologically, modulations of effector and regulatory cells as well as proinflammatory cytokines were observed under ECP treatment: (1) GvHD-relevant cell subsets like CD62L+ NK cells and newly defined CD19hiCD20hi B cells were modulated, but (2) quantity and quality of anti-viral/anti-leukemic effector cells were preserved. (3) The development of MDSCs was promoted and switched from an inactivated subset (CD33-CD11b+) to an activated subset (CD33+CD11b+). (4) The frequency of Foxp3+CD4+ regulatory T cells (Tregs) and CD24+CD38hi regulatory B cells was considerably increased in aGvHD patients, and Foxp3+CD8+ Tregs in cGvHD patients. (5) Proinflammatory cytokines like IL-1β, IL-6, IL-8, and TNF-α were significantly reduced. In summary, ECP constitutes an effective immunomodulatory therapy for patients with steroid-refractory/resistant GvHD without impairment of anti-viral/leukemia effects.
Collapse
Affiliation(s)
- Lei Wang
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Ming Ni
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany.,Department of Hematology, the Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | | | - Leopold Sellner
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Jean-Marc Hoffmann
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Thomas Luft
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Ute Hegenbart
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Stefan Schönland
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Christian Kleist
- Department of Nuclear Medicine, University Clinic Heidelberg, Heidelberg, Germany
| | - Martin Sill
- Division Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Bao-An Chen
- Department of Hematology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Patrick Wuchter
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany.,German Red Cross Blood Service, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology Mannheim, Mannheim, Germany
| | - Volker Eckstein
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - William Krüger
- Department of Internal Medicine C, Haematology, Oncology, Stem Cell Transplantation, Palliative Care, University Clinic Greifswald, Greifswald, Germany
| | - Inken Hilgendorf
- Department of Internal Medicine II, University Clinic Jena, Jena, Germany
| | - Ronit Yerushalmi
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Anthony D Ho
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| |
Collapse
|
15
|
Dimitrov S, Gouttefangeas C, Besedovsky L, Jensen ATR, Chandran PA, Rusch E, Businger R, Schindler M, Lange T, Born J, Rammensee HG. Activated integrins identify functional antigen-specific CD8 + T cells within minutes after antigen stimulation. Proc Natl Acad Sci U S A 2018; 115:E5536-E5545. [PMID: 29844168 PMCID: PMC6004473 DOI: 10.1073/pnas.1720714115] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Immediate β2-integrin activation upon T cell receptor stimulation is critical for effective interaction between T cells and their targets and may therefore be used for the rapid identification and isolation of functional T cells. We present a simple and sensitive flow cytometry-based assay to assess antigen-specific T cells using fluorescent intercellular adhesion molecule (ICAM)-1 multimers that specifically bind to activated β2-integrins. The method is compatible with surface and intracellular staining; it is applicable for monitoring of a broad range of virus-, tumor-, and vaccine-specific CD8+ T cells, and for isolating viable antigen-reacting cells. ICAM-1 binding correlates with peptide-MHC multimer binding but, notably, it identifies the fraction of antigen-specific CD8+ T cells with immediate and high functional capability (i.e., expressing high levels of cytotoxic markers and cytokines). Compared with the currently available methods, staining of activated β2-integrins presents the unique advantage of requiring activation times of only several minutes, therefore delivering functional information nearly reflecting the in vivo situation. Hence, the ICAM-1 assay is most suitable for rapid and precise monitoring of functional antigen-specific T cell responses, including for patient samples in a variety of clinical settings, as well as for the isolation of functional T cells for adoptive cell-transfer immunotherapies.
Collapse
Affiliation(s)
- Stoyan Dimitrov
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 72076 Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, 72076 Tübingen, Germany
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany;
| | - Luciana Besedovsky
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72076 Tübingen, Germany
| | - Anja T R Jensen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - P Anoop Chandran
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| | - Elisa Rusch
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| | - Ramona Businger
- Institute of Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Michael Schindler
- Institute of Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Tanja Lange
- Clinic for Rheumatology and Clinical Immunology, University of Lübeck, 23562 Lübeck, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD), 72076 Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, 72076 Tübingen, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany;
- Partner Site Tübingen, German Cancer Consortium (DKTK), 72076 Tübingen, Germany
| |
Collapse
|
16
|
Bidmon N, Kind S, Welters MJP, Joseph-Pietras D, Laske K, Maurer D, Hadrup SR, Schreibelt G, Rae R, Sahin U, Gouttefangeas C, Britten CM, van der Burg SH. Development of an RNA-based kit for easy generation of TCR-engineered lymphocytes to control T-cell assay performance. J Immunol Methods 2018; 458:74-82. [PMID: 29684430 DOI: 10.1016/j.jim.2018.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/03/2018] [Accepted: 04/18/2018] [Indexed: 11/17/2022]
Abstract
Cell-based assays to monitor antigen-specific T-cell responses are characterized by their high complexity and should be conducted under controlled conditions to lower multiple possible sources of assay variation. However, the lack of standard reagents makes it difficult to directly compare results generated in one lab over time and across institutions. Therefore TCR-engineered reference samples (TERS) that contain a defined number of antigen-specific T cells and continuously deliver stable results are urgently needed. We successfully established a simple and robust TERS technology that constitutes a useful tool to overcome this issue for commonly used T-cell immuno-assays. To enable users to generate large-scale TERS, on-site using the most commonly used electroporation (EP) devices, an RNA-based kit approach, providing stable TCR mRNA and an optimized manufacturing protocol were established. In preparation for the release of this immuno-control kit, we established optimal EP conditions on six devices and initiated an extended RNA stability study. Furthermore, we coordinated on-site production of TERS with 4 participants. Finally, a proficiency panel was organized to test the unsupervised production of TERS at different laboratories using the kit approach. The results obtained show the feasibility and robustness of the kit approach for versatile in-house production of cellular control samples.
Collapse
Affiliation(s)
- Nicole Bidmon
- Translational Oncology at the University Medical Center of the Johannes-Gutenberg University Mainz (TRON gGmbH), Freiligrathstraße 12, Mainz 55131, Germany; BioNTech AG, An der Goldgrube 12, 55131 Mainz, Germany
| | - Sonja Kind
- BioNTech AG, An der Goldgrube 12, 55131 Mainz, Germany
| | - Marij J P Welters
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, Leiden, ZA 2333, The Netherlands
| | - Deborah Joseph-Pietras
- ECMC, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton SO16 6YD, United Kingdom
| | - Karoline Laske
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, Tuebingen 72076, Germany
| | - Dominik Maurer
- Immatics biotechnologies GmbH, Paul-Ehrlich-Str. 15, Tuebingen 72076, Germany
| | - Sine Reker Hadrup
- Laboratory of Hematology, University Hospital Herlev, Ringvej 75, Herlev DK-2730, Denmark
| | - Gerty Schreibelt
- Dept. of Tumor Immunology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen, HB 6500, The Netherlands
| | - Richard Rae
- Translational Oncology at the University Medical Center of the Johannes-Gutenberg University Mainz (TRON gGmbH), Freiligrathstraße 12, Mainz 55131, Germany
| | - Ugur Sahin
- Translational Oncology at the University Medical Center of the Johannes-Gutenberg University Mainz (TRON gGmbH), Freiligrathstraße 12, Mainz 55131, Germany; University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, Mainz D-55131, Germany; BioNTech AG, An der Goldgrube 12, 55131 Mainz, Germany
| | - Cécile Gouttefangeas
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, Tuebingen 72076, Germany
| | - Cedrik M Britten
- Translational Oncology at the University Medical Center of the Johannes-Gutenberg University Mainz (TRON gGmbH), Freiligrathstraße 12, Mainz 55131, Germany
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, Leiden, ZA 2333, The Netherlands.
| |
Collapse
|
17
|
Chandran PA, Laske K, Cazaly A, Rusch E, Schmid-Horch B, Rammensee HG, Ottensmeier CH, Gouttefangeas C. Validation of Immunomonitoring Methods for Application in Clinical Studies: The HLA-Peptide Multimer Staining Assay. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2018; 94:342-353. [PMID: 27363684 DOI: 10.1002/cyto.b.21397] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/27/2016] [Accepted: 06/28/2016] [Indexed: 11/11/2022]
Abstract
BACKGROUND Validated assays are essential to generate data with defined specificity, consistency, and reliability. Although the process of validation is required for applying immunoassays in the context of clinical studies, reports on systematic validation of in vitro T cell assays are scarce so far. We recently validated our HLA-peptide multimer staining assay in a systematic manner so as to qualify the method for monitoring antigen-specific T cell responses after immunotherapy. METHODS Parameters of the assay, specificity, precision, linearity, sensitivity, and robustness were assessed systematically. Experiments were designed to address specifically each parameter and are detailed. RESULTS Nonspecific multimer staining was below the acceptance limit of 0.02% multimer(+) CD8(+) cells. The assay showed acceptable precision in all dimensions it was repeated (CV < 10%) and also demonstrated a linear detection (R2 > 0.99) of antigen specific cells. CONCLUSIONS We succeeded in validating the HLA-multimer staining assay in a systematic manner. Additionally, we propose a technical framework and recommendations that can be applied for validating other T cell assessment methods. © 2016 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- P Anoop Chandran
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | - Karoline Laske
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | - Angelica Cazaly
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, United Kingdom
| | - Elisa Rusch
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | | | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | - Christian H Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, United Kingdom
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| |
Collapse
|
18
|
Assay optimisation and technology transfer for multi-site immuno-monitoring in vaccine trials. PLoS One 2017; 12:e0184391. [PMID: 29020010 PMCID: PMC5636064 DOI: 10.1371/journal.pone.0184391] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/23/2017] [Indexed: 11/28/2022] Open
Abstract
Cellular immunological assays are important tools for the monitoring of responses to T-cell-inducing vaccine candidates. As these bioassays are often technically complex and require considerable experience, careful technology transfer between laboratories is critical if high quality, reproducible data that allows comparison between sites, is to be generated. The aim of this study, funded by the European Union Framework Program 7-funded TRANSVAC project, was to optimise Standard Operating Procedures and the technology transfer process to maximise the reproducibility of three bioassays for interferon-gamma responses: enzyme-linked immunosorbent assay (ELISA), ex-vivo enzyme-linked immunospot and intracellular cytokine staining. We found that the initial variability in results generated across three different laboratories reduced following a combination of Standard Operating Procedure harmonisation and the undertaking of side-by-side training sessions in which assay operators performed each assay in the presence of an assay ‘lead’ operator. Mean inter-site coefficients of variance reduced following this training session when compared with the pre-training values, most notably for the ELISA assay. There was a trend for increased inter-site variability at lower response magnitudes for the ELISA and intracellular cytokine staining assays. In conclusion, we recommend that on-site operator training is an essential component of the assay technology transfer process and combined with harmonised Standard Operating Procedures will improve the quality, reproducibility and comparability of data produced across different laboratories. These data may be helpful in ongoing discussions of the potential risk/benefit of centralised immunological assay strategies for large clinical trials versus decentralised units.
Collapse
|
19
|
Kranz LM, Beck JD, Grunwitz C, Hotz C, Vormehr M, Diken M. CIMT 2017: Anniversary symposium - Report on the 15th CIMT Annual Meeting of the Association for Cancer Immunotherapy. Hum Vaccin Immunother 2017; 13:2272-2279. [PMID: 28846471 PMCID: PMC5647989 DOI: 10.1080/21645515.2017.1358327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Lena M Kranz
- a BioNTech RNA Pharmaceuticals GmbH , Mainz , Germany
| | - Jan D Beck
- b TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany
| | | | | | - Mathias Vormehr
- c BioNTech AG , Mainz , Germany.,d Research Center for Immunotherapy (FZI), University Medical Center at the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Mustafa Diken
- b TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany
| |
Collapse
|
20
|
Pedersen NW, Chandran PA, Qian Y, Rebhahn J, Petersen NV, Hoff MD, White S, Lee AJ, Stanton R, Halgreen C, Jakobsen K, Mosmann T, Gouttefangeas C, Chan C, Scheuermann RH, Hadrup SR. Automated Analysis of Flow Cytometry Data to Reduce Inter-Lab Variation in the Detection of Major Histocompatibility Complex Multimer-Binding T Cells. Front Immunol 2017; 8:858. [PMID: 28798746 PMCID: PMC5526901 DOI: 10.3389/fimmu.2017.00858] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/07/2017] [Indexed: 12/22/2022] Open
Abstract
Manual analysis of flow cytometry data and subjective gate-border decisions taken by individuals continue to be a source of variation in the assessment of antigen-specific T cells when comparing data across laboratories, and also over time in individual labs. Therefore, strategies to provide automated analysis of major histocompatibility complex (MHC) multimer-binding T cells represent an attractive solution to decrease subjectivity and technical variation. The challenge of using an automated analysis approach is that MHC multimer-binding T cell populations are often rare and therefore difficult to detect. We used a highly heterogeneous dataset from a recent MHC multimer proficiency panel to assess if MHC multimer-binding CD8+ T cells could be analyzed with computational solutions currently available, and if such analyses would reduce the technical variation across different laboratories. We used three different methods, FLOw Clustering without K (FLOCK), Scalable Weighted Iterative Flow-clustering Technique (SWIFT), and ReFlow to analyze flow cytometry data files from 28 laboratories. Each laboratory screened for antigen-responsive T cell populations with frequency ranging from 0.01 to 1.5% of lymphocytes within samples from two donors. Experience from this analysis shows that all three programs can be used for the identification of high to intermediate frequency of MHC multimer-binding T cell populations, with results very similar to that of manual gating. For the less frequent populations (<0.1% of live, single lymphocytes), SWIFT outperformed the other tools. As used in this study, none of the algorithms offered a completely automated pipeline for identification of MHC multimer populations, as varying degrees of human interventions were needed to complete the analysis. In this study, we demonstrate the feasibility of using automated analysis pipelines for assessing and identifying even rare populations of antigen-responsive T cells and discuss the main properties, differences, and advantages of the different methods tested.
Collapse
Affiliation(s)
- Natasja Wulff Pedersen
- Division of Immunology and Vaccinology, Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - P. Anoop Chandran
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany
| | - Yu Qian
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA, United States
| | - Jonathan Rebhahn
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Nadia Viborg Petersen
- Division of Immunology and Vaccinology, Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Mathilde Dalsgaard Hoff
- Division of Immunology and Vaccinology, Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Scott White
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, United States
| | - Alexandra J. Lee
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA, United States
| | - Rick Stanton
- Human Longevity Inc., San Diego, CA, United States
| | | | | | - Tim Mosmann
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Cécile Gouttefangeas
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, United States
| | - Richard H. Scheuermann
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA, United States
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| | - Sine Reker Hadrup
- Division of Immunology and Vaccinology, Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| |
Collapse
|
21
|
Masucci GV, Cesano A, Hawtin R, Janetzki S, Zhang J, Kirsch I, Dobbin KK, Alvarez J, Robbins PB, Selvan SR, Streicher HZ, Butterfield LH, Thurin M. Validation of biomarkers to predict response to immunotherapy in cancer: Volume I - pre-analytical and analytical validation. J Immunother Cancer 2016; 4:76. [PMID: 27895917 PMCID: PMC5109744 DOI: 10.1186/s40425-016-0178-1] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/20/2016] [Indexed: 12/31/2022] Open
Abstract
Immunotherapies have emerged as one of the most promising approaches to treat patients with cancer. Recently, there have been many clinical successes using checkpoint receptor blockade, including T cell inhibitory receptors such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death-1 (PD-1). Despite demonstrated successes in a variety of malignancies, responses only typically occur in a minority of patients in any given histology. Additionally, treatment is associated with inflammatory toxicity and high cost. Therefore, determining which patients would derive clinical benefit from immunotherapy is a compelling clinical question. Although numerous candidate biomarkers have been described, there are currently three FDA-approved assays based on PD-1 ligand expression (PD-L1) that have been clinically validated to identify patients who are more likely to benefit from a single-agent anti-PD-1/PD-L1 therapy. Because of the complexity of the immune response and tumor biology, it is unlikely that a single biomarker will be sufficient to predict clinical outcomes in response to immune-targeted therapy. Rather, the integration of multiple tumor and immune response parameters, such as protein expression, genomics, and transcriptomics, may be necessary for accurate prediction of clinical benefit. Before a candidate biomarker and/or new technology can be used in a clinical setting, several steps are necessary to demonstrate its clinical validity. Although regulatory guidelines provide general roadmaps for the validation process, their applicability to biomarkers in the cancer immunotherapy field is somewhat limited. Thus, Working Group 1 (WG1) of the Society for Immunotherapy of Cancer (SITC) Immune Biomarkers Task Force convened to address this need. In this two volume series, we discuss pre-analytical and analytical (Volume I) as well as clinical and regulatory (Volume II) aspects of the validation process as applied to predictive biomarkers for cancer immunotherapy. To illustrate the requirements for validation, we discuss examples of biomarker assays that have shown preliminary evidence of an association with clinical benefit from immunotherapeutic interventions. The scope includes only those assays and technologies that have established a certain level of validation for clinical use (fit-for-purpose). Recommendations to meet challenges and strategies to guide the choice of analytical and clinical validation design for specific assays are also provided.
Collapse
Affiliation(s)
- Giuseppe V Masucci
- Department of Oncology-Pathology, Karolinska Institutet, 171 76 Stockholm, Sweden
| | | | - Rachael Hawtin
- Nodality, Inc, 170 Harbor Way, South San Francisco, 94080 CA USA
| | - Sylvia Janetzki
- ZellNet Consulting, Inc, 555 North Avenue, Fort Lee, 07024 NJ USA
| | - Jenny Zhang
- Covaris Inc, 14 Gill St, Woburn, MA 01801 USA
| | - Ilan Kirsch
- Adaptive Biotechnologies, Inc, 1551 Eastlake Ave. E, Seattle, WA 98102 USA
| | - Kevin K Dobbin
- Department of Epidemiology and Biostatistics, College of Public Health, The University of Georgia, 101 Buck Road, Athens, 30602 GA USA
| | - John Alvarez
- Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | | | - Senthamil R Selvan
- Omni Array Biotechnology, 15601 Crabbs Branch Way, Rockville, 20855 MD USA
| | - Howard Z Streicher
- National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, 20892 MD USA
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Magdalena Thurin
- National Cancer Institute, Cancer Diagnosis Program, DCTD, National Institutes of Health, 9609 Medical Center Drive, Bethesda, 20892 MD USA ; Adaptive Biotechnologies, Inc, 1551 Eastlake Ave. E, Seattle, WA 98102 USA
| |
Collapse
|
22
|
Transfer from research/academia to clinical/regulated. Bioanalysis 2016; 8:2169-76. [PMID: 27628669 DOI: 10.4155/bio-2016-4994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We focus here on how the interface in academia has adapted in their approach to assessing the PDs of biological agents to better understand mechanisms at an early stage. This understanding enables drugs to be modified early and to be reassessed before progressing to late stage trials. We discuss how these efforts are now being bolstered by a network of consortia involving industry, academia and regulatory bodies, to bring together resources, knowledge and a harmonization in bioanalytical techniques. We highlight how the regulatory guidance still lags behind the rapid advancement in biologicals and associated analytical techniques, especially in immunotherapies and immunological bioassays. Despite this, new collaborative groups are working together to deliver robust and accurate results essential for identifying the most promising drugs to progress from early phase academic research to late phase industry based trials. We show how the relationship between academia and not-for-profit organizations with large pharma and emerging biotech companies has shifted toward a more collaborative effort in bringing new therapies to the forefront.
Collapse
|
23
|
Wang L, Hückelhoven A, Hong J, Jin N, Mani J, Chen BA, Schmitt M, Schmitt A. Standardization of cryopreserved peripheral blood mononuclear cells through a resting process for clinical immunomonitoring-Development of an algorithm. Cytometry A 2016; 89:246-58. [DOI: 10.1002/cyto.a.22813] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 10/18/2015] [Accepted: 12/11/2015] [Indexed: 01/05/2023]
Affiliation(s)
- Lei Wang
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Angela Hückelhoven
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Jian Hong
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Nan Jin
- Department of Hematology; Zhongda Hospital, Southeast University; Nanjing China
| | - Jiju Mani
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Bao-an Chen
- Department of Hematology; Zhongda Hospital, Southeast University; Nanjing China
| | - Michael Schmitt
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Anita Schmitt
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| |
Collapse
|
24
|
Fenoglio D, Parodi A, Lavieri R, Kalli F, Ferrera F, Tagliamacco A, Guastalla A, Lamperti MG, Giacomini M, Filaci G. Immunogenicity of GX301 cancer vaccine: Four (telomerase peptides) are better than one. Hum Vaccin Immunother 2016; 11:838-50. [PMID: 25714118 PMCID: PMC4514186 DOI: 10.1080/21645515.2015.1012032] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Peptide540–548, peptide611–626, peptide672–686 and peptide766–780, which are derived from human telomerase, constitute the immunogenic component of the GX301 cancer vaccine. The relative immunogenicity of these peptides is unknown, thus it is unsure whether their combined use offers real advantages over single peptide stimulation. Hence, this study compared the number of specific immune responses and responders to each peptide, as well as to their mixture (meaning the co-presence of the 4 peptides in the same culture well), achieved after ex vivo stimulation of PBMC from 21, HLA-A2+ (n.11) or HLA-A2- (n.10), healthy donors. The study was performed on freshly collected PBMC (T0) and on PBMC stimulated for 10 d with single peptides or their mixture (T1). Peptide-specific immune responses were analyzed by Elispot and cytokine intracellular staining by flow cytometry. The results showed that each peptide induced specific immune responses in some subjects, with different panels of responders among the peptides. Moreover, the numbers of responses and responders to the single peptides or their mixture were comparable. Importantly, the overall number of responders to the 4 peptides was higher than to each single peptide, or to their mixture, both at T0 and T1. These data demonstrate the immunogenicity of each of the 4 GX301 telomerase peptides. Moreover, they show the advantage of multi-peptide over single peptide stimulation, providing a clear support to their combined administration in vaccination protocols. However, the data pose a warning against peptide administration as a mixture due to possible interference phenomena during antigen presentation processes.
Collapse
Affiliation(s)
- Daniela Fenoglio
- a Centre of Excellence for Biomedical Research ; University of Genoa ; Genoa , Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mandruzzato S, Brandau S, Britten CM, Bronte V, Damuzzo V, Gouttefangeas C, Maurer D, Ottensmeier C, van der Burg SH, Welters MJP, Walter S. Toward harmonized phenotyping of human myeloid-derived suppressor cells by flow cytometry: results from an interim study. Cancer Immunol Immunother 2016; 65:161-9. [PMID: 26728481 PMCID: PMC4726716 DOI: 10.1007/s00262-015-1782-5] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/12/2015] [Indexed: 01/02/2023]
Abstract
There is an increasing interest for monitoring circulating myeloid-derived suppressor cells (MDSCs) in cancer patients, but there are also divergences in their phenotypic definition. To overcome this obstacle, the Cancer Immunoguiding Program under the umbrella of the Association of Cancer Immunotherapy is coordinating a proficiency panel program that aims at harmonizing MDSC phenotyping. After a consultation period, a two-stage approach was designed to harmonize MDSC phenotype. In the first step, an international consortium of 23 laboratories immunophenotyped 10 putative MDSC subsets on pretested, peripheral blood mononuclear cells of healthy donors to assess the level of concordance and define robust marker combinations for the identification of circulating MDSCs. At this stage, no mandatory requirements to standardize reagents or protocols were introduced. Data analysis revealed a small intra-laboratory, but very high inter-laboratory variance for all MDSC subsets, especially for the granulocytic subsets. In particular, the use of a dead-cell marker altered significantly the reported percentage of granulocytic MDSCs, confirming that these cells are especially sensitive to cryopreservation and/or thawing. Importantly, the gating strategy was heterogeneous and associated with high inter-center variance. Overall, our results document the high variability in MDSC phenotyping in the multicenter setting if no harmonization/standardization measures are applied. Although the observed variability depended on a number of identified parameters, the main parameter associated with variation was the gating strategy. Based on these findings, we propose further efforts to harmonize marker combinations and gating parameters to identify strategies for a robust enumeration of MDSC subsets.
Collapse
Affiliation(s)
- Susanna Mandruzzato
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64, 35128, Padua, Italy.
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy.
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Cedrik M Britten
- TRON Translationale Onkologie an der Universitätsmedizin der Johannes Gutenberg-Universität Mainz GmbH, Mainz, Germany
- Cell Therapy Group, Immuno-Oncology and Combinations, GlaxoSmithKline, Stevenage, UK
| | - Vincenzo Bronte
- Section of Immunology, Department of Pathology and Diagnostics, Verona University Hospital, Verona, Italy
| | - Vera Damuzzo
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64, 35128, Padua, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | | | - Christian Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, Experimental Cancer Medicine Centre, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, UK
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marij J P Welters
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
26
|
BayesFlow: latent modeling of flow cytometry cell populations. BMC Bioinformatics 2016; 17:25. [PMID: 26755197 PMCID: PMC4709953 DOI: 10.1186/s12859-015-0862-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 12/17/2015] [Indexed: 12/23/2022] Open
Abstract
Background Flow cytometry is a widespread single-cell measurement technology with a multitude of clinical and research applications. Interpretation of flow cytometry data is hard; the instrumentation is delicate and can not render absolute measurements, hence samples can only be interpreted in relation to each other while at the same time comparisons are confounded by inter-sample variation. Despite this, most automated flow cytometry data analysis methods either treat samples individually or ignore the variation by for example pooling the data. A key requirement for models that include multiple samples is the ability to visualize and assess inferred variation, since what could be technical variation in one setting would be different phenotypes in another. Results We introduce BayesFlow, a pipeline for latent modeling of flow cytometry cell populations built upon a Bayesian hierarchical model. The model systematizes variation in location as well as shape. Expert knowledge can be incorporated through informative priors and the results can be supervised through compact and comprehensive visualizations. BayesFlow is applied to two synthetic and two real flow cytometry data sets. For the first real data set, taken from the FlowCAP I challenge, BayesFlow does not only give a gating which would place it among the top performers in FlowCAP I for this dataset, it also gives a more consistent treatment of different samples than either manual gating or other automated gating methods. The second real data set contains replicated flow cytometry measurements of samples from healthy individuals. BayesFlow gives here cell populations with clear expression patterns and small technical intra-donor variation as compared to biological inter-donor variation. Conclusions Modeling latent relations between samples through BayesFlow enables a systematic analysis of inter-sample variation. As opposed to other joint gating methods, effort is put at ensuring that the obtained partition of the data corresponds to actual cell populations, and the result is therefore directly biologically interpretable. BayesFlow is freely available at GitHub. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0862-z) contains supplementary material, which is available to authorized users.
Collapse
|
27
|
Santegoets SJAM, Dijkgraaf EM, Battaglia A, Beckhove P, Britten CM, Gallimore A, Godkin A, Gouttefangeas C, de Gruijl TD, Koenen HJPM, Scheffold A, Shevach EM, Staats J, Taskén K, Whiteside TL, Kroep JR, Welters MJP, van der Burg SH. Monitoring regulatory T cells in clinical samples: consensus on an essential marker set and gating strategy for regulatory T cell analysis by flow cytometry. Cancer Immunol Immunother 2015; 64:1271-86. [PMID: 26122357 PMCID: PMC4554737 DOI: 10.1007/s00262-015-1729-x] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/30/2015] [Indexed: 12/18/2022]
Abstract
Regulatory T cell (Treg)-mediated immunosuppression is considered a major obstacle for successful cancer immunotherapy. The association between clinical outcome and Tregs is being studied extensively in clinical trials, but unfortunately, no consensus has been reached about (a) the markers and (b) the gating strategy required to define human Tregs in this context, making it difficult to draw final conclusions. Therefore, we have organized an international workshop on the detection and functional testing of Tregs with leading experts in the field, and 40 participants discussing different analyses and the importance of different markers and context in which Tregs were analyzed. This resulted in a rationally composed ranking list of "Treg markers". Subsequently, the proposed Treg markers were tested to get insight into the overlap/differences between the most frequently used Treg definitions and their utility for Treg detection in various human tissues. Here, we conclude that the CD3, CD4, CD25, CD127, and FoxP3 markers are the minimally required markers to define human Treg cells. Staining for Ki67 and CD45RA showed to provide additional information on the activation status of Tregs. The use of markers was validated in a series of PBMC from healthy donors and cancer patients, as well as in tumor-draining lymph nodes and freshly isolated tumors. In conclusion, we propose an essential marker set comprising antibodies to CD3, CD4, CD25, CD127, Foxp3, Ki67, and CD45RA and a corresponding robust gating strategy for the context-dependent analysis of Tregs by flow cytometry.
Collapse
Affiliation(s)
- Saskia J A M Santegoets
- Department of Clinical Oncology, Leiden University Medical Center (LUMC), Leiden, The Netherlands,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Janetzki S, Price L, Schroeder H, Britten CM, Welters MJP, Hoos A. Guidelines for the automated evaluation of Elispot assays. Nat Protoc 2015; 10:1098-115. [PMID: 26110715 DOI: 10.1038/nprot.2015.068] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The presented protocol for Elispot plate evaluation summarizes how to implement the recommendations developed following the establishment of a large-scale international Elispot plate-reading panel and subsequent multistep consensus-finding process. The panel involved >100 scientists from various immunological backgrounds. The protocol includes the description and justification of steps for setting reading parameters to obtain accurate, reliable and precise automated analysis results of Elispot plates. Further, necessary adjustments for out-of-specification situations are described and examples are provided. The plate analysis, including parameter adjustments, auditing of results and necessary annotations, should be achievable within a time range of 10-30 min per plate. Adoption of these guidelines should enable a further reduction in assay variability and an increase in the reliability and comparability of results obtained by Elispot. These guidelines conclude the ongoing harmonization efforts for the enzymatic Elispot assay.
Collapse
Affiliation(s)
| | - Leah Price
- LBPrice Statistical Consulting Ltd., Karmiel, Israel
| | | | | | - Marij J P Welters
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Axel Hoos
- Department of ImmunoOncology, GlaxoSmith Kline, Collegeville, Pennsylvania, USA
| |
Collapse
|
29
|
White S, Laske K, Welters MJ, Bidmon N, van der Burg SH, Britten CM, Enzor J, Staats J, Weinhold KJ, Gouttefangeas C, Chan C. Managing Multi-center Flow Cytometry Data for Immune Monitoring. Cancer Inform 2015; 13:111-22. [PMID: 26085786 PMCID: PMC4463798 DOI: 10.4137/cin.s16346] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/19/2014] [Accepted: 11/21/2014] [Indexed: 12/17/2022] Open
Abstract
With the recent results of promising cancer vaccines and immunotherapy1–5, immune monitoring has become increasingly relevant for measuring treatment-induced effects on T cells, and an essential tool for shedding light on the mechanisms responsible for a successful treatment. Flow cytometry is the canonical multi-parameter assay for the fine characterization of single cells in solution, and is ubiquitously used in pre-clinical tumor immunology and in cancer immunotherapy trials. Current state-of-the-art polychromatic flow cytometry involves multi-step, multi-reagent assays followed by sample acquisition on sophisticated instruments capable of capturing up to 20 parameters per cell at a rate of tens of thousands of cells per second. Given the complexity of flow cytometry assays, reproducibility is a major concern, especially for multi-center studies. A promising approach for improving reproducibility is the use of automated analysis borrowing from statistics, machine learning and information visualization21–23, as these methods directly address the subjectivity, operator-dependence, labor-intensive and low fidelity of manual analysis. However, it is quite time-consuming to investigate and test new automated analysis techniques on large data sets without some centralized information management system. For large-scale automated analysis to be practical, the presence of consistent and high-quality data linked to the raw FCS files is indispensable. In particular, the use of machine-readable standard vocabularies to characterize channel metadata is essential when constructing analytic pipelines to avoid errors in processing, analysis and interpretation of results. For automation, this high-quality metadata needs to be programmatically accessible, implying the need for a consistent Application Programming Interface (API). In this manuscript, we propose that upfront time spent normalizing flow cytometry data to conform to carefully designed data models enables automated analysis, potentially saving time in the long run. The ReFlow informatics framework was developed to address these data management challenges.
Collapse
Affiliation(s)
- Scott White
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham NC, USA
| | - Karoline Laske
- Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Marij Jp Welters
- Experimental Cancer Immunology and Therapy, Department of Clinical Oncology (K1-P), Leiden University Medical Center, Leiden, the Netherlands
| | - Nicole Bidmon
- Translational Oncology at the University Medical Center of the Johannes-Gutenberg University gGmbH, Mainz, Germany
| | - Sjoerd H van der Burg
- Experimental Cancer Immunology and Therapy, Department of Clinical Oncology (K1-P), Leiden University Medical Center, Leiden, the Netherlands
| | - Cedrik M Britten
- Translational Oncology at the University Medical Center of the Johannes-Gutenberg University gGmbH, Mainz, Germany
| | - Jennifer Enzor
- Sr. Research Analyst, Flow Cytometry Core Facility, Center for AIDS Research, Duke University Medical Center, Durham, NC, USA
| | - Janet Staats
- Scientific/Research Laboratory Manager, Flow Cytometry Core Facility, Center for AIDS Research, Duke University Medical Center, Durham, NC, USA
| | - Kent J Weinhold
- Joseph W. and Dorothy W. Beard Professor of Surgery, Chief, Division of Surgical Sciences, Professor of Immunology and Pathology, Director, Duke Center for AIDS Research (CFAR), Duke University Medical Center, Durham, NC, USA
| | | | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham NC, USA
| |
Collapse
|
30
|
Gouttefangeas C, Chan C, Attig S, Køllgaard TT, Rammensee HG, Stevanović S, Wernet D, thor Straten P, Welters MJP, Ottensmeier C, van der Burg SH, Britten CM. Data analysis as a source of variability of the HLA-peptide multimer assay: from manual gating to automated recognition of cell clusters. Cancer Immunol Immunother 2015; 64:585-98. [PMID: 25854580 PMCID: PMC4528367 DOI: 10.1007/s00262-014-1649-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 12/18/2014] [Indexed: 11/30/2022]
Abstract
Multiparameter flow cytometry is an indispensable method for assessing antigen-specific T cells in basic research and cancer immunotherapy. Proficiency panels have shown that cell sample processing, test protocols and data analysis may all contribute to the variability of the results obtained by laboratories performing ex vivo T cell immune monitoring. In particular, analysis currently relies on a manual, step-by-step strategy employing serial gating decisions based on visual inspection of one- or two-dimensional plots. It is therefore operator dependent and subjective. In the context of continuing efforts to support inter-laboratory T cell assay harmonization, the CIMT Immunoguiding Program organized its third proficiency panel dedicated to the detection of antigen-specific CD8(+) T cells by HLA-peptide multimer staining. We first assessed the contribution of manual data analysis to the variability of reported T cell frequencies within a group of laboratories staining and analyzing the same cell samples with their own reagents and protocols. The results show that data analysis is a source of variation in the multimer assay outcome. To evaluate whether an automated analysis approach can reduce variability of proficiency panel data, we used a hierarchical statistical mixture model to identify cell clusters. Challenges for automated analysis were the need to process non-standardized data sets from multiple centers, and the fact that the antigen-specific cell frequencies were very low in most samples. We show that this automated method can circumvent difficulties inherent to manual gating strategies and is broadly applicable for experiments performed with heterogeneous protocols and reagents.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, Auf der Morgenstelle 15, 72076, Tübingen, Germany,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Fenstermaker RA, Ciesielski MJ. Challenges in the development of a survivin vaccine (SurVaxM) for malignant glioma. Expert Rev Vaccines 2014; 13:377-85. [PMID: 24521310 DOI: 10.1586/14760584.2014.881255] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There is growing interest in immunotherapy for malignant gliomas. This interest stems from a number of immunological observations, together with the failure of conventional therapeutic agents to produce broad and clinically meaningful improvements in survival and quality of life. The challenges faced in translating laboratory-based immunological observations to Phase I and II clinical trials for immunotherapy of gliomas are substantial. Nevertheless, as our understanding of the effects of active specific vaccination in glioma patients grows, results support optimism that such methods may eventually prove useful as an adjunctive treatment for these cancers. This paper highlights a number of barriers encountered in the translational development of a survivin-targeted peptide vaccine (SurVaxM) for patients with malignant gliomas.
Collapse
Affiliation(s)
- Robert A Fenstermaker
- Department of Neurosurgery, Roswell Park Cancer Institute and State University of New York School of Medicine and Biomedical Sciences, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | |
Collapse
|
32
|
Derhovanessian E, Maier AB, Hähnel K, McElhaney JE, Slagboom EP, Pawelec G. Latent infection with cytomegalovirus is associated with poor memory CD4 responses to influenza A core proteins in the elderly. THE JOURNAL OF IMMUNOLOGY 2014; 193:3624-31. [PMID: 25187662 DOI: 10.4049/jimmunol.1303361] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Influenza remains a major pathogen in older people. Infection with CMV and the accumulation of late-differentiated T cells associated with it have been implicated in poor Ab responsiveness to influenza vaccination in the elderly, most of whom are CMV positive. However, whether CMV infection also affects memory T cell responses to influenza remains unknown. To investigate this, we assessed T cell responses to influenza A matrix protein and nucleoprotein ex vivo in 166 Dutch individuals (mean age 62.2 y, range 42-82) and validated the results in a second cohort from North America (mean age 73.1 y, range 65-81, n = 28). We found that less than half of the CMV-infected older subjects mounted a CD4 T cell response to influenza Ags, whereas ∼80% of uninfected elderly did so. A similar proportion of younger subjects possessed influenza A virus-responsive CD4 T cells, and, interestingly, this was the case whether they were CMV-infected. Thus, the effect of CMV was only seen in the older donors, who may have been exposed to the virus for decades. The percentage of donors with CD8 responses to influenza A virus was lower than those with CD4; this was not influenced by whether the subjects were CMV seropositive or seronegative. CMV-seropositive responders had significantly higher frequencies of late-differentiated CD4 T-cells (CD45RA(+/-)CCR7(-)CD27(-)CD28(-)) compared with CMV-infected nonresponders. These data add to the accumulating evidence that infection with CMV has profound but heterogeneous effects on responses to the products of other viruses and have implications for the design of influenza vaccines, especially in the elderly.
Collapse
Affiliation(s)
- Evelyna Derhovanessian
- Department of Internal Medicine II, Centre for Medical Research, University of Tubingen, 72072 Tubingen, Germany;
| | - Andrea B Maier
- Department of Gerontology and Geriatrics, Leiden University Medical Centre, 2333 ZC Leiden, the Netherlands; Netherlands Consortium for Health Aging, Leiden University Medical Centre, 2333 ZC Leiden, the Netherlands; Department of Internal Medicine, Section of Gerontology and Geriatrics, VU University Medical Centre, 1081 HV Amsterdam, the Netherlands
| | - Karin Hähnel
- Department of Internal Medicine II, Centre for Medical Research, University of Tubingen, 72072 Tubingen, Germany
| | - Janet E McElhaney
- Advanced Medical Research Institute of Canada, Sudbury, Ontario, Canada P3E 5J1; and
| | - Eline P Slagboom
- Netherlands Consortium for Health Aging, Leiden University Medical Centre, 2333 ZC Leiden, the Netherlands; Section of Molecular Epidemiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Graham Pawelec
- Department of Internal Medicine II, Centre for Medical Research, University of Tubingen, 72072 Tubingen, Germany
| |
Collapse
|
33
|
Immune monitoring in cancer vaccine clinical trials: critical issues of functional flow cytometry-based assays. BIOMED RESEARCH INTERNATIONAL 2013; 2013:726239. [PMID: 24195078 PMCID: PMC3806162 DOI: 10.1155/2013/726239] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/19/2013] [Indexed: 11/17/2022]
Abstract
The development of immune monitoring assays is essential to determine the immune responses against tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs) and their possible correlation with clinical outcome in cancer patients receiving immunotherapies. Despite the wide range of techniques used, to date these assays have not shown consistent results among clinical trials and failed to define surrogate markers of clinical efficacy to antitumor vaccines. Multiparameter flow cytometry- (FCM-) based assays combining different phenotypic and functional markers have been developed in the past decade for informative and longitudinal analysis of polyfunctional T-cells. These technologies were designed to address the complexity and functional heterogeneity of cancer biology and cellular immunity and to define biomarkers predicting clinical response to anticancer treatment. So far, there is still a lack of standardization of some of these immunological tests. The aim of this review is to overview the latest technologies for immune monitoring and to highlight critical steps involved in some of the FCM-based cellular immune assays. In particular, our laboratory is focused on melanoma vaccine research and thus our main goal was the validation of a functional multiparameter test (FMT) combining different functional and lineage markers to be applied in clinical trials involving patients with melanoma.
Collapse
|
34
|
Britten CM, Walter S, Janetzki S. Immunological Monitoring to Rationally Guide AAV Gene Therapy. Front Immunol 2013; 4:273. [PMID: 24062741 PMCID: PMC3770921 DOI: 10.3389/fimmu.2013.00273] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 08/27/2013] [Indexed: 12/12/2022] Open
Abstract
Recent successes with adeno-associated virus (AAV)-based gene therapies fuel the hope for new treatments for hereditary diseases. Pre-existing as well as therapy-induced immune responses against both AAV and the encoded transgenes have been described and may impact on safety and efficacy of gene therapy approaches. Consequently, monitoring of vector- and transgene-specific immunity is mandated and may rationally guide clinical development. Next to the humoral immune response, the cellular response is central in our understanding of the host reaction in gene therapy. But in contrast to the monitoring of antibodies, which has matured over many decades, sensitive and robust monitoring of T cells is a relatively new development. To make cellular immune assessments fit for purpose, investigators need to know, control and report the critical assay variables that influence the results. In addition, the quality of immune assays needs to be continuously adjusted to allow for exploratory hypothesis generation in early stages and confirmatory hypothesis validation in later stages of clinical development. The concept of immune assay harmonization which includes use of field-wide benchmarks, harmonization guidelines, and external quality control can support the context-specific evolution of immune assays. Multi-center studies pose particular challenges to sample logistics and quality control of sample specimens. Cooperative groups need to define if immune assessments should be performed in one central facility, in peripheral labs or including a combination of both. Finally, engineered reference samples that contain a defined number of antigen-specific T cells may become broadly applicable tools to control assay performance over time or across institutions.
Collapse
Affiliation(s)
- Cedrik Michael Britten
- Translational Oncology, University Medical Center, Johannes Gutenberg-University Mainz (TRON gGmbH) , Mainz , Germany ; Association for Cancer Immunotherapy (CIMT) , Mainz , Germany
| | | | | |
Collapse
|
35
|
McNeil LK, Price L, Britten CM, Jaimes M, Maecker H, Odunsi K, Matsuzaki J, Staats JS, Thorpe J, Yuan J, Janetzki S. A harmonized approach to intracellular cytokine staining gating: Results from an international multiconsortia proficiency panel conducted by the Cancer Immunotherapy Consortium (CIC/CRI). Cytometry A 2013; 83:728-38. [PMID: 23788464 DOI: 10.1002/cyto.a.22319] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 04/18/2013] [Accepted: 05/17/2013] [Indexed: 11/06/2022]
Abstract
Previous results from two proficiency panels of intracellular cytokine staining (ICS) from the Cancer Immunotherapy Consortium and panels from the National Institute of Allergy and Infectious Disease and the Association for Cancer Immunotherapy highlight the variability across laboratories in reported % CD8+ or % CD4+ cytokine-positive cells. One of the main causes of interassay variability in flow cytometry-based assays is due to differences in gating strategies between laboratories, which may prohibit the generation of robust results within single centers and across institutions. To study how gating strategies affect the variation in reported results, a gating panel was organized where all participants analyzed the same set of Flow Cytometry Standard (FCS) files from a four-color ICS assay using their own gating protocol (Phase I) and a gating protocol drafted by consensus from the organizers of the panel (Phase II). Focusing on analysis removed donor, assay, and instrument variation, enabling us to quantify the variability caused by gating alone. One hundred ten participating laboratories applied 110 different gating approaches. This led to high variability in the reported percentage of cytokine-positive cells and consequently in response detection in Phase I. However, variability was dramatically reduced when all laboratories used the same gating strategy (Phase II). Proximity of the cytokine gate to the negative population most impacted true-positive and false-positive response detection. Recommendations are provided for the (1) placement of the cytokine-positive gate, (2) identification of CD4+ CD8+ double-positive T cells, (3) placement of lymphocyte gate, (4) inclusion of dim cells, (5) gate uniformity, and 6) proper adjustment of the biexponential scaling.
Collapse
|
36
|
Singh SK, Tummers B, Schumacher TN, Gomez R, Franken KLMC, Verdegaal EM, Laske K, Gouttefangeas C, Ottensmeier C, Welters MJP, Britten CM, van der Burg SH. The development of standard samples with a defined number of antigen-specific T cells to harmonize T cell assays: a proof-of-principle study. Cancer Immunol Immunother 2013; 62:489-501. [PMID: 22986454 PMCID: PMC3589624 DOI: 10.1007/s00262-012-1351-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 09/02/2012] [Indexed: 11/05/2022]
Abstract
The validation of assays that quantify antigen-specific T cell responses is critically dependent on cell samples that contain clearly defined measurable numbers of antigen-specific T cells. An important requirement is that such cell samples are handled and analyzed in a comparable fashion to peripheral blood mononuclear cells (PBMC). We performed a proof-of-principle study to show that retrovirally TCR-transduced T cells spiked at defined numbers in autologous PBMC can be used as standard samples for HLA/peptide multimer staining. NY-ESO-1157-165-specific, TCR-transduced CD8+ T cell batches were successfully generated from PBMC of several HLA-A*0201 healthy donors, purified by magnetic cell sorting on the basis of HLA tetramer (TM) staining and expanded with specific antigen in vitro. When subsequently spiked into autologous PBMC, the detection of these CD3+CD8+TM+ T cells was highly accurate with a mean accuracy of 91.6 %. The standard cells can be preserved for a substantial period of time in liquid nitrogen. Furthermore, TM staining of fresh and cryopreserved standard samples diluted at decreasing concentrations into autologous cryopreserved unspiked PBMC revealed that the spiked CD3+CD8+TM+ T cells could be accurately detected at all dilutions in a linear fashion with a goodness-of-fit of over 0.99 at a frequency of at least 0.02 % among the CD3+CD8+ T cell population. Notably, the CD3+CD8+TM+ cells of the standard samples were located exactly within the gates used to analyze patient samples and displayed a similar scatter pattern. The performance of the cryopreserved standard samples in the hands of 5 external investigators was good with an inter-laboratory variation of 32.9 % and the doubtless identification of one outlier.
Collapse
Affiliation(s)
- Satwinder Kaur Singh
- Department of Clinical Oncology, Leiden University Medical Center, Building 1, K1-P, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Bart Tummers
- Department of Clinical Oncology, Leiden University Medical Center, Building 1, K1-P, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Ton N. Schumacher
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Raquel Gomez
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kees L. M. C. Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Els M. Verdegaal
- Department of Clinical Oncology, Leiden University Medical Center, Building 1, K1-P, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Karoline Laske
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | | | | | - Marij J. P. Welters
- Department of Clinical Oncology, Leiden University Medical Center, Building 1, K1-P, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Cedrik M. Britten
- Department of the Translational Oncology, University Medical Center of the Johannes-Gutenberg-University, Mainz, Germany
| | - Sjoerd H. van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Building 1, K1-P, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
37
|
Bedke J, Gouttefangeas C, Singh-Jasuja H, Stevanović S, Behnes CL, Stenzl A. Targeted therapy in renal cell carcinoma: moving from molecular agents to specific immunotherapy. World J Urol 2013; 32:31-8. [PMID: 23404195 PMCID: PMC3901931 DOI: 10.1007/s00345-013-1033-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/24/2013] [Indexed: 12/22/2022] Open
Abstract
Non-specific immunotherapy has been for a long time a standard treatment option for patients with metastatic renal cell carcinoma but was redeemed by specific targeted molecular therapies, namely the VEGF and mTOR inhibitors. After moving treatment for mRCC to specific molecular agents with a well-defined mode of action, immunotherapy still needs this further development to increase its accuracy. Nowadays, an evolution from a rather non-specific cytokine treatment to sophisticated targeted approaches in specific immunotherapy led to a re-launch of immunotherapy in clinical studies. Recent steps in the development of immunotherapy strategies are discussed in this review with a special focus on peptide vaccination which aims at a tumor targeting by specific T lymphocytes. In addition, different combinatory strategies with immunomodulating agents like cyclophosphamide or sunitinib are outlined, and the effects of immune checkpoint modulators as anti-CTLA-4 or PD-1 antibodies are discussed.
Collapse
Affiliation(s)
- Jens Bedke
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany,
| | | | | | | | | | | |
Collapse
|