1
|
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
|
2
|
Gorovits B, Azadeh M, Buchlis G, Fiscella M, Harrison T, Havert M, Janetzki S, Jawa V, Long B, Mahnke YD, McDermott A, Milton M, Nelson R, Vettermann C, Wu B. Evaluation of Cellular Immune Response to Adeno-Associated Virus-Based Gene Therapy. AAPS J 2023; 25:47. [PMID: 37101079 PMCID: PMC10132926 DOI: 10.1208/s12248-023-00814-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/14/2023] [Indexed: 04/28/2023] Open
Abstract
The number of approved or investigational late phase viral vector gene therapies (GTx) has been rapidly growing. The adeno-associated virus vector (AAV) technology continues to be the most used GTx platform of choice. The presence of pre-existing anti-AAV immunity has been firmly established and is broadly viewed as a potential deterrent for successful AAV transduction with a possibility of negative impact on clinical efficacy and a connection to adverse events. Recommendations for the evaluation of humoral, including neutralizing and total antibody based, anti-AAV immune response have been presented elsewhere. This manuscript aims to cover considerations related to the assessment of anti-AAV cellular immune response, including review of correlations between humoral and cellular responses, potential value of cellular immunogenicity assessment, and commonly used analytical methodologies and parameters critical for monitoring assay performance. This manuscript was authored by a group of scientists involved in GTx development who represent several pharma and contract research organizations. It is our intent to provide recommendations and guidance to the industry sponsors, academic laboratories, and regulatory agencies working on AAV-based GTx viral vector modalities with the goal of achieving a more consistent approach to anti-AAV cellular immune response assessment.
Collapse
Affiliation(s)
| | - Mitra Azadeh
- Ultragenyx Pharmaceutical Inc, Novato, California, USA
| | - George Buchlis
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Mike Havert
- Gene Therapy Partners, San Diego, California, USA
| | | | - Vibha Jawa
- Bristol Myers Squibb Pharmaceutical, Princeton, New Jersey, USA
| | - Brian Long
- BioMarin Pharmaceutical Inc, Novato, California, USA
| | | | - Andrew McDermott
- Labcorp Early Development Laboratories Inc, Indianapolis, Indiana, USA
| | - Mark Milton
- Lake Boon Pharmaceutical Consulting LLC, Hudson, New York, USA
| | | | | | - Bonnie Wu
- Janssen Pharmaceuticals, Raritan, New Jersey, USA
| |
Collapse
|
3
|
Using chimeric antigen receptor T-cell therapy to fight glioblastoma multiforme: past, present and future developments. J Neurooncol 2021; 156:81-96. [PMID: 34825292 PMCID: PMC8714623 DOI: 10.1007/s11060-021-03902-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022]
Abstract
Introduction Glioblastoma multiforme (GBM) constitutes one of the deadliest tumors to afflict humans, although it is still considered an orphan disease. Despite testing multiple new and innovative therapies in ongoing clinical trials, the median survival for this type of malignancy is less than two years after initial diagnosis, regardless of therapy. One class of promising new therapies are chimeric antigen receptor T cells or CAR-T which have been shown to be very effective at treating refractory liquid tumors such as B-cell malignancies. However, CAR-T effectivity against solid tumors such as GBM has been limited thus far. Methods A Pubmed, Google Scholar, Directory of Open Access Journals, and Web of Science literature search using the terms chimeric antigen receptor or CAR-T, GBM, solid tumor immunotherapy, immunotherapy, and CAR-T combination was performed for publication dates between January 1987 and November 2021. Results In the current review, we present a comprehensive list of CAR-T cells developed to treat GBM, we describe new possible T-cell engineering strategies against GBM while presenting a short introductory history to the reader regarding the origin(s) of this cutting-edge therapy. We have also compiled a unique list of anti-GBM CAR-Ts with their specific protein sequences and their functions as well as an inventory of clinical trials involving CAR-T and GBM. Conclusions The aim of this review is to introduce the reader to the field of T-cell engineering using CAR-Ts to treat GBM and describe the obstacles that may need to be addressed in order to significantly delay the relentless growth of GBM. Supplementary Information The online version contains supplementary material available at 10.1007/s11060-021-03902-8.
Collapse
|
4
|
Chen HX, Song M, Maecker HT, Gnjatic S, Patton D, Lee JJ, Adam SJ, Moravec R, Liu XS, Cerami E, Lindsay J, Tang M, Hodi FS, Wu CJ, Wistuba II, Al-Atrash G, Bernatchez C, Bendall SC, Hewitt SM, Sharon E, Streicher H, Enos RA, Bowman MD, Tatard-Leitman VM, Sanchez-Espiridion B, Ranasinghe S, Pichavant M, Del Valle DM, Yu J, Janssens S, Peterson-Klaus J, Rowe C, Bongers G, Jenq RR, Chang CC, Abrams JS, Mooney M, Doroshow JH, Harris LN, Thurin M. Network for Biomarker Immunoprofiling for Cancer Immunotherapy: Cancer Immune Monitoring and Analysis Centers and Cancer Immunologic Data Commons (CIMAC-CIDC). Clin Cancer Res 2021; 27:5038-5048. [PMID: 33419780 PMCID: PMC8491462 DOI: 10.1158/1078-0432.ccr-20-3241] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/09/2020] [Accepted: 12/23/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Immunoprofiling to identify biomarkers and integration with clinical trial outcomes are critical to improving immunotherapy approaches for patients with cancer. However, the translational potential of individual studies is often limited by small sample size of trials and the complexity of immuno-oncology biomarkers. Variability in assay performance further limits comparison and interpretation of data across studies and laboratories. EXPERIMENTAL DESIGN To enable a systematic approach to biomarker identification and correlation with clinical outcome across trials, the Cancer Immune Monitoring and Analysis Centers and Cancer Immunologic Data Commons (CIMAC-CIDC) Network was established through support of the Cancer MoonshotSM Initiative of the National Cancer Institute (NCI) and the Partnership for Accelerating Cancer Therapies (PACT) with industry partners via the Foundation for the NIH. RESULTS The CIMAC-CIDC Network is composed of four academic centers with multidisciplinary expertise in cancer immunotherapy that perform validated and harmonized assays for immunoprofiling and conduct correlative analyses. A data coordinating center (CIDC) provides the computational expertise and informatics platforms for the storage, integration, and analysis of biomarker and clinical data. CONCLUSIONS This overview highlights strategies for assay harmonization to enable cross-trial and cross-site data analysis and describes key elements for establishing a network to enhance immuno-oncology biomarker development. These include an operational infrastructure, validation and harmonization of core immunoprofiling assays, platforms for data ingestion and integration, and access to specimens from clinical trials. Published in the same volume are reports of harmonization for core analyses: whole-exome sequencing, RNA sequencing, cytometry by time of flight, and IHC/immunofluorescence.
Collapse
Affiliation(s)
- Helen X Chen
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), Bethesda, Maryland.
| | - Minkyung Song
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), Bethesda, Maryland
| | - Holden T Maecker
- The Human Immune Monitoring Center (HIMC), Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California
| | - Sacha Gnjatic
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - David Patton
- Center for Biomedical Informatics and Information Technology, NCI, Bethesda, Maryland
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stacey J Adam
- Foundation for the National Institutes of Health, North Bethesda, Maryland
| | - Radim Moravec
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
- Kelly Services, Rockville, Maryland
| | - Xiaole Shirley Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ethan Cerami
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - James Lindsay
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ming Tang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - F Stephen Hodi
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gheath Al-Atrash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chantale Bernatchez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sean C Bendall
- Department of Pathology, School of Medicine, Stanford University, Stanford, California
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Elad Sharon
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), Bethesda, Maryland
| | - Howard Streicher
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), Bethesda, Maryland
| | | | | | | | - Beatriz Sanchez-Espiridion
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Srinika Ranasinghe
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mina Pichavant
- The Human Immune Monitoring Center (HIMC), Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California
| | - Diane M Del Valle
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joyce Yu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Cathy Rowe
- Center for Biomedical Informatics and Information Technology, NCI, Bethesda, Maryland
- Essex Management, Rockville, Maryland
| | - Gerold Bongers
- Microbiome Translational Center, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Robert R Jenq
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chia-Chi Chang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey S Abrams
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), Bethesda, Maryland
| | - Margaret Mooney
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), Bethesda, Maryland
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Lyndsay N Harris
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Magdalena Thurin
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland.
| |
Collapse
|
5
|
Verdegaal E, van der Kooij MK, Visser M, van der Minne C, de Bruin L, Meij P, Terwisscha van Scheltinga A, Welters MJ, Santegoets S, de Miranda N, Roozen I, Liefers GJ, Kapiteijn E, van der Burg SH. Low-dose interferon-alpha preconditioning and adoptive cell therapy in patients with metastatic melanoma refractory to standard (immune) therapies: a phase I/II study. J Immunother Cancer 2021; 8:jitc-2019-000166. [PMID: 32238469 PMCID: PMC7174065 DOI: 10.1136/jitc-2019-000166] [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] [Accepted: 02/17/2020] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Adoptive cell therapy (ACT) with tumor-reactive T cells has shown consistent clinical efficacy. We evaluated the response to ACT in combination with interferon alpha (IFNa) preconditioning in patients with stage IV metastatic melanoma, most of which were progressive on cytotoxic T-lymphocyte-associated protein 4 and/or programmed cell death protein 1 checkpoint blockade therapy. METHODS Thirty-four patients were treated with ex vivo expanded tumor reactive T cells, derived from mixed lymphocyte autologous tumor cultures, or with autologous tumor-infiltrating lymphocytes and evaluated for clinical response. Clinical and immunological parameters associated with response were also evaluated. RESULTS Best overall response defined as clinical benefit, comprising either complete response, partial response or stable disease >6 months, was observed in 29% of the patients. Forty-three per cent of the 14 immunotherapy-naïve patients and 20% of the 20 patients progressive on prior immunotherapy benefited from ACT. The overall survival (OS) was 90% versus 28.6% at 1 year and 46.7% versus 0% at 3 years follow-up, of responder and non-responder patients, respectively. Median OS was 36 versus 7 months, respectively. IFNa pretreatment resulted in leukopenia, neutropenia and lymphopenia, which was sustained during the treatment in clinical responders and associated with response. Differences in antigen specificity, but not in phenotype, cytokine profile or CD8+ T cell number of the ACT products correlated with clinical response. Cross-reactivity of the ACT products to one or more allogeneic human leukocyte antigen-matched melanoma cell lines was associated with short OS after treatment while the ACT products of very long-term survivors showed no cross-reactivity but recognized patient-specific neoantigens. CONCLUSION This study demonstrates that ACT in combination with a mild IFNa preconditioning regimen can induce clinical benefit even in immunotherapy pretreated patients, although with lower success than in immunotherapy-naïve patients. ACT products comprising neoantigen reactivity may be more effective.
Collapse
Affiliation(s)
- Els Verdegaal
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique K van der Kooij
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Marten Visser
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Caroline van der Minne
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Linda de Bruin
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Pauline Meij
- GMP Facility Leiden, Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anton Terwisscha van Scheltinga
- GMP Facility Leiden, Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marij J Welters
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Saskia Santegoets
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Noel de Miranda
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Inge Roozen
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerrit Jan Liefers
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
6
|
Bader P, Klingebiel T. [CAR T-cell therapy for children and adolescents with acute lymphatic leukemia]. Internist (Berl) 2021; 62:597-604. [PMID: 33950273 DOI: 10.1007/s00108-021-01041-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Acute lymphatic leukemia is the most common malignant disease in children. Despite a good prognosis, new therapeutic concepts are needed for patients with refractory and relapsed disease. QUESTION The importance of chimeric antigen receptor (CAR) T‑cell therapy for patients with refractory and post-stem cell transplantation. MATERIAL AND METHODS Overview of studies in childhood; presentation of extraction, use, side effects and prognostic factors. RESULTS In 30 patients with a leukemia burden of <5% at the time of lymphocytic chemotherapy, the probability of event-free survival after 12 months was 0.70; in patients with a leukemia burden of >5%, the probability of event-free survival was only 0.23. CONCLUSIONS Further improvements in vector design will help to improve the effectiveness of this treatment. It will become important to isolate factors that will make it possible to identify patients who will respond to this therapy in the long term.
Collapse
Affiliation(s)
- Peter Bader
- Klinik für Kinder- und Jugendmedizin, Schwerpunkt Stammzelltransplantation und Immunologie, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Deutschland.
| | - Thomas Klingebiel
- Klinik für Kinder- und Jugendmedizin, Schwerpunkt Stammzelltransplantation und Immunologie, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Deutschland
| |
Collapse
|
7
|
García de Guadiana-Romualdo L, Morell-García D, Morales-Indiano C, Bauça JM, Alcaide Martín MJ, Esparza del Valle C, Gutiérrez Revilla JI, Urrechaga E, Álamo JM, Hernando Holgado AM, Lorenzo-Lozano MC, Sánchez Fdez-Pacheco S, de la Hera Cagigal P, Juncos Tobarra MA, Vílchez JA, Vírseda Chamorro I, Gutiérrez Garcia I, Pastor Murcia Y, Sahuquillo Frías L, Altimira Queral L, Nuez-Zaragoza E, Adell Ruiz de León J, Ruiz Ripa A, Salas Gómez-Pablos P, Cebreiros López I, Fernández Uriarte A, Larruzea Á, López Yepes ML, Esteban Torrella P, Zamorano Andrés MC, Pedregosa Díaz J, Acevedo Alcaraz C, Blazquez-Manzanera AL, Padilla Jiménez AML, Baamonde Calzada MC, Vera M, Cháfer Rudilla M, Canalda Campás M, García Muñoz S, Vicente Gutiérrez L, Jiménez Añón L, Pérez Martínez A, Pons Castillo A, González Tamayo R, Férriz Vivancos J, Rodríguez-Fraga O, Ferrer Díaz de Brito Fernández V, Aguadero V, García Arévalo MG, Arnaldos Carrillo M, González Morales M, Núñez Gárate M, Ruiz Iruela C, Sancho-Rodríguez N, Vila Pérez M, Egea-Caparrós JM, Sáenz L, Blasco Barbero Á, Galán Ortega A. Characteristics and laboratory findings on admission to the emergency department among 2873 hospitalized patients with COVID-19: the impact of adjusted laboratory tests in multicenter studies. A multicenter study in Spain (BIOCOVID-Spain study). Scand J Clin Lab Invest 2021; 81:187-193. [PMID: 33591234 PMCID: PMC7898295 DOI: 10.1080/00365513.2021.1881997] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/31/2020] [Accepted: 01/24/2021] [Indexed: 02/08/2023]
Abstract
Identification of predictors for severe disease progression is key for risk stratification in COVID-19 patients. We aimed to describe the main characteristics and identify the early predictors for severe outcomes among hospitalized patients with COVID-19 in Spain. This was an observational, retrospective cohort study (BIOCOVID-Spain study) including COVID-19 patients admitted to 32 Spanish hospitals. Demographics, comorbidities and laboratory tests were collected. Outcome was in-hospital mortality. For analysis, laboratory tests values were previously adjusted to assure the comparability of results among participants. Cox regression was performed to identify predictors. Study population included 2873 hospitalized COVID-19 patients. Nine variables were independent predictors for in-hospital mortality, including creatinine (Hazard ratio [HR]:1.327; 95% Confidence Interval [CI]: 1.040-1.695, p = .023), troponin (HR: 2.150; 95% CI: 1.155-4.001; p = .016), platelet count (HR: 0.994; 95% CI: 0.989-0.998; p = .004) and C-reactive protein (HR: 1.037; 95% CI: 1.006-1.068; p = .019). This is the first multicenter study in which an effort was carried out to adjust the results of laboratory tests measured with different methodologies to guarantee their comparability. We reported a comprehensive information about characteristics in a large cohort of hospitalized COVID-19 patients, focusing on the analytical features. Our findings may help to identify patients early at a higher risk for an adverse outcome.
Collapse
Affiliation(s)
| | - Daniel Morell-García
- Laboratory Medicine Department, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | | | - Josep Miquel Bauça
- Laboratory Medicine Department, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | | | - Clara Esparza del Valle
- Laboratory Medicine Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | | | | | - José M. Álamo
- Biochemical Laboratory, Hospital Marina Baixa, Villajoyosa, Spain
| | | | | | | | | | - María A. Juncos Tobarra
- Laboratory Medicine Department, Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | - Juan A. Vílchez
- Laboratory Medicine Department. Hospital, Universitario Morales Meseguer, Murcia, Spain
| | | | | | - Yolanda Pastor Murcia
- Laboratory Medicine Department, Consorci Hospital General Universitari de València, Valencia, Spain
| | | | - Laura Altimira Queral
- Laboratory Medicine Department, Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, Spain
| | - Elisa Nuez-Zaragoza
- Clinical Laboratory Department, Parc Tauli Hospital Universitari, Sabadell, Spain
| | | | - Alicia Ruiz Ripa
- Laboratory Medicine Department, Hospital de Mataró, Mataró, Spain
| | | | - Iria Cebreiros López
- Laboratory Medicine Department, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | | | - Álex Larruzea
- Laboratory Medicine Deparment, Hospital Fundació Sanitària Mollet, Barcelona, Spain
| | | | | | | | | | - Cristina Acevedo Alcaraz
- Laboratory Medicine Department. Hospital, Universitario Los Arcos del Mar Menor, San Javier, Spain
| | | | | | | | - Marina Vera
- Biochemical Laboratory, Hospital Marina Baixa, Villajoyosa, Spain
| | | | | | - Sara García Muñoz
- Laboratory Medicine Department, Hospital Universitario de Basurto, Bilbao, Spain
| | - Luis Vicente Gutiérrez
- Laboratory Medicine Department, Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | - Laura Jiménez Añón
- Laboratory Medicine Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | | | - Ruth González Tamayo
- Laboratory Medicine Department, Hospital Universitario de Torrevieja, Torrevieja, Spain
| | - Jorge Férriz Vivancos
- Laboratory Medicine Department, Consorci Hospital General Universitari de València, Valencia, Spain
| | | | | | - Vicente Aguadero
- Clinical Laboratory Department, Parc Tauli Hospital Universitari, Sabadell, Spain
| | | | | | | | | | - Cristina Ruiz Iruela
- Laboratory Medicine Deparment, Hospital Fundació Sanitària Mollet, Barcelona, Spain
| | | | - Marti Vila Pérez
- Laboratory Medicine Department, Hospital Verge de la Cinta, Tortosa, Spain
| | - José M. Egea-Caparrós
- Laboratory Medicine Department. Hospital, Universitario Los Arcos del Mar Menor, San Javier, Spain
| | - Luis Sáenz
- Laboratory Medicine Department. Hospital General, Universitario Rafael Méndez, Lorca, Spain
| | | | - Amparo Galán Ortega
- Comisión de Magnitudes Biológicas relacionadas con la Urgencia Médica, Sociedad Española de Medicina de Laboratorio (SEQC-ML), Barcelona, Spain
| |
Collapse
|
8
|
Hoos A, Janetzki S, Britten CM. Advancing the field of cancer immunotherapy: MIATA consensus guidelines become available to improve data reporting and interpretation for T-cell immune monitoring. Oncoimmunology 2021; 1:1457-1459. [PMID: 23264891 PMCID: PMC3525600 DOI: 10.4161/onci.22308] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Axel Hoos
- Glaxo Smith Kline, Collegeville, PA USA ; Cancer Immunotherapy Consortium; Cancer Research Institute; New York, NY USA
| | | | | |
Collapse
|
9
|
Hu-Lieskovan S, Bhaumik S, Dhodapkar K, Grivel JCJB, Gupta S, Hanks BA, Janetzki S, Kleen TO, Koguchi Y, Lund AW, Maccalli C, Mahnke YD, Novosiadly RD, Selvan SR, Sims T, Zhao Y, Maecker HT. SITC cancer immunotherapy resource document: a compass in the land of biomarker discovery. J Immunother Cancer 2020; 8:e000705. [PMID: 33268350 PMCID: PMC7713206 DOI: 10.1136/jitc-2020-000705] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2020] [Indexed: 02/07/2023] Open
Abstract
Since the publication of the Society for Immunotherapy of Cancer's (SITC) original cancer immunotherapy biomarkers resource document, there have been remarkable breakthroughs in cancer immunotherapy, in particular the development and approval of immune checkpoint inhibitors, engineered cellular therapies, and tumor vaccines to unleash antitumor immune activity. The most notable feature of these breakthroughs is the achievement of durable clinical responses in some patients, enabling long-term survival. These durable responses have been noted in tumor types that were not previously considered immunotherapy-sensitive, suggesting that all patients with cancer may have the potential to benefit from immunotherapy. However, a persistent challenge in the field is the fact that only a minority of patients respond to immunotherapy, especially those therapies that rely on endogenous immune activation such as checkpoint inhibitors and vaccination due to the complex and heterogeneous immune escape mechanisms which can develop in each patient. Therefore, the development of robust biomarkers for each immunotherapy strategy, enabling rational patient selection and the design of precise combination therapies, is key for the continued success and improvement of immunotherapy. In this document, we summarize and update established biomarkers, guidelines, and regulatory considerations for clinical immune biomarker development, discuss well-known and novel technologies for biomarker discovery and validation, and provide tools and resources that can be used by the biomarker research community to facilitate the continued development of immuno-oncology and aid in the goal of durable responses in all patients.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Kavita Dhodapkar
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | | | - Sumati Gupta
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Brent A Hanks
- Duke University Medical Center, Durham, North Carolina, USA
| | | | | | - Yoshinobu Koguchi
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Amanda W Lund
- Oregon Health and Science University, Portland, Oregon, USA
| | | | | | | | | | - Tasha Sims
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | | |
Collapse
|
10
|
Shemesh CS, Hsu JC, Hosseini I, Shen BQ, Rotte A, Twomey P, Girish S, Wu B. Personalized Cancer Vaccines: Clinical Landscape, Challenges, and Opportunities. Mol Ther 2020; 29:555-570. [PMID: 33038322 DOI: 10.1016/j.ymthe.2020.09.038] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/15/2020] [Accepted: 09/26/2020] [Indexed: 12/21/2022] Open
Abstract
Tremendous innovation is underway among a rapidly expanding repertoire of promising personalized immune-based treatments. Therapeutic cancer vaccines (TCVs) are attractive systemic immunotherapies that activate and expand antigen-specific CD8+ and CD4+ T cells to enhance anti-tumor immunity. Our review highlights key issues impacting TCVs in clinical practice and reports on progress in development. We review the mechanism of action, immune-monitoring, dosing strategies, combinations, obstacles, and regulation of cancer vaccines. Most trials of personalized TCVs are ongoing and represent diverse platforms with predominantly early investigations of mRNA, DNA, or peptide-based targeting strategies against neoantigens in solid tumors, with many in combination immunotherapies. Multiple delivery systems, routes of administration, and dosing strategies are used. Intravenous or intramuscular administration is common, including delivery by lipid nanoparticles. Absorption and biodistribution impact antigen uptake, expression, and presentation, affecting the strength, speed, and duration of immune response. The emerging trials illustrate the complexity of developing this class of innovative immunotherapies. Methodical testing of the multiple potential factors influencing immune responses, as well as refined quantitative methodologies to facilitate optimal dosing strategies, could help resolve uncertainty of therapeutic approaches. To increase the likelihood of success in bringing these medicines to patients, several unique development challenges must be overcome.
Collapse
Affiliation(s)
- Colby S Shemesh
- Department of Clinical Pharmacology Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Joy C Hsu
- Department of Clinical Pharmacology Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Iraj Hosseini
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ben-Quan Shen
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Anand Rotte
- Department of Clinical Pharmacology Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Patrick Twomey
- Department of Product Development Safety, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Sandhya Girish
- Department of Clinical Pharmacology Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Benjamin Wu
- Department of Clinical Pharmacology Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
11
|
Weis P, Helm J, Page L, Lauruschkat CD, Lazariotou M, Einsele H, Loeffler J, Ullmann AJ, Wurster S. Development and evaluation of a whole blood-based approach for flow cytometric quantification of CD154+ mould-reactive T cells. Med Mycol 2020; 58:187-196. [PMID: 31095327 DOI: 10.1093/mmy/myz038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/30/2019] [Accepted: 04/02/2019] [Indexed: 12/17/2022] Open
Abstract
CD154+ mould-reactive T cells were proposed as a novel biomarker in the diagnosis of invasive mycoses. As PBMC-based protocols for flow cytometric quantification of these cells are logistically challenging and susceptible to preanalytic delays, this study evaluated and optimized a whole blood-based method for the detection of mould-reactive T cells. Blood collection tubes containing costimulatory antibodies and Aspergillus fumigatus mycelial lysates were inoculated with heparinized whole blood from healthy adults, and detection rates of CD154+/CD4+A. fumigatus reactive T cells were compared with PBMC-based detection using samples from the same donors. In contrast to the PBMC-based method, double costimulation with αCD28 and αCD49d was crucial for reliable whole blood stimulation. Optimizing stimulation schemes for both matrixes, significantly higher specific T-cell detection rates were achieved by the whole blood-based method, whereas the unspecific background stimulation remained low. MHC II-dependent CD154+ upregulation was demonstrated for both matrixes. Excellent correlation and reproducible conversion factors between whole blood and PBMC-based results were observed. Using frozen ready-to-use test tubes containing costimulatory antibodies and lysates, detection rates of specific T cells were comparable to freshly prepared blood collection tubes. The optimized whole blood-based protocol was also used to detect Rhizopus arrhizus and Rhizomucor pusillus reactive T cells, resulting in 1.5- to 2.7-fold higher detection rates compared with PBMC-based measurement. In summary, the whole blood protocol is a robust, highly sensitive, and cost-effective method for mould-reactive T-cell quantification, allowing for point-of-care sample stimulation and contributing to better assay standardization in multi-centre evaluation of mould reactive T-cell quantification.
Collapse
Affiliation(s)
- Philipp Weis
- University Hospital of Wuerzburg, Department of Internal Medicine II, Division of Infectious Diseases, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Johanna Helm
- University Hospital of Wuerzburg, Department of Internal Medicine II, Division of Infectious Diseases, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Lukas Page
- University Hospital of Wuerzburg, Department of Internal Medicine II, Division of Infectious Diseases, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Chris D Lauruschkat
- University Hospital of Wuerzburg, Department of Internal Medicine II, Division of Infectious Diseases, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Maria Lazariotou
- University Hospital of Wuerzburg, Department of Internal Medicine II, Division of Infectious Diseases, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Hermann Einsele
- University Hospital of Wuerzburg, Department of Internal Medicine II, Division of Infectious Diseases, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Juergen Loeffler
- University Hospital of Wuerzburg, Department of Internal Medicine II, Division of Infectious Diseases, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Andrew J Ullmann
- University Hospital of Wuerzburg, Department of Internal Medicine II, Division of Infectious Diseases, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Sebastian Wurster
- University Hospital of Wuerzburg, Department of Internal Medicine II, Division of Infectious Diseases, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany.,The University of Texas MD Anderson Cancer Center, Department of Infectious Diseases, 1515 Holcombe Boulevard, Houston, TX 77030, United States of America
| |
Collapse
|
12
|
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
|
13
|
Wakabayashi G, Lee YC, Luh F, Kuo CN, Chang WC, Yen Y. Development and clinical applications of cancer immunotherapy against PD-1 signaling pathway. J Biomed Sci 2019; 26:96. [PMID: 31801525 PMCID: PMC6894306 DOI: 10.1186/s12929-019-0588-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/13/2019] [Indexed: 12/26/2022] Open
Abstract
Dramatic advances in immune therapy have emerged as a promising strategy in cancer therapeutics. In addition to chemotherapy and radiotherapy, inhibitors targeting immune-checkpoint molecules such as cytotoxic T-lymphocyte antigen-4 (CTLA-4), programmed cell death receptor-1 (PD-1) and its ligand (PD-L1) demonstrate impressive clinical benefits in clinical trials. In this review, we present background information about therapies involving PD-1/PD-L1 blockade and provide an overview of current clinical trials. Furthermore, we present recent advances involving predictive biomarkers associated with positive therapeutic outcomes in cancer immunotherapy.
Collapse
Affiliation(s)
| | - Yu-Ching Lee
- Center for Cancer Transnational Research, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan, 110
| | - Frank Luh
- Sino-American Cancer Foundation, 668 Arrow Grand Circle, Suite 101, Covina, California, 91722, USA
| | - Chun-Nan Kuo
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University; Department of Pharmacy, Integrative Therapy Center for Gastroenterologic Cancers, Wan Fang Hospital; Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan, 110
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University; Department of Pharmacy, Integrative Therapy Center for Gastroenterologic Cancers, Wan Fang Hospital; Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan, 110.
| | - Yun Yen
- PhD Program for Cancer Biology and Drug Discovery, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan, 110.
| |
Collapse
|
14
|
Marliot F, Lafontaine L, Galon J. Immunoscore assay for the immune classification of solid tumors: Technical aspects, improvements and clinical perspectives. Methods Enzymol 2019; 636:109-128. [PMID: 32178816 DOI: 10.1016/bs.mie.2019.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The past two decades witnessed the appreciation of the importance of specific tumor-infiltrating immune cells in influencing tumor evolution. The discovery that a favorable immune contexture is linked to a prolonged patients' survival, and more specifically that intratumoral cytotoxic T lymphocytes hold powerful prognostic value, provided the foundations for the development of the Immunoscore. Immunoscore is a digital pathology, IHC-based immune assay measuring the densities of CD3+ and CD8+ T cells at different tumor locations, linking them with patients' clinical outcome. The present chapter provides a detailed overview of the assay development and procedure, from the bench to the data analysis, and of the internationally approved and validated guidelines to perform a consensus Immunoscore for colon cancer patients. Assay strengths and limitations are also discussed, as well as the clinical implications of its introduction in routine practice.
Collapse
Affiliation(s)
- Florence Marliot
- INSERM, Laboratory of Integrative Cancer Immunology, Équipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; Centre de Recherche des Cordeliers, Paris, France; Laboratory of Immunology, AP-HP, European Hospital Georges Pompidou, Paris, France
| | - Lucie Lafontaine
- INSERM, Laboratory of Integrative Cancer Immunology, Équipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; Centre de Recherche des Cordeliers, Paris, France
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Équipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; Centre de Recherche des Cordeliers, Paris, France.
| |
Collapse
|
15
|
Arora S, Velichinskii R, Lesh RW, Ali U, Kubiak M, Bansal P, Borghaei H, Edelman MJ, Boumber Y. Existing and Emerging Biomarkers for Immune Checkpoint Immunotherapy in Solid Tumors. Adv Ther 2019; 36:2638-2678. [PMID: 31410780 PMCID: PMC6778545 DOI: 10.1007/s12325-019-01051-z] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Indexed: 02/06/2023]
Abstract
In the last few years, immunotherapy has transformed the way we treat solid tumors, including melanoma, lung, head neck, breast, renal, and bladder cancers. Durable responses and long-term survival benefit has been experienced by many cancer patients, with favorable toxicity profiles of immunotherapeutic agents relative to chemotherapy. Cures have become possible in some patients with metastatic disease. Additional approvals of immunotherapy drugs and in combination with other agents are anticipated in the near future. Multiple additional immunotherapy drugs are in earlier stages of clinical development, and their testing in additional tumor types is under way. Despite considerable early success and relatively fewer side effects, the majority of cancer patients do not respond to checkpoint inhibitors. Additionally, while the drugs are generally well tolerated, there is still the potential for significant, unpredictable and even fatal toxicity with these agents. Improved biomarkers may help to better select patients who are more likely to respond to these drugs. Two key biologically important predictive tissue biomarkers, specifically, PD-L1 and mismatch repair deficiency, have been FDA-approved in conjunction with the checkpoint inhibitor, pembrolizumab. Tumor mutation burden, another promising biomarker, is emerging in several tumor types, and may also soon receive approval. Finally, several other tissue and liquid biomarkers are emerging that could help guide single-agent immunotherapy and in combination with other agents. Of these, one promising investigational biomarker is alteration or deficiency in DNA damage response (DDR) pathways, with altered DDR observed in a broad spectrum of tumors. Here, we provide a critical overview of current, emerging, and investigational biomarkers in the context of response to immunotherapy in solid tumors.
Collapse
Affiliation(s)
- Sanjeevani Arora
- Program in Cancer Prevention and Control, Fox Chase Cancer Center, Philadelphia, PA, USA.
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Rodion Velichinskii
- Program in Cancer Prevention and Control, Fox Chase Cancer Center, Philadelphia, PA, USA
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Randy W Lesh
- Program in Cancer Prevention and Control, Fox Chase Cancer Center, Philadelphia, PA, USA
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
- Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | - Usman Ali
- Division of Hospital Medicine, Department of Medicine, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Michal Kubiak
- Internal Medicine Residency Program, Centegra Health System, McHenry Hospital and Rosalind Franklin University, Mchenry, IL, USA
| | | | - Hossein Borghaei
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Martin J Edelman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yanis Boumber
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA.
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
| |
Collapse
|
16
|
Angell HK, Bruni D, Barrett JC, Herbst R, Galon J. The Immunoscore: Colon Cancer and Beyond. Clin Cancer Res 2019; 26:332-339. [DOI: 10.1158/1078-0432.ccr-18-1851] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/11/2019] [Accepted: 08/08/2019] [Indexed: 01/28/2023]
|
17
|
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
|
18
|
Gouttefangeas C, Rammensee HG. Personalized cancer vaccines: adjuvants are important, too. Cancer Immunol Immunother 2018; 67:1911-1918. [PMID: 29644387 PMCID: PMC11028305 DOI: 10.1007/s00262-018-2158-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/28/2018] [Indexed: 12/30/2022]
Abstract
Therapeutic cancer vaccines have shown limited clinical efficacy so far. Nevertheless, in the meantime, our understanding of immune cell function and the interactions of immune cells with growing tumors has advanced considerably. We are now in a position to invest this knowledge into the design of more powerful vaccines and therapy combinations aimed at increasing immunogenicity and decreasing tumor-induced immunosuppression. This review focuses essentially on peptide-based human vaccines. We will discuss two aspects that are critical for increasing their intrinsic immunogenicity: the selection of the antigen(s) to be targeted, and the as yet unmet need for strong adjuvants.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
| | - Hans-Georg Rammensee
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| |
Collapse
|
19
|
Implementing liquid biopsies into clinical decision making for cancer immunotherapy. Oncotarget 2018; 8:48507-48520. [PMID: 28501851 PMCID: PMC5564665 DOI: 10.18632/oncotarget.17397] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/04/2017] [Indexed: 02/06/2023] Open
Abstract
During the last decade, novel immunotherapeutic strategies, in particular antibodies directed against immune checkpoint inhibitors, have revolutionized the treatment of different malignancies leading to an improved survival of patients. Identification of immune-related biomarkers for diagnosis, prognosis, monitoring of immune responses and selection of patients for specific cancer immunotherapies is urgently required and therefore areas of intensive research. Easily accessible samples in particular liquid biopsies (body fluids), such as blood, saliva or urine, are preferred for serial tumor biopsies. Although monitoring of immune and tumor responses prior, during and post immunotherapy has led to significant advances of patients’ outcome, valid and stable prognostic biomarkers are still missing. This might be due to the limited capacity of the technologies employed, reproducibility of results as well as assay stability and validation of results. Therefore solid approaches to assess immune regulation and modulation as well as to follow up the nature of the tumor in liquid biopsies are urgently required to discover valuable and relevant biomarkers including sample preparation, timing of the collection and the type of liquid samples. This article summarizes our knowledge of the well-known liquid material in a new context as liquid biopsy and focuses on collection and assay requirements for the analysis and the technical developments that allow the implementation of different high-throughput assays to detect alterations at the genetic and immunologic level, which could be used for monitoring treatment efficiency, acquired therapy resistance mechanisms and the prognostic value of the liquid biopsies.
Collapse
|
20
|
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
|
21
|
Ward BJ, Pillet S, Charland N, Trepanier S, Couillard J, Landry N. The establishment of surrogates and correlates of protection: Useful tools for the licensure of effective influenza vaccines? Hum Vaccin Immunother 2018; 14:647-656. [PMID: 29252098 PMCID: PMC5861778 DOI: 10.1080/21645515.2017.1413518] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The search for a test that can predict vaccine efficacy is an important part of any vaccine development program. Although regulators hesitate to acknowledge any test as a true ‘correlate of protection’, there are many precedents for defining ‘surrogate’ assays. Surrogates can be powerful tools for vaccine optimization, licensure, comparisons between products and development of improved products. When such tests achieve ‘reference’ status however, they can inadvertently become barriers to new technologies that do not work the same way as existing vaccines. This is particularly true when these tests are based upon circularly-defined ‘reference’ or, even worse, proprietary reagents. The situation with inactivated influenza vaccines is a good example of this phenomenon. The most frequently used tests to define vaccine-induced immunity are all serologic assays: hemagglutination inhibition (HI), single radial hemolysis (SRH) and microneutralization (MN). The first two, and particularly the HI assay, have achieved reference status and criteria have been established in many jurisdictions for their use in licensing new vaccines and to compare the performance of different vaccines. However, all of these assays are based on biological reagents that are notoriously difficult to standardize and can vary substantially by geography, by chance (i.e. developing reagents in eggs that may not antigenitically match wild-type viruses) and by intention (ie: choosing reagents that yield the most favorable results). This review describes attempts to standardize these assays to improve their performance as surrogates, the dangers of over-reliance on ‘reference’ serologic assays, the ways that manufacturers can exploit the existing regulatory framework to make their products ‘look good’ and the implications of this long-established system for the introduction of novel influenza vaccines.
Collapse
Affiliation(s)
- Brian J Ward
- a Research Institute of the McGill University Health Centre, Infectious Diseases Division , Montreal , QC , Canada.,b Medicago Inc , Québec , QC , Canada
| | - Stephane Pillet
- a Research Institute of the McGill University Health Centre, Infectious Diseases Division , Montreal , QC , Canada.,b Medicago Inc , Québec , QC , Canada
| | | | | | | | | |
Collapse
|
22
|
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
|
23
|
Gulley JL, Berzofsky JA, Butler MO, Cesano A, Fox BA, Gnjatic S, Janetzki S, Kalavar S, Karanikas V, Khleif SN, Kirsch I, Lee PP, Maccalli C, Maecker H, Schlom J, Seliger B, Siebert J, Stroncek DF, Thurin M, Yuan J, Butterfield LH. Immunotherapy biomarkers 2016: overcoming the barriers. J Immunother Cancer 2017; 5:29. [PMID: 28653584 PMCID: PMC5359902 DOI: 10.1186/s40425-017-0225-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/13/2017] [Indexed: 12/18/2022] Open
Abstract
This report summarizes the symposium, 'Immunotherapy Biomarkers 2016: Overcoming the Barriers', which was held on April 1, 2016 at the National Institutes of Health in Bethesda, Maryland. The symposium, cosponsored by the Society for Immunotherapy of Cancer (SITC) and the National Cancer Institute (NCI), focused on emerging immunotherapy biomarkers, new technologies, current hurdles to further progress, and recommendations for advancing the field of biomarker development.
Collapse
Affiliation(s)
- James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, 10 Center Dr., 13 N240, Bethesda, MD, 20892, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, 41 Medlars Dr, Bldg 41 Rm D702D, Bethesda, MD, 20892, USA
| | - Marcus O Butler
- Princess Margaret Cancer Center/Ontario Cancer Institute, RM 9-622, 610 University Ave, Toronto, ON, Canada
| | - Alessandra Cesano
- NanoString, Inc., 500 Fairview Avenue North, Seattle, WA, 98109, USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR, 97213, USA
| | - Sacha Gnjatic
- Department of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, S5-105, 1470 Madison Avenue, Box 1128, New York, NY, 10029, USA
| | - Sylvia Janetzki
- ZellNet Consulting, Inc., 555 North Avenue, Fort Lee, NJ, 07024, USA
| | - Shyam Kalavar
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, 1401 Rockville Pike, Rockville, MD, 20852, USA
| | - Vaios Karanikas
- Roche Innovation Center Zurich, Wagistrasse 18, Schlieren, Switzerland
| | - Samir N Khleif
- Georgia Cancer Center, Augusta University, 1120 15th Street, CN-2101A, Augusta, GA, 30912, USA
| | - Ilan Kirsch
- Adaptive Biotechnologies, Inc., 1551 Eastlake Ave. E., Seattle, WA, 98102, USA
| | - Peter P Lee
- Department of Immuno-oncology, City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Cristina Maccalli
- Department of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Holden Maecker
- Stanford University Medical Center, 299 Campus Drive, Stanford, CA, 94303, USA
| | - Jeffrey Schlom
- National Cancer Institute, National Institutes of Health, 10 Center Drive, Bldg. 10, Room 8B09, Bethesda, MD, 20892, USA
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, Halle, Germany
| | - Janet Siebert
- CytoAnalytics, 3500 South Albion Street, Cherry Hills Village, CO, 80113, USA
| | - David F Stroncek
- Department of Transfusion Medicine, National Institutes of Health, 10 Center Drive, Building 10, Room 3C720, Bethesda, MD, 20892, USA
| | - Magdalena Thurin
- National Cancer Institute, Cancer Diagnosis Program, DCTD, National Institutes of Health, 9609 Medical Center Drive, Bethesda, 20892, MD, USA
| | - Jianda Yuan
- Early Clinical Oncology Development, Merck Research Laboratories, Rahway, NJ, 07065, USA
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
24
|
Mellman I, Hubbard-Lucey VM, Tontonoz MJ, Kalos MD, Chen DS, Allison JP, Drake CG, Levitsky H, Lonberg N, van der Burg SH, Fearon DT, Wherry EJ, Lowy I, Vonderheide RH, Hwu P. De-Risking Immunotherapy: Report of a Consensus Workshop of the Cancer Immunotherapy Consortium of the Cancer Research Institute. Cancer Immunol Res 2016; 4:279-88. [PMID: 27036972 DOI: 10.1158/2326-6066.cir-16-0045] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
With the recent FDA approvals of pembrolizumab and nivolumab, and a host of additional immunomodulatory agents entering clinical development each year, the field of cancer immunotherapy is changing rapidly. Strategies that can assist researchers in choosing the most promising drugs and drug combinations to move forward through clinical development are badly needed in order to reduce the likelihood of late-stage clinical trial failures. On October 5, 2014, the Cancer Immunotherapy Consortium of the Cancer Research Institute, a collaborative think tank composed of stakeholders from academia, industry, regulatory agencies, and patient interest groups, met to discuss strategies for de-risking immunotherapy development, with a focus on integrating preclinical and clinical studies, and conducting smarter early-phase trials, particularly for combination therapies. Several recommendations were made, including making better use of clinical data to inform preclinical research, obtaining adequate tissues for biomarker studies, and choosing appropriate clinical trial endpoints to identify promising drug candidates and combinations in nonrandomized early-phase trials.
Collapse
Affiliation(s)
| | | | | | | | | | - James P Allison
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Hy Levitsky
- Roche Innovation Center, Zurich, Switzerland
| | | | | | | | - E John Wherry
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Israel Lowy
- Regeneron Pharmaceuticals, Tarrytown, New York
| | - Robert H Vonderheide
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Patrick Hwu
- The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
25
|
Song L, Lachno DR, Hanlon D, Shepro A, Jeromin A, Gemani D, Talbot JA, Racke MM, Dage JL, Dean RA. A digital enzyme-linked immunosorbent assay for ultrasensitive measurement of amyloid-β 1-42 peptide in human plasma with utility for studies of Alzheimer's disease therapeutics. ALZHEIMERS RESEARCH & THERAPY 2016; 8:58. [PMID: 27978855 PMCID: PMC5160015 DOI: 10.1186/s13195-016-0225-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 11/18/2016] [Indexed: 12/18/2022]
Abstract
Background Amyloid-β 1–42 peptide (Aβ1–42) is associated with plaque formation in the brain of patients with Alzheimer’s disease (AD). Pharmacodynamic studies of AD therapeutics that lower the concentrations of Aβ1–42 in peripheral blood require highly sensitive assays for its measurement. A digital enzyme-linked immunosorbent assay (ELISA) using single molecule array (Simoa) technology has been developed that provides improved sensitivity compared with conventional ELISA methods using the same antibody reagents. Methods A sensitive digital ELISA for measurement of Aβ1–42 using antibodies 3D6 and 21F12 was developed. Assay performance was evaluated by repeated testing of pooled human plasma and buffer diluent quality control samples to determine relative accuracy, intra- and inter-assay precision, limit of detection (LOD), lower limit of quantification (LLOQ), dilutional linearity, and spike recovery. The optimized assay was used to quantify Aβ1–42 in clinical samples from patients treated with the β-site amyloid precursor protein cleaving enzyme 1 inhibitor LY2886721. Results The prototype assay measured Aβ1–42 with an LOD of 0.3 pg/ml and an LLOQ of 2.8 pg/ml in plasma, calibrated using an Aβ1–42 peptide standard from Fujirebio. Assay precision was acceptable with intra- and inter-assay coefficients of variation both being ≤10%. Dilutional linearity was demonstrated in sample diluent and immunodepleted human plasma. Analyte spike recovery ranged from 51% to 93% with a mean of 80%. This assay was able to quantify Aβ1–42 in all of the 84 clinical samples tested. A rapid reduction in levels of Aβ1–42 was detected within 1 h after drug treatment, and a dose-dependent decrease of Aβ1–42 levels was also observed over the time course of sample collection. Conclusions This digital ELISA has potential utility in clinical applications for quantification of Aβ1–42 in plasma where high sensitivity and precision are required.
Collapse
Affiliation(s)
- Linan Song
- Quanterix Corporation, Lexington, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Analytical Aspects of the Implementation of Biomarkers in Clinical Transplantation. Ther Drug Monit 2016; 38 Suppl 1:S80-92. [PMID: 26418704 DOI: 10.1097/ftd.0000000000000230] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In response to the urgent need for new reliable biomarkers to complement the guidance of the immunosuppressive therapy, a huge number of biomarker candidates to be implemented in clinical practice have been introduced to the transplant community. This includes a diverse range of molecules with very different molecular weights, chemical and physical properties, ex vivo stabilities, in vivo kinetic behaviors, and levels of similarity to other molecules, etc. In addition, a large body of different analytical techniques and assay protocols can be used to measure biomarkers. Sometimes, a complex software-based data evaluation is a prerequisite for appropriate interpretation of the results and for their reporting. Although some analytical procedures are of great value for research purposes, they may be too complex for implementation in a clinical setting. Whereas the proof of "fitness for purpose" is appropriate for validation of biomarker assays used in exploratory drug development studies, a higher level of analytical validation must be achieved and eventually advanced analytical performance might be necessary before diagnostic application in transplantation medicine. A high level of consistency of results between laboratories and between methods (if applicable) should be obtained and maintained to make biomarkers effective instruments in support of therapeutic decisions. This overview focuses on preanalytical and analytical aspects to be considered for the implementation of new biomarkers for adjusting immunosuppression in a clinical setting and highlights critical points to be addressed on the way to make them suitable as diagnostic tools. These include but are not limited to appropriate method validation, standardization, education, automation, and commercialization.
Collapse
|
27
|
Analytical Validation and Cross-Validation of an NFAT-Regulated Gene Expression Assay for Pharmacodynamic Monitoring of Therapy With Calcineurin Inhibitors. Ther Drug Monit 2016; 38:711-716. [DOI: 10.1097/ftd.0000000000000340] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Affiliation(s)
- Cedrik M Britten
- GlaxoSmithKline, Immuno-Oncology and Combinations, Oncology Research and Development, Stevenage, UK
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, ZA Leiden, The Netherlands
| | - Cécile Gouttefangeas
- Interfaculty Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| |
Collapse
|
29
|
Körber N, Behrends U, Hapfelmeier A, Protzer U, Bauer T. Validation of an IFNγ/IL2 FluoroSpot assay for clinical trial monitoring. J Transl Med 2016; 14:175. [PMID: 27297580 PMCID: PMC4906590 DOI: 10.1186/s12967-016-0932-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/31/2016] [Indexed: 11/29/2022] Open
Abstract
Background The FluoroSpot assay, an advancement of the ELISpot assay, enables simultaneous measurement of different analytes secreted at a single-cell level. This allows parallel detection of several cytokines secreted by immune cells upon antigen recognition. Easier standardization, higher sensitivity and reduced labour intensity render FluoroSpot assays an interesting alternative to flow-cytometry based assays for analysis of clinical samples. While the use of immunoassays to study immunological primary and secondary endpoints becomes increasingly attractive, assays used require pre-trial validation. Here we describe the assay validation (precision, specificity and linearity) of a FluoroSpot immunological endpoint assay detecting Interferon γ (IFNγ) and Interleukin 2 (IL2) for use in clinical trial immune monitoring. Methods We validated an IFNγ/IL2 FluoroSpot assay to determine Epstein-Barr virus (EBV)-specific cellular immune responses (IFNγ, IL2 and double positive IFNγ + IL2 responses), using overlapping peptide pools corresponding to EBV-proteins BZLF1 and EBNA3A. Assay validation was performed using cryopreserved PBMC of 16 EBV-seropositive and 6 EBV-seronegative donors. Precision was assessed by (i) testing 16 donors using three replicates per assay (intra-assay precision/repeatability) (ii) using two plates in parallel (intermediate precision/plate-to-plate variability) and (iii) by performing the assays on three different days (inter-assay precision/reproducibility). In addition, we determined specificity, linearity and quantification limits of the assay. Further we tested precision across the two assay systems, IFNγ/IL2 FluoroSpot and the corresponding enzymatic single cytokine ELISpot. Results The validation revealed: (1) a high intra-assay precision (coefficient of variation (CV) 9.96, 8.85 and 13.05 %), intermediate precision (CV 6.48, 10.20 and 12.97 %) and reproducibility (CV 20.81 %, 12,75 % and 12.07 %) depending on the analyte and antigen used; (2) a specificity of 100 %; (3) a linearity with R2 values from 0.93 to 0.99 depending on the analyte. The testing of the precision across the two assay systems, adduced a concordance correlation coefficient pc = 0.99 for IFNγ responses and pc = 0.93 for IL2 responses, indicating a large agreement between both assay methods. Conclusions The validated primary endpoint assay, an EBV peptide pool specific IFNγ/IL2 FluoroSpot assay was found to be suitable for the detection of EBV-specific immune responses subject to the requirement of standardized assay procedure and data analysis. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0932-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nina Körber
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Schneckenburgerstr. 8, 81675, Munich, Germany
| | - Uta Behrends
- Clinical Cooperation Group Pediatric Tumor Immunology, Children's Hospital, Technische Universität München/Helmholtz Zentrum München, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Alexander Hapfelmeier
- Institute of Medical Statistics and Epidemiology, Technische Universität München, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Schneckenburgerstr. 8, 81675, Munich, Germany.,Clinical Cooperation Group, Immune Monitoring, Helmholtz Zentrum München/Technische Universität München, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Tanja Bauer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Schneckenburgerstr. 8, 81675, Munich, Germany. .,Clinical Cooperation Group, Immune Monitoring, Helmholtz Zentrum München/Technische Universität München, Munich, Germany. .,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
30
|
Kohrt HE, Tumeh PC, Benson D, Bhardwaj N, Brody J, Formenti S, Fox BA, Galon J, June CH, Kalos M, Kirsch I, Kleen T, Kroemer G, Lanier L, Levy R, Lyerly HK, Maecker H, Marabelle A, Melenhorst J, Miller J, Melero I, Odunsi K, Palucka K, Peoples G, Ribas A, Robins H, Robinson W, Serafini T, Sondel P, Vivier E, Weber J, Wolchok J, Zitvogel L, Disis ML, Cheever MA. Immunodynamics: a cancer immunotherapy trials network review of immune monitoring in immuno-oncology clinical trials. J Immunother Cancer 2016; 4:15. [PMID: 26981245 PMCID: PMC4791805 DOI: 10.1186/s40425-016-0118-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/15/2016] [Indexed: 12/26/2022] Open
Abstract
The efficacy of PD-1/PD-L1 targeted therapies in addition to anti-CTLA-4 solidifies immunotherapy as a modality to add to the anticancer arsenal. Despite raising the bar of clinical efficacy, immunologically targeted agents raise new challenges to conventional drug development paradigms by highlighting the limited relevance of assessing standard pharmacokinetics (PK) and pharmacodynamics (PD). Specifically, systemic and intratumoral immune effects have not consistently correlated with standard relationships between systemic dose, toxicity, and efficacy for cytotoxic therapies. Hence, PK and PD paradigms remain inadequate to guide the selection of doses and schedules, both starting and recommended Phase 2 for immunotherapies. The promise of harnessing the immune response against cancer must also be considered in light of unique and potentially serious toxicities. Refining immune endpoints to better inform clinical trial design represents a high priority challenge. The Cancer Immunotherapy Trials Network investigators review the immunodynamic effects of specific classes of immunotherapeutic agents to focus immune assessment modalities and sites, both systemic and importantly intratumoral, which are critical to the success of the rapidly growing field of immuno-oncology.
Collapse
Affiliation(s)
- Holbrook E Kohrt
- Division of Oncology, Stanford Cancer Institute, Stanford University Medical Center, 269 Campus Drive, CCSR 1105, Stanford, CA 94305-5151 USA
| | - Paul C Tumeh
- Division of Dermatology, Department of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - Don Benson
- Division of Hematology/Oncology, Ohio State University, Columbus, OH USA
| | - Nina Bhardwaj
- Medicine, Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY USA
| | - Joshua Brody
- Medicine, Hematology and Medical Oncology, Mount Sinai Hospital, Ruttenberg Treatment Center, New York, NY USA
| | - Silvia Formenti
- Department of Radiation Oncology, New York Weill Cornell Medical Center, New York, NY USA
| | - Bernard A Fox
- SOM-Molecular Microbiology & Immunology Department, Laboratory of Molecular and Tumor Immunology, OHSU Cancer Institute, Portland, OR USA
| | - Jerome Galon
- INSERM, Integrative Cancer Immunology Team, Cordeliers Research Center, Paris, France
| | - Carl H June
- Perelman School of Medicine, University of Pennsylvania, Pathology and Laboratory Medicine, Philadelphia, PA USA
| | - Michael Kalos
- Cancer Immunobiology, Eli Lilly & Company, New York, NY USA
| | - Ilan Kirsch
- Translational Medicine, Adaptive Biotechnologies Corp, Seattle, WA USA
| | - Thomas Kleen
- Immune Monitoring, Epiontis GmbH, Berlin, Germany
| | - Guido Kroemer
- Faculty of Medicine, University of Paris Descartes, Paris, France
| | - Lewis Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, CA USA
| | - Ron Levy
- Division of Oncology, Stanford School of Medicine, Stanford, CA USA
| | - H Kim Lyerly
- Duke University School of Medicine, Durham, NC USA
| | - Holden Maecker
- Human Immune Monitoring Center Shared Resource, Stanford Cancer Institute, Stanford, CA USA
| | | | - Jos Melenhorst
- Product Development and Correlative Sciences, Smilow Center for Translational Research, Philadelphia, PA USA
| | - Jeffrey Miller
- Division of Hematology, Experimental Therapeutics, University of Minnesota, Oncology and Transplantation, Minneapolis, MN USA
| | - Ignacio Melero
- Centro de Investigacion Medica Aplicada, Universidad de Navarra, Avda. Pamplona, Spain
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY USA
| | | | - George Peoples
- Cancer Vaccine Development Program, Brooke Army Medical Center, Houston, TX USA
| | - Antoni Ribas
- Tumor Immunology Program Area, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA USA
| | | | - William Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | | | - Paul Sondel
- Cellular & Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI USA
| | - Eric Vivier
- Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | | | - Jedd Wolchok
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY USA
| | - Laurence Zitvogel
- Institut National de la Santé et Recherche Médicale, Institut GrustaveRoussy, Villejuif, France
| | - Mary L Disis
- Tumor Vaccine Group, University of Washington, Seattle, WA USA
| | - Martin A Cheever
- Fred Hutchinson Cancer Research Center, 1100 Eastlake Ave N., E3-300, PO Box 19024, Seattle, WA 98109-1023 USA
| | | |
Collapse
|
31
|
Yang J, Diaz N, Adelsberger J, Zhou X, Stevens R, Rupert A, Metcalf JA, Baseler M, Barbon C, Imamichi T, Lempicki R, Cosentino LM. The effects of storage temperature on PBMC gene expression. BMC Immunol 2016; 17:6. [PMID: 26979060 PMCID: PMC4791795 DOI: 10.1186/s12865-016-0144-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 03/04/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Cryopreservation of peripheral blood mononuclear cells (PBMCs) is a common and essential practice in conducting research. There are different reports in the literature as to whether cryopreserved PBMCs need to only be stored ≤ -150 °C or can be stored for a specified time at -80 °C. Therefore, we performed gene expression analysis on cryopreserved PBMCs stored at both temperatures for 14 months and PBMCs that underwent temperature cycling 104 times between these 2 storage temperatures. Real-time RT-PCR was performed to confirm the involvement of specific genes associated with identified cellular pathways. All cryopreserved/stored samples were compared to freshly isolated PBMCs and between storage conditions. RESULTS We identified a total of 1,367 genes whose expression after 14 months of storage was affected >3 fold in PBMCs following isolation, cryopreservation and thawing as compared to freshly isolated PBMC aliquots that did not undergo cryopreservation. Sixty-six of these genes were shared among two or more major stress-related cellular pathways (stress responses, immune activation and cell death). Thirteen genes involved in these pathways were tested by real-time RT-PCR and the results agreed with the corresponding microarray data. There was no significant change on the gene expression if the PBMCs experienced brief but repetitive temperature cycling as compared to those that were constantly kept ≤ -150 °C. However, there were 18 genes identified to be different when PBMCs were stored at -80 °C but did not change when stored < -150 °C. A correlation was also found between the expressions of 2'-5'- oligoadenylate synthetase (OAS2), a known interferon stimulated gene (IFSG), and poor PBMC recovery post-thaw. PBMC recovery and viability were better when the cells were stored ≤ -150 °C as compared to -80 °C. CONCLUSIONS Not only is the viability and recovery of PBMCs affected during cryopreservation but also their gene expression pattern, as compared to freshly isolated PBMCs. Different storage temperature of PBMCs can activate or suppress different genes, but the cycling between -80 °C and -150 °C did not produce significant alterations in gene expression when compared to PBMCs stored ≤ -150 °C. Further analysis by gene expression of various PBMC processing and cryopreservation procedures is currently underway, as is identifying possible molecular mechanisms.
Collapse
Affiliation(s)
- Jun Yang
- />Leidos Biomedical Research, Inc., Frederick, MD 21702 USA
| | - Norma Diaz
- />Leidos Biomedical Research, Inc., Frederick, MD 21702 USA
| | | | - Xueyuan Zhou
- />Leidos Biomedical Research, Inc., Frederick, MD 21702 USA
| | - Randy Stevens
- />Leidos Biomedical Research, Inc., Frederick, MD 21702 USA
| | - Adam Rupert
- />Leidos Biomedical Research, Inc., Frederick, MD 21702 USA
| | - Julia A. Metcalf
- />Division of Clinical Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD 20852 USA
| | - Mike Baseler
- />Leidos Biomedical Research, Inc., Frederick, MD 21702 USA
| | | | | | | | | |
Collapse
|
32
|
Hoos A. Development of immuno-oncology drugs - from CTLA4 to PD1 to the next generations. Nat Rev Drug Discov 2016; 15:235-47. [PMID: 26965203 DOI: 10.1038/nrd.2015.35] [Citation(s) in RCA: 460] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Since the regulatory approval of ipilimumab in 2011, the field of cancer immunotherapy has been experiencing a renaissance. This success is based on progress in both preclinical and clinical science, including the development of new methods of investigation. Immuno-oncology has become a sub-specialty within oncology owing to its unique science and its potential for substantial and long-term clinical benefit. Immunotherapy agents do not directly attack the tumour but instead mobilize the immune system - this can be achieved through various approaches that utilize adaptive or innate immunity. Therefore, immuno-oncology drug development encompasses a broad range of agents, including antibodies, peptides, proteins, small molecules, adjuvants, cytokines, oncolytic viruses, bi-specific molecules and cellular therapies. This Perspective summarizes the recent history of cancer immunotherapy, including the factors that led to its success, provides an overview of novel drug-development considerations, summarizes three generations of immunotherapies that have been developed since 2011 and, thus, illustrates the breadth of opportunities these new generations of immunotherapies represent.
Collapse
Affiliation(s)
- Axel Hoos
- Oncology Research and Development, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, USA
| |
Collapse
|
33
|
Rekers NV, von Herrath MG, Wesley JD. Immunotherapies and immune biomarkers in Type 1 diabetes: A partnership for success. Clin Immunol 2015; 161:37-43. [PMID: 26122172 DOI: 10.1016/j.clim.2015.05.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 12/16/2022]
Abstract
The standard of care (SoC) for Type 1 diabetes (T1D) today is much the same as it was in the early 1920s, simply with more insulin options-fast-acting, slow-acting, injectable, and inhalable insulins. However, these well-tolerated treatments only manage the symptoms and complications, but do nothing to halt the underlying immune response. There is an unmet need for better treatment options for T1D that address all aspects of the disease. For decades, we have successfully treated T1D in preclinical animal models with immune-modifying therapies that have not demonstrated comparable efficacy in humans. The path to bringing such options to the clinic will depend on the implementation and standard inclusion of biomarkers of immune and therapeutic efficacy in T1D clinical trials, and dictate if we can create a new SoC that treats the underlying autoimmunity as well as the symptoms it causes.
Collapse
Affiliation(s)
- Niels V Rekers
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA; Pacific Northwest Diabetes Research Institute, Seattle, WA, USA
| | | | - Johnna D Wesley
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA.
| |
Collapse
|
34
|
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
|
35
|
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
|
36
|
Klaver Y, Kunert A, Sleijfer S, Debets R, Lamers CHJ. Adoptive T-cell therapy: a need for standard immune monitoring. Immunotherapy 2015; 7:513-33. [DOI: 10.2217/imt.15.23] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cancer immune therapy, in particular the use of checkpoint inhibitors and adoptive transfer of T cells has recently demonstrated significant clinical responses against several tumor types. Unfortunately, these therapies are frequently accompanied by severe toxicities, underscoring the need for markers that provide information on therapy response. Monitoring immune responses in the tumor microenvironment and peripheral blood prior to and during these therapies will provide better insight into the mechanisms underlying clinical activities, and will potentially enable the identification of such markers. In this review, we present an overview of adoptive T-cell trials conducted with a special focus on immune monitoring, and argue that accurate monitoring of T cells is pivotal to further development of immune therapies to treat cancer.
Collapse
Affiliation(s)
- Yarne Klaver
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Andre Kunert
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Cor HJ Lamers
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
37
|
Bidmon N, Attig S, Rae R, Schröder H, Omokoko TA, Simon P, Kuhn AN, Kreiter S, Sahin U, Gouttefangeas C, van der Burg SH, Britten CM. Generation of TCR-Engineered T Cells and Their Use To Control the Performance of T Cell Assays. THE JOURNAL OF IMMUNOLOGY 2015; 194:6177-89. [DOI: 10.4049/jimmunol.1400958] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 04/09/2015] [Indexed: 11/19/2022]
|
38
|
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
|
39
|
Kaiser AD, Assenmacher M, Schröder B, Meyer M, Orentas R, Bethke U, Dropulic B. Towards a commercial process for the manufacture of genetically modified T cells for therapy. Cancer Gene Ther 2015; 22:72-8. [PMID: 25613483 PMCID: PMC4356749 DOI: 10.1038/cgt.2014.78] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 11/05/2014] [Indexed: 12/12/2022]
Abstract
The recent successes of adoptive T-cell immunotherapy for the treatment of hematologic malignancies have highlighted the need for manufacturing processes that are robust and scalable for product commercialization. Here we review some of the more outstanding issues surrounding commercial scale manufacturing of personalized-adoptive T-cell medicinal products. These include closed system operations, improving process robustness and simplifying work flows, reducing labor intensity by implementing process automation, scalability and cost, as well as appropriate testing and tracking of products, all while maintaining strict adherence to Current Good Manufacturing Practices and regulatory guidelines. A decentralized manufacturing model is proposed, where in the future patients' cells could be processed at the point-of-care in the hospital.
Collapse
Affiliation(s)
- A D Kaiser
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | | | - B Schröder
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - M Meyer
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - R Orentas
- Lentigen Technology Inc., Gaithersburg, MD, USA
| | - U Bethke
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - B Dropulic
- Lentigen Technology Inc., Gaithersburg, MD, USA
| |
Collapse
|
40
|
Hadrup SR, Maurer D, Laske K, Frøsig TM, Andersen SR, Britten CM, van der Burg SH, Walter S, Gouttefangeas C. Cryopreservation of MHC multimers: Recommendations for quality assurance in detection of antigen specific T cells. Cytometry A 2015; 87:37-48. [PMID: 25297339 PMCID: PMC4309491 DOI: 10.1002/cyto.a.22575] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 08/24/2014] [Accepted: 09/24/2014] [Indexed: 11/16/2022]
Abstract
Fluorescence-labeled peptide-MHC class I multimers serve as ideal tools for the detection of antigen-specific T cells by flow cytometry, enabling functional and phenotypical characterization of specific T cells at the single cell level. While this technique offers a number of unique advantages, MHC multimer reagents can be difficult to handle in terms of stability and quality assurance. The stability of a given fluorescence-labeled MHC multimer complex depends on both the stability of the peptide-MHC complex itself and the stability of the fluorochrome. Consequently, stability is difficult to predict and long-term storage is generally not recommended. We investigated here the possibility of cryopreserving MHC multimers, both in-house produced and commercially available, using a wide range of peptide-MHC class I multimers comprising virus and cancer-associated epitopes of different affinities presented by various HLA-class I molecules. Cryopreservation of MHC multimers was feasible for at least 6 months, when they were dissolved in buffer containing 5-16% glycerol (v/v) and 0.5% serum albumin (w/v). The addition of cryoprotectants was tolerated across three different T-cell staining protocols for all fluorescence labels tested (PE, APC, PE-Cy7 and Quantum dots). We propose cryopreservation as an easily implementable method for stable storage of MHC multimers and recommend the use of cryopreservation in long-term immunomonitoring projects, thereby eliminating the variability introduced by different batches and inconsistent stability.
Collapse
Affiliation(s)
- Sine Reker Hadrup
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), University Hospital HerlevHerlev, Denmark
| | | | - Karoline Laske
- Department of Immunology, Institute for Cell Biology, University of TübingenGermany and DKTK, DKFZ partner site Tübingen, Germany
| | - Thomas Mørch Frøsig
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), University Hospital HerlevHerlev, Denmark
| | - Sofie Ramskov Andersen
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), University Hospital HerlevHerlev, Denmark
| | - Cedrik M Britten
- Translational Oncology, University Medical Center, Johannes Gutenberg-University Mainz gGmbHMainz, Germany
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical CenterLeiden, The Netherlands
| | | | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of TübingenGermany and DKTK, DKFZ partner site Tübingen, Germany
| |
Collapse
|
41
|
Improvement of IFNg ELISPOT Performance Following Overnight Resting of Frozen PBMC Samples Confirmed Through Rigorous Statistical Analysis. Cells 2014; 4:1-18. [PMID: 25546016 PMCID: PMC4381205 DOI: 10.3390/cells4010001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/16/2014] [Indexed: 01/18/2023] Open
Abstract
Immune monitoring of functional responses is a fundamental parameter to establish correlates of protection in clinical trials evaluating vaccines and therapies to boost antigen-specific responses. The IFNg ELISPOT assay is a well-standardized and validated method for the determination of functional IFNg-producing T-cells in peripheral blood mononuclear cells (PBMC); however, its performance greatly depends on the quality and integrity of the cryopreserved PBMC. Here, we investigate the effect of overnight (ON) resting of the PBMC on the detection of CD8-restricted peptide-specific responses by IFNg ELISPOT. The study used PBMC from healthy donors to evaluate the CD8 T-cell response to five pooled or individual HLA-A2 viral peptides. The results were analyzed using a modification of the existing distribution free resampling (DFR) recommended for the analysis of ELISPOT data to ensure the most rigorous possible standard of significance. The results of the study demonstrate that ON resting of PBMC samples prior to IFNg ELISPOT increases both the magnitude and the statistical significance of the responses. In addition, a comparison of the results with a 13-day preculture of PBMC with the peptides before testing demonstrates that ON resting is sufficient for the efficient evaluation of immune functioning.
Collapse
|
42
|
Sebastian M, Papachristofilou A, Weiss C, Früh M, Cathomas R, Hilbe W, Wehler T, Rippin G, Koch SD, Scheel B, Fotin-Mleczek M, Heidenreich R, Kallen KJ, Gnad-Vogt U, Zippelius A. Phase Ib study evaluating a self-adjuvanted mRNA cancer vaccine (RNActive®) combined with local radiation as consolidation and maintenance treatment for patients with stage IV non-small cell lung cancer. BMC Cancer 2014; 14:748. [PMID: 25288198 PMCID: PMC4195907 DOI: 10.1186/1471-2407-14-748] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 09/25/2014] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Advanced non-small cell lung cancer (NSCLC) represents a significant unmet medical need. Despite advances with targeted therapies in a small subset of patients, fewer than 20% of patients survive for more than two years after diagnosis. Cancer vaccines are a promising therapeutic approach that offers the potential for durable responses through the engagement of the patient's own immune system. CV9202 is a self-adjuvanting mRNA vaccine that targets six antigens commonly expressed in NSCLC (NY-ESO-1, MAGEC1, MAGEC2, 5 T4, survivin, and MUC1). METHODS/DESIGN The trial will assess the safety and tolerability of CV9202 vaccination combined with local radiation designed to enhance immune responses and will include patients with stage IV NSCLC and a response or stable disease after first-line chemotherapy or therapy with an EGFR tyrosine kinase inhibitor. Three histological and molecular subtypes of NSCLC will be investigated (squamous and non-squamous cell with/without EGFR mutations). All patients will receive two initial vaccinations with CV9202 prior to local radiotherapy (5 GY per day for four successive days) followed by further vaccinations until disease progression. The primary endpoint of the study is the number of patients experiencing Grade >3 treatment-related adverse events. Pharmacodynamic analyses include the assessment of immune responses to the antigens encoded by CV9202 and others not included in the panel (antigen spreading) and standard efficacy assessments. DISCUSSION RNActive self-adjuvanted mRNA vaccines offer the potential for simultaneously inducing immune responses to a wide panel of antigens commonly expressed in tumors. This trial will assess the feasibility of this approach in combination with local radiotherapy in NSCLC patients. TRIAL REGISTRATION Clinicaltrials.gov: NCT01915524/EudraCT No.: 2012-004230-41.
Collapse
Affiliation(s)
- Martin Sebastian
- />Department of Hematology and Oncology, Johann-Wolfgang-Goethe-Universität, Frankfurt, Germany
| | | | - Christian Weiss
- />Department of Radiation Therapy and Oncology, Goethe University, Frankfurt am Main, Germany
| | - Martin Früh
- />Department of Medical Oncology and Hematology, Kantonsspital St Gallen, St Gallen, Switzerland
| | | | - Wolfgang Hilbe
- />Department of General Internal Medicine, Oncology, University Hospital, Innsbruck, Austria
| | - Thomas Wehler
- />Third Department of Internal Medicine, University Hospital Mainz, Mainz, Germany
| | | | | | | | | | | | | | | | - Alfred Zippelius
- />Department of Oncology, University Hospital Basel, Petersgraben 4, CH - 4031 Basel, Switzerland
| |
Collapse
|
43
|
Dutruel C, Thole J, Geels M, Mollenkopf HJ, Ottenhoff T, Guzman CA, Fletcher HA, Leroy O, Kaufmann SH. TRANSVAC workshop on standardisation and harmonisation of analytical platforms for HIV, TB and malaria vaccines: ‘How can big data help?’. Vaccine 2014; 32:4365-4368. [DOI: 10.1016/j.vaccine.2014.06.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 05/26/2014] [Accepted: 06/06/2014] [Indexed: 01/08/2023]
|
44
|
Janetzki S, Britten CM. The role of the reporting framework MIATA within current efforts to advance immune monitoring. J Immunol Methods 2014; 409:6-8. [PMID: 24816466 DOI: 10.1016/j.jim.2014.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/01/2014] [Accepted: 05/02/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Sylvia Janetzki
- ZellNet Consulting, Inc., 555 North Avenue, Suite 25-S, Fort Lee, NJ 07024, USA.
| | - Cedrik M Britten
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz gGmbH, Verfügungsgebäude 708, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
45
|
Sanchez AM, Denny TN, O'Gorman M. Introduction to a Special Issue of the Journal of Immunological Methods: Building global resource programs to support HIV/AIDS clinical trial studies. J Immunol Methods 2014; 409:1-5. [PMID: 24910413 DOI: 10.1016/j.jim.2014.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 05/30/2014] [Indexed: 11/30/2022]
Abstract
This Special Issue of the Journal of Immunological Methods includes 16 manuscripts describing quality assurance activities related to virologic and immunologic monitoring of six global laboratory resource programs that support international HIV/AIDS clinical trial studies: Collaboration for AIDS Vaccine Discovery (CAVD); Center for HIV/AIDS Vaccine Immunology (CHAVI); External Quality Assurance Program Oversight Laboratory (EQAPOL); HIV Vaccine Trial Network (HVTN); International AIDS Vaccine Initiative (IAVI); and Immunology Quality Assessment (IQA). The reports from these programs address the many components required to develop comprehensive quality control activities and subsequent quality assurance programs for immune monitoring in global clinical trials including: all aspects of processing, storing, and quality assessment of PBMC preparations used ubiquitously in HIV clinical trials, the development and optimization of assays for CD8 HIV responses and HIV neutralization, a comprehensive global HIV virus repository, and reports on the development and execution of novel external proficiency testing programs for immunophenotyping, intracellular cytokine staining, ELISPOT and luminex based cytokine measurements. In addition, there are articles describing the implementation of Good Clinical Laboratory Practices (GCLP) in a large quality assurance laboratory, the development of statistical methods specific for external proficiency testing assessment, a discussion on the ability to set objective thresholds for measuring rare events by flow cytometry, and finally, a manuscript which addresses a framework for the structured reporting of T cell immune function based assays. It is anticipated that this series of manuscripts covering a wide range of quality assurance activities associated with the conduct of global clinical trials will provide a resource for individuals and programs involved in improving the harmonization, standardization, accuracy, and sensitivity of virologic and immunologic testing.
Collapse
Affiliation(s)
- Ana M Sanchez
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA.
| | - Thomas N Denny
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Maurice O'Gorman
- Departments of Pathology and Pediatrics, Keck School of Medicine, University of Southern California, USA; Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
46
|
Ontiveros N, Tye-Din JA, Hardy MY, Anderson RP. Ex-vivo whole blood secretion of interferon (IFN)-γ and IFN-γ-inducible protein-10 measured by enzyme-linked immunosorbent assay are as sensitive as IFN-γ enzyme-linked immunospot for the detection of gluten-reactive T cells in human leucocyte antigen (HLA)-DQ2·5(+) -associated coeliac disease. Clin Exp Immunol 2014; 175:305-15. [PMID: 24192268 DOI: 10.1111/cei.12232] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2013] [Indexed: 01/22/2023] Open
Abstract
T cell cytokine release assays are used to diagnose infectious diseases, but not autoimmune or allergic disease. Coeliac disease (CD) is a common T cell-mediated disease diagnosed by the presence of gluten-dependent intestinal inflammation and serology. Many patients cannot be diagnosed with CD because they reduce dietary gluten before medical workup. Oral gluten challenge in CD patients treated with gluten-free diet (GFD) mobilizes gluten-reactive T cells measurable by interferon (IFN)-γ enzyme-linked immunospot (ELISPOT) or major histocompatibility complex (MHC) class II tetramers. Immunodominant peptides are quite consistent in the 90% of patients who possess HLA-DQ2·5. We aimed to develop whole blood assays to detect gluten-specific T cells. Blood was collected before and after gluten challenge from GFD donors confirmed to have CD (n = 27, all HLA-DQ2·5(+) ), GFD donors confirmed not to have CD (n = 6 HLA-DQ2·5(+) , 11 HLA-DQ2·5(-) ) and donors with CD not following GFD (n = 4, all HLA-DQ2·5(+) ). Plasma IFN-γ and IFN-γ inducible protein-10 (IP-10) were measured by enzyme-linked immunosorbent assay (ELISA) after whole blood incubation with peptides or gliadin, and correlated with IFN-γ ELISPOT. No T cell assay could distinguish between CD patients and controls prior to gluten challenge, but after gluten challenge the whole blood IFN-γ ELISA and the ELISPOT were both 85% sensitive and 100% specific for HLA-DQ2·5(+) CD patients; the whole blood IP-10 ELISA was 94% sensitive and 100% specific. We conclude that whole blood cytokine release assays are sensitive and specific for detection of gluten-reactive T cells in CD; further clinical studies addressing the utility of these tests in patients with an uncertain diagnosis of CD is warranted.
Collapse
Affiliation(s)
- N Ontiveros
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia; Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | | | | | | |
Collapse
|
47
|
Development and implementation of a proficiency testing program for Luminex bead-based cytokine assays. J Immunol Methods 2014; 409:62-71. [PMID: 24801479 DOI: 10.1016/j.jim.2014.04.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 04/10/2014] [Accepted: 04/23/2014] [Indexed: 11/21/2022]
Abstract
Luminex bead array assays are widely used for rapid biomarker quantification due to the ability to measure up to 100 unique analytes in a single well of a 96-well plate. There has been, however, no comprehensive analysis of variables impacting assay performance, nor development of a standardized proficiency testing program for laboratories performing these assays. To meet this need, the NIH/NIAID and the Cancer Immunotherapy Consortium of the Cancer Research Institute collaborated to develop and implement a Luminex assay proficiency testing program as part of the NIH/NIAID-sponsored External Quality Assurance Program Oversight Laboratory (EQAPOL) at Duke University. The program currently monitors 25 domestic and international sites with two external proficiency panels per year. Each panel includes a de-identified commercial Luminex assay kit with standards to quantify human IFNγ, TNFα, IL-6, IL-10 and IL-2, and a series of recombinant cytokine-spiked human serum samples. All aspects of panel development, testing and shipping are performed under GCLP by EQAPOL support teams. Following development testing, a comprehensive site proficiency scoring system comprised of timeliness, protocol adherence, accuracy and precision was implemented. The overall mean proficiency score across three rounds of testing has remained stable (EP3: 76%, EP4: 75%, EP5: 77%); however, a more detailed analysis of site reported results indicates a significant improvement of intra- (within) and inter- (between) site variation, suggesting that training and remediation for poor performing sites may be having a positive impact on proficiency. Through continued proficiency testing, identification of variables affecting Luminex assay outcomes will strengthen efforts to bring standardization to the field.
Collapse
|
48
|
Galon J, Mlecnik B, Bindea G, Angell HK, Berger A, Lagorce C, Lugli A, Zlobec I, Hartmann A, Bifulco C, Nagtegaal ID, Palmqvist R, Masucci GV, Botti G, Tatangelo F, Delrio P, Maio M, Laghi L, Grizzi F, Asslaber M, D'Arrigo C, Vidal-Vanaclocha F, Zavadova E, Chouchane L, Ohashi PS, Hafezi-Bakhtiari S, Wouters BG, Roehrl M, Nguyen L, Kawakami Y, Hazama S, Okuno K, Ogino S, Gibbs P, Waring P, Sato N, Torigoe T, Itoh K, Patel PS, Shukla SN, Wang Y, Kopetz S, Sinicrope FA, Scripcariu V, Ascierto PA, Marincola FM, Fox BA, Pagès F. Towards the introduction of the 'Immunoscore' in the classification of malignant tumours. J Pathol 2014; 232:199-209. [PMID: 24122236 PMCID: PMC4255306 DOI: 10.1002/path.4287] [Citation(s) in RCA: 1061] [Impact Index Per Article: 96.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 02/06/2023]
Abstract
The American Joint Committee on Cancer/Union Internationale Contre le Cancer (AJCC/UICC) TNM staging system provides the most reliable guidelines for the routine prognostication and treatment of colorectal carcinoma. This traditional tumour staging summarizes data on tumour burden (T), the presence of cancer cells in draining and regional lymph nodes (N) and evidence for distant metastases (M). However, it is now recognized that the clinical outcome can vary significantly among patients within the same stage. The current classification provides limited prognostic information and does not predict response to therapy. Multiple ways to classify cancer and to distinguish different subtypes of colorectal cancer have been proposed, including morphology, cell origin, molecular pathways, mutation status and gene expression-based stratification. These parameters rely on tumour-cell characteristics. Extensive literature has investigated the host immune response against cancer and demonstrated the prognostic impact of the in situ immune cell infiltrate in tumours. A methodology named 'Immunoscore' has been defined to quantify the in situ immune infiltrate. In colorectal cancer, the Immunoscore may add to the significance of the current AJCC/UICC TNM classification, since it has been demonstrated to be a prognostic factor superior to the AJCC/UICC TNM classification. An international consortium has been initiated to validate and promote the Immunoscore in routine clinical settings. The results of this international consortium may result in the implementation of the Immunoscore as a new component for the classification of cancer, designated TNM-I (TNM-Immune).
Collapse
Affiliation(s)
- Jérôme Galon
- INSERM, U872, Laboratory of Integrative Cancer ImmunologyParis, France
- Université Paris DescartesParis, France
- Centre de Recherche des CordeliersUniversité Pierre et Marie Curie Paris 6, France
| | - Bernhard Mlecnik
- INSERM, U872, Laboratory of Integrative Cancer ImmunologyParis, France
- Université Paris DescartesParis, France
- Centre de Recherche des CordeliersUniversité Pierre et Marie Curie Paris 6, France
| | - Gabriela Bindea
- INSERM, U872, Laboratory of Integrative Cancer ImmunologyParis, France
- Université Paris DescartesParis, France
- Centre de Recherche des CordeliersUniversité Pierre et Marie Curie Paris 6, France
| | - Helen K Angell
- INSERM, U872, Laboratory of Integrative Cancer ImmunologyParis, France
- Université Paris DescartesParis, France
- Centre de Recherche des CordeliersUniversité Pierre et Marie Curie Paris 6, France
| | - Anne Berger
- Digestive Surgery Department, Georges Pompidou European HospitalParis, France
| | | | | | - Inti Zlobec
- Institute of Pathology, University of BernSwitzerland
| | | | - Carlo Bifulco
- Department of Pathology, Providence Portland Medical CenterPortland, OR, USA
| | - Iris D Nagtegaal
- Pathology Department, Radboud University Nijmegen Medical CentreThe Netherlands
| | | | | | - Gerardo Botti
- Department of Pathology, Istituto Nazionale per lo Studio e la Cura dei Tumouri, ‘Fondazione G Pascale’Naples, Italy
| | - Fabiana Tatangelo
- Department of Pathology, Istituto Nazionale per lo Studio e la Cura dei Tumouri, ‘Fondazione G Pascale’Naples, Italy
| | - Paolo Delrio
- Colorectal Surgery Department, Istituto Nazionale per lo Studio e la Cura dei Tumouri, ‘Fondazione G Pascale’Naples, Italy
| | - Michele Maio
- Division of Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano TumouriSiena, Italy
| | - Luigi Laghi
- Molecular Gastroenterology and Department of Gastroenterology, Humanitas Clinical and Research CentreRozzano, Milan, Italy
| | - Fabio Grizzi
- Molecular Gastroenterology and Department of Gastroenterology, Humanitas Clinical and Research CentreRozzano, Milan, Italy
| | | | - Corrado D'Arrigo
- Department of Histopathology, Dorset County HospitalDorchester, UK
| | - Fernando Vidal-Vanaclocha
- CEU-San Pablo University School of Medicine and HM Hospital of Madrid Scientific Foundation, Institute of Applied Molecular Medicine (IMMA)Madrid, Spain
| | - Eva Zavadova
- Department of Oncology, Charles University in Prague, First Faculty of Medicine, Department of Oncology of the First Faculty of Medicine and General HospitalPrague, Czech Republic
| | | | - Pamela S Ohashi
- Ontario Cancer Institute and Campbell Family Institute for Cancer Research, Princess Margaret Hospital, University Health NetworkToronto, ON, Canada
| | - Sara Hafezi-Bakhtiari
- Department of Pathology and Laboratory Medicine, University Health NetworkToronto, ON, Canada
| | - Bradly G Wouters
- Department of Pathology and Laboratory Medicine, University Health NetworkToronto, ON, Canada
| | - Michael Roehrl
- UHN Program in BioSpecimen Sciences, University Health NetworkToronto, ON, Canada
| | - Linh Nguyen
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, University Health NetworkToronto, ON, Canada
| | - Yutaka Kawakami
- Division of Cellular Signalling, Institute for Advanced Medical Research, Keio University School of MedicineTokyo, Japan
| | - Shoichi Hazama
- Department of Digestive Surgery and Surgical Oncology, Yamaguchi University Graduate School of MedicineJapan
| | - Kiyotaka Okuno
- Department of Surgery, Kinki University School of MedicineOsaka-sayama, Japan
| | - Shuji Ogino
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School and Department of Medical Oncology, Dana-Farber Cancer InstituteBoston, MA, USA
| | - Peter Gibbs
- Department of Medical Oncology, Royal Melbourne HospitalAustralia
| | - Paul Waring
- Department of Pathology, University of MelbourneAustralia
| | - Noriyuki Sato
- Department of Pathology, Sapporo Medical University School of MedicineJapan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of MedicineJapan
| | - Kyogo Itoh
- Department of Immunology and Immunotherapy, Kurume University School of MedicineJapan
| | - Prabhu S Patel
- The Gujarat Cancer and Research InstituteAsarwa, Ahmedabad, India
| | - Shilin N Shukla
- The Gujarat Cancer and Research InstituteAsarwa, Ahmedabad, India
| | - Yili Wang
- Institute for Cancer Research, Centre of Translational Medicine, Xi'an Jiaotong UniversityXian, People's Republic of China
| | | | | | - Viorel Scripcariu
- Department of Surgery, University of Medicine and Pharmacy ‘Gr T Popa’, Department of Surgical Oncology, Regional Institute of OncologyIaşi, Romania
| | - Paolo A Ascierto
- Medical Oncology and Innovative Therapies Unit, Istituto Nazionale per lo Studio e la Cura dei Tumouri, Fondazione ‘G Pascale’Napoli, Italy
| | | | - Bernard A Fox
- Laboratory of Molecular and Tumour Immunology, Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical CenterPortland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science UniversityPortland, OR, USA
| | - Franck Pagès
- INSERM, U872, Laboratory of Integrative Cancer ImmunologyParis, France
- Université Paris DescartesParis, France
- Centre de Recherche des CordeliersUniversité Pierre et Marie Curie Paris 6, France
- Immunomonitoring Platform, Laboratory of Immunology, Georges Pompidou European HospitalParis, France
| |
Collapse
|
49
|
Ascierto PA, Kalos M, Schaer DA, Callahan MK, Wolchok JD. Biomarkers for immunostimulatory monoclonal antibodies in combination strategies for melanoma and other tumor types. Clin Cancer Res 2013; 19:1009-20. [PMID: 23460532 DOI: 10.1158/1078-0432.ccr-12-2982] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Modulation of the immune system by targeting coinhibitory and costimulatory receptors has become a promising new approach of immunotherapy for cancer. The recent approval of the CTLA-4-blocking antibody ipilimumab for the treatment of melanoma was a watershed event, opening up a new era in the field of immunotherapy. Ipilimumab was the first treatment to ever show enhanced overall survival (OS) for patients with stage IV melanoma. However, measuring response rates using standard Response Evaluation Criteria in Solid Tumors (RECIST) or modified World Health Organization criteria or progression-free survival does not accurately capture the potential for clinical benefit for ipilimumab-treated patients. As immunotherapy approaches are translated into more tumor types, it is important to study biomarkers, which may be more predictive of OS to identify the patients most likely to have clinical benefit. Ipilimumab is the first-in-class of a series of immunomodulating antibodies that are in clinical development. Anti-PD1 (nivolumab and MK-3475), anti-PD-L1 (BMS-936 559, RG7446, and MEDI4736), anti-CD137 (urelumab), anti-OX40, anti-GITR, and anti-CD40 monoclonal antibodies are just some of the agents that are being actively investigated in clinical trials, each having the potential for combination with the ipilimumab to enhance its effectiveness. Development of rational combinations of immunomodulatory antibodies with small-molecule pathway inhibitor therapies such as vemurafenib makes the discovery of predictive biomarkers even more important. Identifying reliable biomarkers is a necessary step in personalizing the treatment of each patient's cancer through a baseline assessment of tumor gene expression and/or immune profile to optimize therapy for the best chance of therapeutic success.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori Fondazione G Pascale, Napoli, Italy.
| | | | | | | | | |
Collapse
|
50
|
Kutscher S, Dembek CJ, Deckert S, Russo C, Körber N, Bogner JR, Geisler F, Umgelter A, Neuenhahn M, Albrecht J, Cosma A, Protzer U, Bauer T. Overnight resting of PBMC changes functional signatures of antigen specific T- cell responses: impact for immune monitoring within clinical trials. PLoS One 2013; 8:e76215. [PMID: 24146841 PMCID: PMC3795753 DOI: 10.1371/journal.pone.0076215] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/21/2013] [Indexed: 11/19/2022] Open
Abstract
Polyfunctional CD4 or CD8 T cells are proposed to represent a correlate of immune control for persistent viruses as well as for vaccine mediated protection against infection. A well-suited methodology to study complex functional phenotypes of antiviral T cells is the combined staining of intracellular cytokines and phenotypic marker expression using polychromatic flow cytometry. In this study we analyzed the effect of an overnight resting period at 37°C on the quantity and functionality of HIV-1, EBV, CMV, HBV and HCV specific CD4 and CD8 T-cell responses in a cohort of 21 individuals. We quantified total antigen specific T cells by multimer staining and used 10-color intracellular cytokine staining (ICS) to determine IFNγ, TNFα, IL2 and MIP1β production. After an overnight resting significantly higher numbers of functionally active T cells were detectable by ICS for all tested antigen specificities, whereas the total number of antigen specific T cells determined by multimer staining remained unchanged. Overnight resting shifted the quality of T-cell responses towards polyfunctionality and increased antigen sensitivity of T cells. Our data suggest that the observed effect is mediated by T cells rather than by antigen presenting cells. We conclude that overnight resting of PBMC prior to ex vivo analysis of antiviral T-cell responses represents an efficient method to increase sensitivity of ICS-based methods and has a prominent impact on the functional phenotype of T cells.
Collapse
Affiliation(s)
- Sarah Kutscher
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
| | - Claudia J. Dembek
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Simone Deckert
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
| | - Carolina Russo
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Nina Körber
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Johannes R. Bogner
- Department of Infectious Diseases/Med. Klinik und Poliklinik, University Hospital of Munich/Ludwig Maximilians Universität, Munich, Germany
| | - Fabian Geisler
- Department of Internal Medicine II, Klinikum rechts der Isar/Technische Universität München, Munich, Germany
| | - Andreas Umgelter
- Department of Internal Medicine II, Klinikum rechts der Isar/Technische Universität München, Munich, Germany
| | - Michael Neuenhahn
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
- Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - Julia Albrecht
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
- Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | | | - Ulrike Protzer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Tanja Bauer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
- * E-mail:
| |
Collapse
|