1
|
Bilich T, Nelde A, Bauer J, Walz S, Roerden M, Salih HR, Weisel K, Besemer B, Marcu A, Lübke M, Schuhmacher J, Neidert MC, Rammensee HG, Stevanović S, Walz JS. Mass spectrometry-based identification of a B-cell maturation antigen-derived T-cell epitope for antigen-specific immunotherapy of multiple myeloma. Blood Cancer J 2020; 10:24. [PMID: 32111817 PMCID: PMC7048774 DOI: 10.1038/s41408-020-0288-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 10/28/2019] [Indexed: 01/10/2023] Open
Abstract
The B-cell maturation antigen (BCMA) is currently being evaluated as promising tumor-associated surface antigen for T-cell-based immunotherapy approaches, such as CAR T cells and bispecific antibodies, in multiple myeloma (MM). Cytotoxic T cells bearing BCMA-specific T-cell receptors might further allow targeting HLA-presented antigens derived from the intracellular domain of BCMA. By analyzing a mass spectrometry-acquired immunopeptidome dataset of primary MM samples and MM cell lines for BCMA-derived HLA ligands, we identified the naturally presented HLA-B*18-restricted ligand P(BCMA)B*18. Additionally, P(BCMA)B*18 was identified on primary CLL samples, thereby expanding the range for possible applications. P(BCMA)B*18 induced multifunctional BCMA-specific cells de novo from naïve CD8+ T cells of healthy volunteers. These T cells exhibited antigen-specific lysis of autologous peptide-loaded cells. Even in the immunosuppressive context of MM, we detected spontaneous memory T-cell responses against P(BCMA)B*18 in patients. By applying CTLA-4 and PD-1 inhibition in vitro we induced multifunctional P(BCMA)B*18-specific CD8+ T cells in MM patients lacking preexisting BCMA-directed immune responses. Finally, we could show antigen-specific lysis of autologous peptide-loaded target cells and even MM.1S cells naturally presenting P(BCMA)B*18 using patient-derived P(BCMA)B*18-specific T cells. Hence, this BCMA-derived T-cell epitope represents a promising target for T-cell-based immunotherapy and monitoring following immunotherapy in B-cell malignancy patients.
Collapse
Affiliation(s)
- Tatjana Bilich
- University Hospital Tübingen, Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tübingen, Germany
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Annika Nelde
- University Hospital Tübingen, Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tübingen, Germany
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Jens Bauer
- University Hospital Tübingen, Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tübingen, Germany
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Simon Walz
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
- University Hospital Tübingen, Department of Urology, Tübingen, Germany
| | - Malte Roerden
- University Hospital Tübingen, Department of Hematology and Oncology, Tübingen, Germany
| | - Helmut R Salih
- University Hospital Tübingen, Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tübingen, Germany
| | - Katja Weisel
- University Hospital Tübingen, Department of Hematology and Oncology, Tübingen, Germany
- University Hospital Hamburg-Eppendorf, Department of Oncology, Hamburg-Eppendorf, Germany
| | - Britta Besemer
- University Hospital Tübingen, Department of Hematology and Oncology, Tübingen, Germany
| | - Ana Marcu
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Maren Lübke
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Juliane Schuhmacher
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Marian C Neidert
- University Hospital Zurich and University of Zurich, Department of Neurosurgery, Clinical Neuroscience Center, Zurich, Switzerland
| | - Hans-Georg Rammensee
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Stefan Stevanović
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Juliane S Walz
- University Hospital Tübingen, Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tübingen, Germany.
| |
Collapse
|
2
|
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
|
3
|
Wen PY, Reardon DA, Armstrong TS, Phuphanich S, Aiken RD, Landolfi JC, Curry WT, Zhu JJ, Glantz M, Peereboom DM, Markert JM, LaRocca R, O'Rourke DM, Fink K, Kim L, Gruber M, Lesser GJ, Pan E, Kesari S, Muzikansky A, Pinilla C, Santos RG, Yu JS. A Randomized Double-Blind Placebo-Controlled Phase II Trial of Dendritic Cell Vaccine ICT-107 in Newly Diagnosed Patients with Glioblastoma. Clin Cancer Res 2019; 25:5799-5807. [PMID: 31320597 DOI: 10.1158/1078-0432.ccr-19-0261] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/30/2019] [Accepted: 07/11/2019] [Indexed: 01/20/2023]
Abstract
PURPOSE To evaluate the results of the randomized, double-blind, placebo-controlled phase II clinical trial of ICT-107 in patients with newly diagnosed glioblastoma. PATIENTS AND METHODS We conducted a double-blinded randomized phase II trial of ICT-107 in newly diagnosed patients with glioblastoma (GBM) and tested efficacy, safety, quality of life (QoL), and immune response. HLA-A1+ and/or -A2+-resected patients with residual tumor ≤1 cm3 received radiotherapy and concurrent temozolomide. Following completion of radiotherapy, 124 patients, randomized 2:1, received ICT-107 [autologous dendritic cells (DC) pulsed with six synthetic peptide epitopes targeting GBM tumor/stem cell-associated antigens MAGE-1, HER-2, AIM-2, TRP-2, gp100, and IL13Rα2] or matching control (unpulsed DC). Patients received induction ICT-107 or control weekly × 4 followed by 12 months of adjuvant temozolomide. Maintenance vaccinations occurred at 1, 3, and 6 months and every 6 months thereafter. RESULTS ICT-107 was well tolerated, with no difference in adverse events between the treatment and control groups. The primary endpoint, median overall survival (OS), favored ICT-107 by 2.0 months in the intent-to-treat (ITT) population but was not statistically significant. Progression-free survival (PFS) in the ITT population was significantly increased in the ICT-107 cohort by 2.2 months (P = 0.011). The frequency of HLA-A2 primary tumor antigen expression was higher than that for HLA-A1 patients, and HLA-A2 patients had higher immune response (via Elispot). HLA-A2 patients achieved a meaningful therapeutic benefit with ICT-107, in both the MGMT methylated and unmethylated prespecified subgroups, whereas only HLA-A1 methylated patients had an OS benefit. CONCLUSIONS PFS was significantly improved in ICT-107-treated patients with maintenance of QoL. Patients in the HLA-A2 subgroup showed increased ICT-107 activity clinically and immunologically.
Collapse
Affiliation(s)
- Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - David A Reardon
- Center For Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Robert D Aiken
- Rutgers-Cancer Institute of New Jersey, New Brunswick, New Jersey
| | | | | | - Jay-Jiguang Zhu
- University of Texas Health Sciences Center at Houston (UTHealth), Houston, Texas
| | - Michael Glantz
- Penn State Hershey Medical Center, Hershey, Pennsylvania
| | | | | | | | - Donald M O'Rourke
- Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Karen Fink
- Baylor Scott and White Neuro-Oncology Associates, Dallas, Texas
| | - Lyndon Kim
- Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | | | - Glenn J Lesser
- Wake Forest Baptist Medical Center, Winston-Salem, North Carolina
| | - Edward Pan
- University of Texas, Southwest Medical Center, Dallas, Texas
| | - Santosh Kesari
- John Wayne Cancer Institute and Pacific Neuroscience Institute, Santa Monica, California
| | - Alona Muzikansky
- Alona Muzikansky, Massachusetts General Hospital, Boston, Massachusetts
| | - Clemencia Pinilla
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Radleigh G Santos
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - John S Yu
- Cedars-Sinai Medical Center, Los Angeles, California.,Immunocellular Therapeutics, Calabasas, California.,Precision Lifesciences Group, Nashville, TN
| |
Collapse
|
4
|
Staats J, Divekar A, McCoy JP, Maecker HT. Guidelines for Gating Flow Cytometry Data for Immunological Assays. Methods Mol Biol 2019; 2032:81-104. [PMID: 31522414 DOI: 10.1007/978-1-4939-9650-6_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
"Gating" refers to the selection of successive subpopulations of cells for analysis in flow cytometry. It is usually performed manually, based on expert knowledge of cell characteristics. However, there can be considerable disagreement in how gates should be applied, even between individuals experienced in the field. While clinical software often automates gating, and some guidelines do exist (especially for clinical assays), there are no comprehensive guidelines across the various types of immunological assays performed using flow cytometry. Here we attempt to provide such guidelines, focused on the most general and pervasive types of gates, why they are important, and what recommendations can be made regarding their use. We do so through the display of example data, collected by academic, government, and industry representatives. These guidelines should be of value to both novice and experienced flow cytometrists analyzing a wide variety of immunological assays.
Collapse
Affiliation(s)
- Janet Staats
- Duke Immune Profiling Core, Duke University Medical Center, Durham, NC, USA
| | - Anagha Divekar
- Department for Cellular Analysis, Biolegend, San Diego, CA, USA
| | | | - Holden T Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
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
|
6
|
Chandran PA, Laske K, Cazaly A, Rusch E, Schmid-Horch B, Rammensee HG, Ottensmeier CH, Gouttefangeas C. Validation of Immunomonitoring Methods for Application in Clinical Studies: The HLA-Peptide Multimer Staining Assay. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2018; 94:342-353. [PMID: 27363684 DOI: 10.1002/cyto.b.21397] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/27/2016] [Accepted: 06/28/2016] [Indexed: 11/11/2022]
Abstract
BACKGROUND Validated assays are essential to generate data with defined specificity, consistency, and reliability. Although the process of validation is required for applying immunoassays in the context of clinical studies, reports on systematic validation of in vitro T cell assays are scarce so far. We recently validated our HLA-peptide multimer staining assay in a systematic manner so as to qualify the method for monitoring antigen-specific T cell responses after immunotherapy. METHODS Parameters of the assay, specificity, precision, linearity, sensitivity, and robustness were assessed systematically. Experiments were designed to address specifically each parameter and are detailed. RESULTS Nonspecific multimer staining was below the acceptance limit of 0.02% multimer(+) CD8(+) cells. The assay showed acceptable precision in all dimensions it was repeated (CV < 10%) and also demonstrated a linear detection (R2 > 0.99) of antigen specific cells. CONCLUSIONS We succeeded in validating the HLA-multimer staining assay in a systematic manner. Additionally, we propose a technical framework and recommendations that can be applied for validating other T cell assessment methods. © 2016 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- P Anoop Chandran
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | - Karoline Laske
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | - Angelica Cazaly
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, United Kingdom
| | - Elisa Rusch
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | | | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | - Christian H Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, United Kingdom
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| |
Collapse
|
7
|
Scheikl-Gatard T, Tosch C, Lemonnier F, Rooke R. Identification of new MUC1 epitopes using HLA-transgenic animals: implication for immunomonitoring. J Transl Med 2017; 15:154. [PMID: 28679396 PMCID: PMC5499006 DOI: 10.1186/s12967-017-1254-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 06/24/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The success of immunotherapeutics in oncology and the search for further improvements has prompted revisiting the use of cancer vaccines. In this context, knowledge of the immunogenic epitopes and the monitoring of the immune response cancer vaccines generate are essential. MUC1 has been considered one of the most important tumor associated antigen for decades. METHODS To identify HLA-restricted MUC1 peptides we used eight human MHC class I transgenic mouse lines, together covering more than 80% of the human population. MUC1 peptides were identified by vaccinating each line with full length MUC1 coding sequences and using an IFNγ ELIspot restimulation assay. Relevant peptides were tested in a flow cytometry-based tetramer assay and for their capacity to serve as target in an in vivo killing assay. RESULTS Four previously identified MUC1 peptides were confirmed and five are described here for the first time. These nine peptide-MHC combinations were further characterized. Six gave above-background tetramer staining of splenocytes from immunized animals and three peptides were induced more than 5% specific in vivo killing. CONCLUSIONS These data describe for the first time five new HLA class I-restricted peptides and revisit some that were previously described. They also emphasize the importance of using in vivo/ex vivo models to screen for immunogenic peptides and define the functions for individual peptide-HLA combinations.
Collapse
Affiliation(s)
| | - Caroline Tosch
- Transgene SA, 400 Bld Gonthier d'Andernach, 67400, Illkirch Graffenstaden, France
| | - François Lemonnier
- Unité INSERM 1016, Département Endocrinologie, Métabolisme et Diabète. Equipe Immunologie des Diabètes, Bâtiment Cassini, 123 Bd Port Royal, 75014, Paris, France
| | - Ronald Rooke
- Institut de Recherche Servier, 125 Chemin de Ronde, 78290, Croissy, France.
| |
Collapse
|
8
|
Matko S, Teichert M, Tunger A, Schmitz M, Bornhauser M, Tonn T, Odendahl M. Enumeration of WT1-specific CD8 + T cells for clinical application using an MHC Streptamer based no-wash single-platform flow-cytometric assay. Cytometry A 2017; 91:1001-1008. [PMID: 28544366 DOI: 10.1002/cyto.a.23146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/02/2017] [Accepted: 05/11/2017] [Indexed: 11/08/2022]
Abstract
The advent of novel strategies to generate leukemia-associated-antigen (LAA)-specific T cells for adoptive immunotherapies creates a demand for standardized good laboratory practice (GLP)-compliant enumeration assays to provide a secure clinical environment-whether it is to identify potential donors, define therapeutic doses for transplantation, or monitor clinical success. Here, we introduce a no-wash assay based on single-platform cell enumeration and Streptamer staining to determine the Wilms' tumor antigen 1 (WT1)-specific T cell immunity in clinical samples. We analyzed the performance of the WT1-specific MHC Streptamers in direct comparison to CMV- and EBV-specific MHC Streptamer staining by spiking antigen-specific T cells in PBMCs. The accuracy of the assay was high for all performed experiments with a mean recovery of 94% and a linear regression of 0.988. Differences were apparent regarding the limit of detection/quantification (LOD/LOQ). While results obtained for WT1 yielded an LOD/LOQ of 0.08 ± 0.04% and 0.11 ± 0.06% (1.33 ± 0.32 cells/µl and 1.9 ± 0.14 cells/µl), the overall LOD/LOQ was notably lower and accounted to 0.02 ± 0.02% and 0.05 ± 0.03% (0.60 ± 0.03 cells/µl and 1.27 ± 0.58 cells/µl). Subsequent screening of 22 healthy individuals revealed significantly higher values for WT1 (0.04 ± 0.02% and 1.5 ± 0.9 cells/µl) than for the irrelevant HIV pol (0.016 ± 0.01% and 0.5 ± 0.4 cells/µl). In contrast, no increased frequencies were observed for WT1-specific T cells compared to HIV-specific T cells using a classical wash-protocol. These findings strongly suggest the use of no-wash single-platform assays in combination with MHC Streptamer staining for the detection of low affinity LAA-specific T cells due to its high accuracy and sensitivity. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Sarah Matko
- Department for Experimental Transfusion Medicine, German Red Cross Blood Donation Service North-East, Dresden, Germany.,Institute for Transfusion Medicine, Medical Faculty, Technical University (TU) Dresden, Germany
| | - Madeleine Teichert
- Department for Experimental Transfusion Medicine, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Antje Tunger
- Institute of Immunology, Medical Faculty, TU Dresden, Germany.,National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty, TU Dresden, Germany.,National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Germany.,German Consortium for Translational Cancer Research (DKTK) Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Bornhauser
- National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Germany.,German Consortium for Translational Cancer Research (DKTK) Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Torsten Tonn
- Department for Experimental Transfusion Medicine, German Red Cross Blood Donation Service North-East, Dresden, Germany.,Institute for Transfusion Medicine, Medical Faculty, Technical University (TU) Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Germany.,German Consortium for Translational Cancer Research (DKTK) Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcus Odendahl
- Department for Experimental Transfusion Medicine, German Red Cross Blood Donation Service North-East, Dresden, Germany
| |
Collapse
|
9
|
Masucci GV, Cesano A, Hawtin R, Janetzki S, Zhang J, Kirsch I, Dobbin KK, Alvarez J, Robbins PB, Selvan SR, Streicher HZ, Butterfield LH, Thurin M. Validation of biomarkers to predict response to immunotherapy in cancer: Volume I - pre-analytical and analytical validation. J Immunother Cancer 2016; 4:76. [PMID: 27895917 PMCID: PMC5109744 DOI: 10.1186/s40425-016-0178-1] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/20/2016] [Indexed: 12/31/2022] Open
Abstract
Immunotherapies have emerged as one of the most promising approaches to treat patients with cancer. Recently, there have been many clinical successes using checkpoint receptor blockade, including T cell inhibitory receptors such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death-1 (PD-1). Despite demonstrated successes in a variety of malignancies, responses only typically occur in a minority of patients in any given histology. Additionally, treatment is associated with inflammatory toxicity and high cost. Therefore, determining which patients would derive clinical benefit from immunotherapy is a compelling clinical question. Although numerous candidate biomarkers have been described, there are currently three FDA-approved assays based on PD-1 ligand expression (PD-L1) that have been clinically validated to identify patients who are more likely to benefit from a single-agent anti-PD-1/PD-L1 therapy. Because of the complexity of the immune response and tumor biology, it is unlikely that a single biomarker will be sufficient to predict clinical outcomes in response to immune-targeted therapy. Rather, the integration of multiple tumor and immune response parameters, such as protein expression, genomics, and transcriptomics, may be necessary for accurate prediction of clinical benefit. Before a candidate biomarker and/or new technology can be used in a clinical setting, several steps are necessary to demonstrate its clinical validity. Although regulatory guidelines provide general roadmaps for the validation process, their applicability to biomarkers in the cancer immunotherapy field is somewhat limited. Thus, Working Group 1 (WG1) of the Society for Immunotherapy of Cancer (SITC) Immune Biomarkers Task Force convened to address this need. In this two volume series, we discuss pre-analytical and analytical (Volume I) as well as clinical and regulatory (Volume II) aspects of the validation process as applied to predictive biomarkers for cancer immunotherapy. To illustrate the requirements for validation, we discuss examples of biomarker assays that have shown preliminary evidence of an association with clinical benefit from immunotherapeutic interventions. The scope includes only those assays and technologies that have established a certain level of validation for clinical use (fit-for-purpose). Recommendations to meet challenges and strategies to guide the choice of analytical and clinical validation design for specific assays are also provided.
Collapse
Affiliation(s)
- Giuseppe V Masucci
- Department of Oncology-Pathology, Karolinska Institutet, 171 76 Stockholm, Sweden
| | | | - Rachael Hawtin
- Nodality, Inc, 170 Harbor Way, South San Francisco, 94080 CA USA
| | - Sylvia Janetzki
- ZellNet Consulting, Inc, 555 North Avenue, Fort Lee, 07024 NJ USA
| | - Jenny Zhang
- Covaris Inc, 14 Gill St, Woburn, MA 01801 USA
| | - Ilan Kirsch
- Adaptive Biotechnologies, Inc, 1551 Eastlake Ave. E, Seattle, WA 98102 USA
| | - Kevin K Dobbin
- Department of Epidemiology and Biostatistics, College of Public Health, The University of Georgia, 101 Buck Road, Athens, 30602 GA USA
| | - John Alvarez
- Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | | | - Senthamil R Selvan
- Omni Array Biotechnology, 15601 Crabbs Branch Way, Rockville, 20855 MD USA
| | - Howard Z Streicher
- National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, 20892 MD USA
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Magdalena Thurin
- National Cancer Institute, Cancer Diagnosis Program, DCTD, National Institutes of Health, 9609 Medical Center Drive, Bethesda, 20892 MD USA ; Adaptive Biotechnologies, Inc, 1551 Eastlake Ave. E, Seattle, WA 98102 USA
| |
Collapse
|
10
|
Rampling R, Peoples S, Mulholland PJ, James A, Al-Salihi O, Twelves CJ, McBain C, Jefferies S, Jackson A, Stewart W, Lindner J, Kutscher S, Hilf N, McGuigan L, Peters J, Hill K, Schoor O, Singh-Jasuja H, Halford SE, Ritchie JWA. A Cancer Research UK First Time in Human Phase I Trial of IMA950 (Novel Multipeptide Therapeutic Vaccine) in Patients with Newly Diagnosed Glioblastoma. Clin Cancer Res 2016; 22:4776-4785. [PMID: 27225692 PMCID: PMC5026298 DOI: 10.1158/1078-0432.ccr-16-0506] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/11/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE To perform a two-cohort, phase I safety and immunogenicity study of IMA950 in addition to standard chemoradiotherapy and adjuvant temozolomide in patients with newly diagnosed glioblastoma. IMA950 is a novel glioblastoma-specific therapeutic vaccine containing 11 tumor-associated peptides (TUMAP), identified on human leukocyte antigen (HLA) surface receptors in primary human glioblastoma tissue. EXPERIMENTAL DESIGN Patients were HLA-A*02-positive and had undergone tumor resection. Vaccination comprised 11 intradermal injections with IMA950 plus granulocyte macrophage colony-stimulating factor (GM-CSF) over a 24-week period, beginning 7 to 14 days prior to initiation of chemoradiotherapy (Cohort 1) or 7 days after chemoradiotherapy (Cohort 2). Safety was assessed according to NCI CTCAE Version 4.0 and TUMAP-specific T-cell immune responses determined. Secondary observations included progression-free survival (PFS), pretreatment regulatory T cell (Treg) levels, and the effect of steroids on T-cell responses. RESULTS Forty-five patients were recruited. Related adverse events included minor injection site reactions, rash, pruritus, fatigue, neutropenia and single cases of allergic reaction, anemia and anaphylaxis. Two patients experienced grade 3 dose-limiting toxicity of fatigue and anaphylaxis. Of 40 evaluable patients, 36 were TUMAP responders and 20 were multi-TUMAP responders, with no important differences between cohorts. No effect of pretreatment Treg levels on IMA950 immunogenicity was observed, and steroids did not affect TUMAP responses. PFS rates were 74% at 6 months and 31% at 9 months. CONCLUSIONS IMA950 plus GM-CSF was well-tolerated with the primary immunogenicity endpoint of observing multi-TUMAP responses in at least 30% of patients exceeded. Further development of IMA950 is encouraged. Clin Cancer Res; 22(19); 4776-85. ©2016 AACRSee related commentary by Lowenstein and Castro, p. 4760.
Collapse
Affiliation(s)
- Roy Rampling
- University of Glasgow, Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Sharon Peoples
- Edinburgh Centre for Neuro-Oncology, Western General Hospital, Edinburgh, United Kingdom
| | - Paul J Mulholland
- Department of Oncology, University College London Hospitals, London, United Kingdom
| | - Allan James
- University of Glasgow, Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Omar Al-Salihi
- Adult Neuro-Oncology, Southampton University Hospitals NHS Trust, Southampton, United Kingdom
| | - Christopher J Twelves
- Cancer Research UK Clinical Centre, St James's University Hospital, Leeds, United Kingdom
| | - Catherine McBain
- The Christie NHS Foundation Trust, Withington, Manchester, United Kingdom
| | - Sarah Jefferies
- Cambridge Cancer Trials Centre, Oncology Clinical Trials, Addensbrooke's Hospital, Cambridge, United Kingdom
| | - Alan Jackson
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, United Kingdom
| | - Willie Stewart
- Department of Neuropathology, The Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Juha Lindner
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | - Norbert Hilf
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | - Lesley McGuigan
- Cancer Research UK Centre for Drug Development, London, United Kingdom
| | - Jane Peters
- Cancer Research UK Centre for Drug Development, London, United Kingdom
| | - Karen Hill
- Cancer Research UK Centre for Drug Development, London, United Kingdom
| | | | | | - Sarah E Halford
- Cancer Research UK Centre for Drug Development, London, United Kingdom
| | - James W A Ritchie
- Cancer Research UK Centre for Drug Development, London, United Kingdom.
| |
Collapse
|
11
|
Mandruzzato S, Brandau S, Britten CM, Bronte V, Damuzzo V, Gouttefangeas C, Maurer D, Ottensmeier C, van der Burg SH, Welters MJP, Walter S. Toward harmonized phenotyping of human myeloid-derived suppressor cells by flow cytometry: results from an interim study. Cancer Immunol Immunother 2016; 65:161-9. [PMID: 26728481 PMCID: PMC4726716 DOI: 10.1007/s00262-015-1782-5] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/12/2015] [Indexed: 01/02/2023]
Abstract
There is an increasing interest for monitoring circulating myeloid-derived suppressor cells (MDSCs) in cancer patients, but there are also divergences in their phenotypic definition. To overcome this obstacle, the Cancer Immunoguiding Program under the umbrella of the Association of Cancer Immunotherapy is coordinating a proficiency panel program that aims at harmonizing MDSC phenotyping. After a consultation period, a two-stage approach was designed to harmonize MDSC phenotype. In the first step, an international consortium of 23 laboratories immunophenotyped 10 putative MDSC subsets on pretested, peripheral blood mononuclear cells of healthy donors to assess the level of concordance and define robust marker combinations for the identification of circulating MDSCs. At this stage, no mandatory requirements to standardize reagents or protocols were introduced. Data analysis revealed a small intra-laboratory, but very high inter-laboratory variance for all MDSC subsets, especially for the granulocytic subsets. In particular, the use of a dead-cell marker altered significantly the reported percentage of granulocytic MDSCs, confirming that these cells are especially sensitive to cryopreservation and/or thawing. Importantly, the gating strategy was heterogeneous and associated with high inter-center variance. Overall, our results document the high variability in MDSC phenotyping in the multicenter setting if no harmonization/standardization measures are applied. Although the observed variability depended on a number of identified parameters, the main parameter associated with variation was the gating strategy. Based on these findings, we propose further efforts to harmonize marker combinations and gating parameters to identify strategies for a robust enumeration of MDSC subsets.
Collapse
Affiliation(s)
- Susanna Mandruzzato
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64, 35128, Padua, Italy.
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy.
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Cedrik M Britten
- TRON Translationale Onkologie an der Universitätsmedizin der Johannes Gutenberg-Universität Mainz GmbH, Mainz, Germany
- Cell Therapy Group, Immuno-Oncology and Combinations, GlaxoSmithKline, Stevenage, UK
| | - Vincenzo Bronte
- Section of Immunology, Department of Pathology and Diagnostics, Verona University Hospital, Verona, Italy
| | - Vera Damuzzo
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64, 35128, Padua, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | | | - Christian Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, Experimental Cancer Medicine Centre, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, UK
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marij J P Welters
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
12
|
Higuchi Y, Koya T, Yuzawa M, Yamaoka N, Mizuno Y, Yoshizawa K, Hirabayashi K, Kobayashi T, Sano K, Shimodaira S. Enzyme-Linked Immunosorbent Spot Assay for the Detection of Wilms' Tumor 1-Specific T Cells Induced by Dendritic Cell Vaccination. Biomedicines 2015; 3:304-315. [PMID: 28536414 PMCID: PMC5344226 DOI: 10.3390/biomedicines3040304] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/23/2015] [Accepted: 11/27/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Despite recent advances in cancer immunotherapy and the development of various assays for T cell assessment, a lack of universal standards within immune monitoring remains. The objective of this study was to evaluate the enzyme-linked immunosorbent spot (ELISpot) assay in comparison with major histocompatibility complex-tetramer analysis in the context of dendritic cell (DC)-based cancer immunotherapy. METHODS The ELISpot assay was performed on peripheral blood mononuclear cells to assess reproducibility, daily precision, and linearity using HLA-A*24:02-restricted Cytomegalovirus peptide. Wilms' tumor 1 (WT1) antigen-specific cytotoxic T cells were then evaluated by both the ELISpot assay and WT1 tetramer analysis in peripheral blood from 46 cancer patients who received DC vaccinations pulsed with human leukocyte antigen (HLA)-A*24:02-restricted modified WT1 peptides. RESULTS The ELISpot assay was proven to have reproducibility (coefficient of variation (CV) ranged from 7.4% to 16.3%), daily precision (CV ranged from 5.0% to 17.3%), and linearity (r = 0.96-0.98). WT1-specific immune responses were detected by the ELISpot assay in 34 out of 46 patients (73.9%) post-vaccination. A Spearman's rank-correlation coefficient of 0.82 between the ELISpot assay and WT1 tetramer analysis was obtained. CONCLUSION This is the first report of a comparison of an ELISpot assay and tetramer analysis in the context of dendritic cell (DC)-based cancer immunotherapy. The ELISpot assay has reproducibility, linearity, and excellent correlation with the WT1 tetramer analysis. These findings suggest that the validated ELISpot assay is useful to monitor the acquired immunity by DC vaccination targeting WT1.
Collapse
Affiliation(s)
- Yumiko Higuchi
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto 390-8621, Japan.
- Department of Health and Medical Sciences, Shinshu University Graduate School of Medicine, Matsumoto 390-8621, Japan.
| | - Terutsugu Koya
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto 390-8621, Japan.
| | - Miki Yuzawa
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto 390-8621, Japan.
| | - Naoko Yamaoka
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto 390-8621, Japan.
| | - Yumiko Mizuno
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto 390-8621, Japan.
| | - Kiyoshi Yoshizawa
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto 390-8621, Japan.
| | - Koichi Hirabayashi
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto 390-8621, Japan.
| | - Takashi Kobayashi
- Shinshu Cancer Center, Shinshu University Hospital, Matsumoto 390-8621, Japan.
| | - Kenji Sano
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto 390-8621, Japan.
| | - Shigetaka Shimodaira
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto 390-8621, Japan.
| |
Collapse
|
13
|
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
|
14
|
Abstract
Cancer vaccines are designed to promote tumor specific immune responses, particularly cytotoxic CD8 positive T cells that are specific to tumor antigens. The earliest vaccines, which were developed in 1994-95, tested non-mutated, shared tumor associated antigens that had been shown to be immunogenic and capable of inducing clinical responses in a minority of people with late stage cancer. Technological developments in the past few years have enabled the investigation of vaccines that target mutated antigens that are patient specific. Several platforms for cancer vaccination are being tested, including peptides, proteins, antigen presenting cells, tumor cells, and viral vectors. Standard of care treatments, such as surgery and ablation, chemotherapy, and radiotherapy, can also induce antitumor immunity, thereby having cancer vaccine effects. The monitoring of patients' immune responses at baseline and after standard of care treatment is shedding light on immune biomarkers. Combination therapies are being tested in clinical trials and are likely to be the best approach to improving patient outcomes.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Departments of Medicine, Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
15
|
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
|
16
|
Chudley L, McCann KJ, Coleman A, Cazaly AM, Bidmon N, Britten CM, van der Burg SH, Gouttefangeas C, Jandus C, Laske K, Maurer D, Romero P, Schröder H, Stynenbosch LFM, Walter S, Welters MJP, Ottensmeier CH. Harmonisation of short-term in vitro culture for the expansion of antigen-specific CD8(+) T cells with detection by ELISPOT and HLA-multimer staining. Cancer Immunol Immunother 2014; 63:1199-211. [PMID: 25134947 PMCID: PMC4209099 DOI: 10.1007/s00262-014-1593-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 08/01/2014] [Indexed: 10/31/2022]
Abstract
Ex vivo ELISPOT and multimer staining are well-established tests for the assessment of antigen-specific T cells. Many laboratories are now using a period of in vitro stimulation (IVS) to enhance detection. Here, we report the findings of a multi-centre panel organised by the Association for Cancer Immunotherapy Immunoguiding Program to investigate the impact of IVS protocols on the detection of antigen-specific T cells of varying ex vivo frequency. Five centres performed ELISPOT and multimer staining on centrally prepared PBMCs from 3 donors, both ex vivo and following IVS. A harmonised IVS protocol was designed based on the best-performing protocol(s), which was then evaluated in a second phase on 2 donors by 6 centres. All centres were able to reliably detect antigen-specific T cells of high/intermediate frequency both ex vivo (Phase I) and post-IVS (Phase I and II). The highest frequencies of antigen-specific T cells ex vivo were mirrored in the frequencies following IVS and in the detection rates. However, antigen-specific T cells of a low/undetectable frequency ex vivo were not reproducibly detected post-IVS. Harmonisation of the IVS protocol reduced the inter-laboratory variation observed for ELISPOT and multimer analyses by approximately 20 %. We further demonstrate that results from ELISPOT and multimer staining correlated after (P < 0.0001 and R (2) = 0.5113), but not before IVS. In summary, IVS was shown to be a reproducible method that benefitted from method harmonisation.
Collapse
Affiliation(s)
- Lindsey Chudley
- Cancer Sciences Unit, Faculty of Medicine, Experimental Cancer Medicine Centre, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD UK
| | - Katy J. McCann
- Cancer Sciences Unit, Faculty of Medicine, Experimental Cancer Medicine Centre, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD UK
| | - Adam Coleman
- Cancer Sciences Unit, Faculty of Medicine, Experimental Cancer Medicine Centre, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD UK
| | - Angelica M. Cazaly
- Cancer Sciences Unit, Faculty of Medicine, Experimental Cancer Medicine Centre, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD UK
| | - Nicole Bidmon
- Translational Oncology, University Medical Center, Johannes-Gutenberg University GmbH, Mainz, Germany
| | - Cedrik M. Britten
- Translational Oncology, University Medical Center, Johannes-Gutenberg University GmbH, Mainz, Germany
| | - Sjoerd H. van der Burg
- Department of Clinical Oncology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Cecile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, Eberhard-Karls University, Tübingen, Germany
| | - Camilla Jandus
- Translational Tumour Immunology, Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Karoline Laske
- Department of Immunology, Institute for Cell Biology, Eberhard-Karls University, Tübingen, Germany
| | | | - Pedro Romero
- Translational Tumour Immunology, Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Helene Schröder
- Translational Oncology, University Medical Center, Johannes-Gutenberg University GmbH, Mainz, Germany
| | | | | | - Marij J. P. Welters
- Department of Clinical Oncology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Christian H. Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, Experimental Cancer Medicine Centre, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD UK
- Somers Cancer Research Building (Mailpoint 824), Cancer Sciences Unit, Faculty of Medicine, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD UK
| |
Collapse
|
17
|
Baxevanis CN, Anastasopoulou EA, Voutsas IF, Papamichail M, Perez SA. Immune biomarkers: how well do they serve prognosis in human cancers? Expert Rev Mol Diagn 2014; 15:49-59. [PMID: 25345403 DOI: 10.1586/14737159.2015.965684] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In order to be optimally efficacious, therapeutic cancer vaccines must induce robust tumor-specific CD8(+) cytotoxic T cells, which are responsible for tumor cell lysis. Unlike cytotoxic drugs, which act directly on the tumor, cancer vaccines demonstrate new kinetics involving the generation of specific cellular immune responses, which need to be translated into antitumor responses to delay tumor progression and improve survival. These delayed kinetics of action establish a new concept of benefit in the long term, which implies a slow down in tumor growth rates, than a marked reduction in tumor size. Therefore, there is a significant need to identify intermediate biomarkers so that clinical responses can be evaluated in a timely manner. Therapeutic vaccination as a modality for cancer treatment has received significant attention with multiple clinical trials demonstrating improvements in overall survival. Significant challenges to this modality remain, including increasing vaccine potency and minimizing treatment-related toxicities and identifying prognostic and predictive biomarkers of clinical benefit that may guide to select and optimize the therapeutic strategies for patients most likely to gain benefit.
Collapse
Affiliation(s)
- Constantin N Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras avenue, Athens 11522, Greece
| | | | | | | | | |
Collapse
|
18
|
Borchers S, Ogonek J, Varanasi PR, Tischer S, Bremm M, Eiz-Vesper B, Koehl U, Weissinger EM. Multimer monitoring of CMV-specific T cells in research and in clinical applications. Diagn Microbiol Infect Dis 2013; 78:201-12. [PMID: 24331953 DOI: 10.1016/j.diagmicrobio.2013.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/11/2013] [Accepted: 11/04/2013] [Indexed: 10/26/2022]
Abstract
Multimer monitoring has become a standard technique for detection of antigen-specific T cells. The term "multimer" refers to a group of reagents based on the multimerisation of molecules in order to raise avidity and thus stabilize binding to their ligand. Multimers for detection of antigen-specific T-cell responses are based on major histocompatibility complex class I peptide complexes. Multimer staining enables fast and direct visualization of antigen-specific T cells; thus, it is widely applied to assess antiviral immunity, e.g., monitor patients in vaccination trials or confirm purity of cell products for adoptive transfer. Assessment of T-cell immunity against persistent pathogens like cytomegalovirus (CMV) is of major importance in immunosuppressed patients. Recent advancements of multimers facilitate reversible labeling and allow isolation of epitope-specific T cells for adoptive transfer. Here, we give an overview on the different multimers and their applications, with an emphasis on CMV-specific T-cell responses.
Collapse
Affiliation(s)
- Sylvia Borchers
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany; Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover, Germany; German Centre for Infection Research (DZIF), Partnerside Hannover-Braunschweig, Germany.
| | - Justyna Ogonek
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany.
| | - Pavankumar R Varanasi
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany; Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover, Germany; German Centre for Infection Research (DZIF), Partnerside Hannover-Braunschweig, Germany.
| | - Sabine Tischer
- Institute of Transfusion Medicine, MHH, Hannover, Germany.
| | - Melanie Bremm
- Pediatric Hematology and Oncology, Johann Wolfgang Goethe-University, Frankfurt, Germany.
| | - Britta Eiz-Vesper
- Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover, Germany; Institute of Transfusion Medicine, MHH, Hannover, Germany.
| | - Ulrike Koehl
- Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover, Germany; Institute for Cellular Therapeutics, MHH, Hannover, Germany.
| | - Eva M Weissinger
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany; Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover, Germany; German Centre for Infection Research (DZIF), Partnerside Hannover-Braunschweig, Germany.
| |
Collapse
|
19
|
Immune monitoring in cancer vaccine clinical trials: critical issues of functional flow cytometry-based assays. BIOMED RESEARCH INTERNATIONAL 2013; 2013:726239. [PMID: 24195078 PMCID: PMC3806162 DOI: 10.1155/2013/726239] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/19/2013] [Indexed: 11/17/2022]
Abstract
The development of immune monitoring assays is essential to determine the immune responses against tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs) and their possible correlation with clinical outcome in cancer patients receiving immunotherapies. Despite the wide range of techniques used, to date these assays have not shown consistent results among clinical trials and failed to define surrogate markers of clinical efficacy to antitumor vaccines. Multiparameter flow cytometry- (FCM-) based assays combining different phenotypic and functional markers have been developed in the past decade for informative and longitudinal analysis of polyfunctional T-cells. These technologies were designed to address the complexity and functional heterogeneity of cancer biology and cellular immunity and to define biomarkers predicting clinical response to anticancer treatment. So far, there is still a lack of standardization of some of these immunological tests. The aim of this review is to overview the latest technologies for immune monitoring and to highlight critical steps involved in some of the FCM-based cellular immune assays. In particular, our laboratory is focused on melanoma vaccine research and thus our main goal was the validation of a functional multiparameter test (FMT) combining different functional and lineage markers to be applied in clinical trials involving patients with melanoma.
Collapse
|
20
|
Britten CM, Walter S, Janetzki S. Immunological Monitoring to Rationally Guide AAV Gene Therapy. Front Immunol 2013; 4:273. [PMID: 24062741 PMCID: PMC3770921 DOI: 10.3389/fimmu.2013.00273] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 08/27/2013] [Indexed: 12/12/2022] Open
Abstract
Recent successes with adeno-associated virus (AAV)-based gene therapies fuel the hope for new treatments for hereditary diseases. Pre-existing as well as therapy-induced immune responses against both AAV and the encoded transgenes have been described and may impact on safety and efficacy of gene therapy approaches. Consequently, monitoring of vector- and transgene-specific immunity is mandated and may rationally guide clinical development. Next to the humoral immune response, the cellular response is central in our understanding of the host reaction in gene therapy. But in contrast to the monitoring of antibodies, which has matured over many decades, sensitive and robust monitoring of T cells is a relatively new development. To make cellular immune assessments fit for purpose, investigators need to know, control and report the critical assay variables that influence the results. In addition, the quality of immune assays needs to be continuously adjusted to allow for exploratory hypothesis generation in early stages and confirmatory hypothesis validation in later stages of clinical development. The concept of immune assay harmonization which includes use of field-wide benchmarks, harmonization guidelines, and external quality control can support the context-specific evolution of immune assays. Multi-center studies pose particular challenges to sample logistics and quality control of sample specimens. Cooperative groups need to define if immune assessments should be performed in one central facility, in peripheral labs or including a combination of both. Finally, engineered reference samples that contain a defined number of antigen-specific T cells may become broadly applicable tools to control assay performance over time or across institutions.
Collapse
Affiliation(s)
- Cedrik Michael Britten
- Translational Oncology, University Medical Center, Johannes Gutenberg-University Mainz (TRON gGmbH) , Mainz , Germany ; Association for Cancer Immunotherapy (CIMT) , Mainz , Germany
| | | | | |
Collapse
|
21
|
Janetzki S, Hoos A, Melief CJM, Odunsi K, Romero P, Britten CM. Structured reporting of T cell assay results. CANCER IMMUNITY 2013; 13:13. [PMID: 23882158 PMCID: PMC3718734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
|
22
|
Aarntzen EHJG, Bol K, Schreibelt G, Jacobs JFM, Lesterhuis WJ, Van Rossum MM, Adema GJ, Figdor CG, Punt CJA, De Vries IJM. Skin-test infiltrating lymphocytes early predict clinical outcome of dendritic cell-based vaccination in metastatic melanoma. Cancer Res 2012; 72:6102-10. [PMID: 23010076 DOI: 10.1158/0008-5472.can-12-2479] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The identification of responding patients early during treatment would improve the capability to develop effective new immunotherapies more rapidly. Here, we describe a bioassay that may link early T-cell-mediated immune responses to later clinical benefits. This bioassay rests upon the tenet of immunotherapy that tumor-specific effector T cells capable of invading peripheral tissue can recognize tumor antigens and exert cytotoxic functions there. To show its utility, we conducted a retrospective study of a large cohort of metastatic melanoma patients (n = 91) enrolled in dendritic cell (DC)-based vaccination protocols to examine a hypothesized correlation of posttreatment skin-infiltrating lymphocytes (SKIL) with overall survival (OS). Stringent immunologic criteria were defined to identify long-term survivors. The presence of tumor-associated antigen (TAA)-specific CD8(+) T cell populations within SKILs (criterion I) was highly predictive for long-term survival. Further restriction by selecting for the presence of TAA-specific CD8(+) T cells specifically recognizing tumor peptide (criterion II) was also associated with improved OS. Recognition of naturally processed antigen (criterion III) maximized the accuracy of the test, with a median OS of 24.1 versus 9.9 months (P = 0.001). Our results show that detailed characterization of SKILs can permit an accurate selection of metastatic melanoma patients who benefit most from DC-based vaccination. This simple and robust bioassay integrates multiple aspects of cellular functions that mediate effective immune responses, thereby offering an effective tool to rapidly identify patients who are responding to immunotherapy at an early stage of treatment.
Collapse
Affiliation(s)
- Erik H J G Aarntzen
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
|
24
|
Lasso P, Mesa D, Bolaños N, Cuéllar A, Guzmán F, Cucunuba Z, Rosas F, Velasco V, Thomas MC, López MC, González JM, Puerta CJ. Chagasic patients are able to respond against a viral antigen from influenza virus. BMC Infect Dis 2012; 12:198. [PMID: 22920436 PMCID: PMC3511223 DOI: 10.1186/1471-2334-12-198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 08/13/2012] [Indexed: 12/13/2022] Open
Abstract
Background Trypanosoma cruzi, the etiological agent of Chagas’ disease, is an obligate intracellular parasite which induces a CD8+ T cell immune response with secretion of cytokines and release of cytotoxic granules. Although an immune-suppressive effect of T. cruzi on the acute phase of the disease has been described, little is known about the capacity of CD8+ T cell from chronic chagasic patients to respond to a non-T. cruzi microbial antigen. Methods In the present paper, the frequency, phenotype and the functional activity of the CD8+ T cells specific from Flu-MP*, an influenza virus epitope, were determined in 13 chagasic patients and 5 healthy donors. Results The results show that Flu-MP* peptide specific CD8+ T cells were found with similar frequencies in both groups. In addition, Flu-MP* specific CD8+ T cells were distributed in the early or intermediate/late differentiation stages without showing enrichment of a specific sub-population. The mentioned Flu-MP* specific CD8+ T cells from chagasic patients were predominately TEM (CCR7- CD62L-), producing IL-2, IFNγ, CD107a/b and perforin, and did not present significant differences when compared with those from healthy donors. Conclusions Our results support the hypothesis that there is no CD8+ T cell nonspecific immune-suppression during chronic Chagas disease infection. Nonetheless, other viral antigens must be studied in order to confirm our findings.
Collapse
Affiliation(s)
- Paola Lasso
- Laboratorio de Parasitología Molecular, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
van der Burg SH, Kalos M, Gouttefangeas C, Janetzki S, Ottensmeier C, Welters MJP, Romero P, Britten CM, Hoos A. Harmonization of immune biomarker assays for clinical studies. Sci Transl Med 2012; 3:108ps44. [PMID: 22072636 DOI: 10.1126/scitranslmed.3002785] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Assays that measure a patient's immune response play an increasingly important role in the development of immunotherapies. The inherent complexity of these assays and independent protocol development between laboratories result in high data variability and poor reproducibility. Quality control through harmonization--based on integration of laboratory-specific protocols with standard operating procedures and assay performance benchmarks--is one way to overcome these limitations. Harmonization guidelines can be widely implemented to address assay performance variables. This process enables objective interpretation and comparison of data across clinical trial sites and also facilitates the identification of relevant immune biomarkers, guiding the development of new therapies.
Collapse
Affiliation(s)
- Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Developers of cancer immunotherapy have struggled for decades to achieve clinical success in using the patient's immune system to treat cancer. In the absence of a defined development paradigm for immunotherapies, conventional criteria established for chemotherapy were applied to these agents. This article summarizes the recent lessons for development of agents in the immunotherapy space, describes the systematic creation of a new clinical development paradigm for cancer immunotherapies and integrates this paradigm with the emerging methodological framework for a new clinical sub-specialty of immuno-oncology, which was driven by the collaborative work between the Cancer Immunotherapy Consortium (CIC) of the Cancer Research Institute in the US and the Association for Cancer Immunotherapy (CIMT) in Europe. This new framework provides a better defined development path and a foundation for more reproducible success of future therapies.
Collapse
Affiliation(s)
- Axel Hoos
- Cancer Immunotherapy Consortium (CIC; formerly Cancer Vaccine Consortium) of the Cancer Research Institute; New York, NY USA
| | - Cedrik Britten
- Association for Immunotherapy of Cancer; Mainz, Germany
- Ribological GmbH; Mainz, Germany
| |
Collapse
|
27
|
Abstract
During more than 25 years of application in immunological sciences, ELISPOT has been established as a routine, robust, versatile, and reliable assay. From basic research to clinical immune monitoring, ELISPOT is being used to address the quantification and (to a lesser extent) functional characterization of immune cells secreting different molecules in the context of health and disease, immune intervention, and therapy in humans and other species [Kalyuzhny (Ed.) (2005) Handbook of Elispot: methods and protocols, Vol. 302, Humana Press Inc., Totowa, NJ]. Over the last decade, ELISPOT assays have been increasingly implemented as an immune-monitoring tool in clinical trials [Schmittel et al. J Immunother 23:289-295, 2000; Whiteside Immunol Invest 29:149-162, 2000; Nagata et al. Ann N Y Acad Sci 1037:10-15, 2004; Cox et al. (2005) Cellular immune assays for evaluation of vaccine efficacy in developing countries., In Manual of Clinical Immunology Laboratory (Rose, N. R., Hamilton, R. G., and Detrick, B., Eds.), p 301, ASM Press, Washington, DC; Cox et al. Methods 38:274-282, 2006]. While the principles of the original protocol have changed little since its first introduction [Czerkinsky J Immunol Methods 110:29-36, 1988], individual laboratories have adapted assay procedures based on experimental needs, availability of reagents and equipment, obtained recommendations, and gained experience, leading to a wide disparity of applied ELISPOT protocols with inevitable consequences. This chapter addresses the resulting challenges for ELISPOT use in clinical trial settings, and discusses the influence of harmonization strategies as a tool for overcoming these challenges. Furthermore, harmonization is discussed in the context of assay standardization and validation strategies.
Collapse
|
28
|
Fox BA, Schendel DJ, Butterfield LH, Aamdal S, Allison JP, Ascierto PA, Atkins MB, Bartunkova J, Bergmann L, Berinstein N, Bonorino CC, Borden E, Bramson JL, Britten CM, Cao X, Carson WE, Chang AE, Characiejus D, Choudhury AR, Coukos G, de Gruijl T, Dillman RO, Dolstra H, Dranoff G, Durrant LG, Finke JH, Galon J, Gollob JA, Gouttefangeas C, Grizzi F, Guida M, Håkansson L, Hege K, Herberman RB, Hodi FS, Hoos A, Huber C, Hwu P, Imai K, Jaffee EM, Janetzki S, June CH, Kalinski P, Kaufman HL, Kawakami K, Kawakami Y, Keilholtz U, Khleif SN, Kiessling R, Kotlan B, Kroemer G, Lapointe R, Levitsky HI, Lotze MT, Maccalli C, Maio M, Marschner JP, Mastrangelo MJ, Masucci G, Melero I, Melief C, Murphy WJ, Nelson B, Nicolini A, Nishimura MI, Odunsi K, Ohashi PS, O'Donnell-Tormey J, Old LJ, Ottensmeier C, Papamichail M, Parmiani G, Pawelec G, Proietti E, Qin S, Rees R, Ribas A, Ridolfi R, Ritter G, Rivoltini L, Romero PJ, Salem ML, Scheper RJ, Seliger B, Sharma P, Shiku H, Singh-Jasuja H, Song W, Straten PT, Tahara H, Tian Z, van Der Burg SH, von Hoegen P, Wang E, Welters MJP, Winter H, Withington T, Wolchok JD, Xiao W, Zitvogel L, et alFox BA, Schendel DJ, Butterfield LH, Aamdal S, Allison JP, Ascierto PA, Atkins MB, Bartunkova J, Bergmann L, Berinstein N, Bonorino CC, Borden E, Bramson JL, Britten CM, Cao X, Carson WE, Chang AE, Characiejus D, Choudhury AR, Coukos G, de Gruijl T, Dillman RO, Dolstra H, Dranoff G, Durrant LG, Finke JH, Galon J, Gollob JA, Gouttefangeas C, Grizzi F, Guida M, Håkansson L, Hege K, Herberman RB, Hodi FS, Hoos A, Huber C, Hwu P, Imai K, Jaffee EM, Janetzki S, June CH, Kalinski P, Kaufman HL, Kawakami K, Kawakami Y, Keilholtz U, Khleif SN, Kiessling R, Kotlan B, Kroemer G, Lapointe R, Levitsky HI, Lotze MT, Maccalli C, Maio M, Marschner JP, Mastrangelo MJ, Masucci G, Melero I, Melief C, Murphy WJ, Nelson B, Nicolini A, Nishimura MI, Odunsi K, Ohashi PS, O'Donnell-Tormey J, Old LJ, Ottensmeier C, Papamichail M, Parmiani G, Pawelec G, Proietti E, Qin S, Rees R, Ribas A, Ridolfi R, Ritter G, Rivoltini L, Romero PJ, Salem ML, Scheper RJ, Seliger B, Sharma P, Shiku H, Singh-Jasuja H, Song W, Straten PT, Tahara H, Tian Z, van Der Burg SH, von Hoegen P, Wang E, Welters MJP, Winter H, Withington T, Wolchok JD, Xiao W, Zitvogel L, Zwierzina H, Marincola FM, Gajewski TF, Wigginton JM, Disis ML. Defining the critical hurdles in cancer immunotherapy. J Transl Med 2011; 9:214. [PMID: 22168571 PMCID: PMC3338100 DOI: 10.1186/1479-5876-9-214] [Show More Authors] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 12/14/2011] [Indexed: 02/07/2023] Open
Abstract
Scientific discoveries that provide strong evidence of antitumor effects in preclinical models often encounter significant delays before being tested in patients with cancer. While some of these delays have a scientific basis, others do not. We need to do better. Innovative strategies need to move into early stage clinical trials as quickly as it is safe, and if successful, these therapies should efficiently obtain regulatory approval and widespread clinical application. In late 2009 and 2010 the Society for Immunotherapy of Cancer (SITC), convened an "Immunotherapy Summit" with representatives from immunotherapy organizations representing Europe, Japan, China and North America to discuss collaborations to improve development and delivery of cancer immunotherapy. One of the concepts raised by SITC and defined as critical by all parties was the need to identify hurdles that impede effective translation of cancer immunotherapy. With consensus on these hurdles, international working groups could be developed to make recommendations vetted by the participating organizations. These recommendations could then be considered by regulatory bodies, governmental and private funding agencies, pharmaceutical companies and academic institutions to facilitate changes necessary to accelerate clinical translation of novel immune-based cancer therapies. The critical hurdles identified by representatives of the collaborating organizations, now organized as the World Immunotherapy Council, are presented and discussed in this report. Some of the identified hurdles impede all investigators; others hinder investigators only in certain regions or institutions or are more relevant to specific types of immunotherapy or first-in-humans studies. Each of these hurdles can significantly delay clinical translation of promising advances in immunotherapy yet if overcome, have the potential to improve outcomes of patients with cancer.
Collapse
Affiliation(s)
- Bernard A Fox
- Earle A. Chiles Research Institute, Robert W. Franz Research Center, Providence Cancer Center, Providence Portland Medical Center, Portland, OR, USA
- Department of Molecular Microbiology and Immunology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Dolores J Schendel
- Institute of Molecular Immunology and Clinical Cooperation Group "Immune Monitoring", Helmholtz Centre Munich, German Research Center for Environmental Health, Munich, Germany
| | - Lisa H Butterfield
- Departments of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Surgery University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Steinar Aamdal
- Department of Clinical Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - James P Allison
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Paolo Antonio Ascierto
- Medical Oncology and Innovative Therapy, Instituto Nazionale Tumori-Fondazione 'G. Pascale', Naples, Italy
| | - Michael B Atkins
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jirina Bartunkova
- Institute of Immunology, FOCIS Center of Excellence, 2nd Medical School, Charles University, Prague, Czech Republic
| | - Lothar Bergmann
- Goethe Universität Frankfurt Am Main,Medizinische Klinik II, Frankfurt Am Main, Germany
| | | | - Cristina C Bonorino
- Instituto Nacional para o Controle do Câncer, Instituto de Pesquisas Biomédicas, PUCRS Faculdade de Biociências, PUCRS, Porto Alegre RS Brazil
| | - Ernest Borden
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
- Department of Solid Tumor Oncology, Cleveland Clinic, Cleveland, OH, USA
| | | | - Cedrik M Britten
- University Medical Center Mainz, III. Medical Department, Mainz, Germany
- Ribological GmbH, Mainz, Germany
| | - Xuetao Cao
- Chinese Academy of Medical Sciences, Beijing, China
- Institute of Immunology, National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, China
| | | | - Alfred E Chang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI
| | | | | | - George Coukos
- Ovarian Cancer Research Center, University of Pennsylvania Medical Center, Philadelphia, A, USA
| | - Tanja de Gruijl
- Department of Medical Oncology, VU Medical Center, Cancer Center Amsterdam Amsterdam, The Netherlands
| | - Robert O Dillman
- Hoag Institute for Research and Education, Hoag Cancer Institute, Newport Beach, CA, USA
| | - Harry Dolstra
- Department of Laboratory Medicine, Nijmegen Centre for Molecular Life Sciences, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Glenn Dranoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lindy G Durrant
- Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - James H Finke
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Jerome Galon
- INSERM U872, Cordeliers Research Center, Paris, France
| | | | - Cécile Gouttefangeas
- Institute for Cell Biology, Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | | | | | - Leif Håkansson
- University of Lund, Lund, Sweden
- CanImGuide Therapeutics AB, Hoellviken, Sweden
| | - Kristen Hege
- University of California, San Francisco, CA and Celgene Corporation, San Francisco, CA, USA
| | | | - F Stephen Hodi
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Axel Hoos
- Bristol-Myers Squibb Company, Wallingford, Connecticut, USA
| | - Christoph Huber
- Translational Oncology & Immunology Centre TRON at the Mainz University Medical Center, Mainz, Germany
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Kohzoh Imai
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Elizabeth M Jaffee
- Department of Oncology, the Sidney Kimmel Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | - Carl H June
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pawel Kalinski
- Department of Surgery University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Howard L Kaufman
- Rush University Cancer Center, Rush University Medical Center, Chicago, IL, USA
| | - Koji Kawakami
- School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Ulrich Keilholtz
- Dept. of Hematology and Medical Oncology, Charité Comprehensive Cancer Center, Berlin, Germany
| | | | - Rolf Kiessling
- Department of Oncology - Pathology, Cancer Center Karolinska, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Beatrix Kotlan
- Department of Molecular Immunology and Toxicology, Center of Surgical and Molecular Tumor pathology, National Institute of Oncology, Budapest, Hungary
| | - Guido Kroemer
- INSERM, U848, Institut Gustave Roussy, Villejuif, France
| | - Rejean Lapointe
- Research Center, University Hospital, Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de, Montréal, Montréal, Québec, Canada
| | - Hyam I Levitsky
- School of Medicine, Oncology Center, Johns Hopkins University, Baltimore, MD, USA
| | - Michael T Lotze
- Departments of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Surgery University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Cristina Maccalli
- Department of Molecular Oncology, Foundation San Raffaele Scientific Institute, Milan, Italy
| | - Michele Maio
- Medical Oncology and Immunotherapy, Department of Oncology, University, Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | | | | | - Giuseppe Masucci
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Ignacio Melero
- Department of Immunology, CIMA, CUN and Medical School University of Navarra, Pamplona, Spain
| | - Cornelius Melief
- Deptartment of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, the Netherlands
| | - William J Murphy
- University of California-Davis Medical Center, Sacramento, CA, USA
| | - Brad Nelson
- Deeley Research Centre, BC Cancer Agency, Victoria, BC, Canada
| | - Andrea Nicolini
- Department of Internal Medicine, University of Pisa, Santa Chiara Hospital, Pisa, Italy
| | - Michael I Nishimura
- Oncology Institute, Loyola University Medical Center, Cardinal Bernardin Cancer Center, Maywood, IL, USA
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Tumor Immunology and Immunotherapy Program, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Pamela S Ohashi
- Ontario Cancer Institute/University Health Network, Toronto, ON, Canada
| | | | - Lloyd J Old
- Ludwig Institute for Cancer Research, New York, NY, USA
| | - Christian Ottensmeier
- Experimental Cancer Medicine Centre, University of Southampton Faculty of Medicine, Southampton, UK
| | - Michael Papamichail
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | - Giorgio Parmiani
- Unit of Immuno-Biotherapy of Melanoma and Solid Tumors, San Raffaele Scientific Institute, Milan, Italy
| | - Graham Pawelec
- Center for Medical Research, University of Tuebingen, Tuebingen, Germany
| | | | - Shukui Qin
- Chinese PLA Cancer Center, Nanjing, China
| | - Robert Rees
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Antoni Ribas
- Department of Medicine, Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - Ruggero Ridolfi
- Immunoterapia e Terapia Cellulare Somatica, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.), Meldola (FC), Italy
| | - Gerd Ritter
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Ludwig Institute for Cancer Research, New York, NY, USA
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, IRCCS Foundation, Istituto Nazionale Tumori, Milan, Italy
| | - Pedro J Romero
- Division of Clinical Onco-Immunology, Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland
| | - Mohamed L Salem
- Immunology and Biotechnology Unit, Department of Zoology, Faculty of Science, Tanta University, Egypt
| | - Rik J Scheper
- Dept. of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | - Hiroshi Shiku
- Department of Cancer Vaccine, Mie University Graduate School of Medicine, Mie, Japan
- Department of Immuno-gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | | | - Wenru Song
- Millennium: The Takeda Oncology Company, Cambridge, MA, USA
| | - Per Thor Straten
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Herlev Hospital, Herlev, Denmark
| | - Hideaki Tahara
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Zhigang Tian
- Institute of Immunology, School of Life Sciences, University of Science & Technology of China, Hefei, China
- Institute of Immunopharmacology & Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Sjoerd H van Der Burg
- Experimental Cancer Immunology and Therapy, Department of Clinical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, MD, USA
- Center for Human Immunology (CHI), NIH, Bethesda, MD, USA
| | - Marij JP Welters
- Experimental Cancer Immunology and Therapy, Department of Clinical Oncology (K1-P), Leiden University Medical Center, Leiden, The Netherlands
| | - Hauke Winter
- Department of Surgery, Klinikum Grosshadern, Ludwig Maximilians University, Munich, Germany
| | | | - Jedd D Wolchok
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Weihua Xiao
- Institute of Immunology, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Laurence Zitvogel
- Institut Gustave Roussy, Center of Clinical Investigations CICBT507, Villejuif, France
| | - Heinz Zwierzina
- Department Haematology and Oncology Innsbruck Medical University, Innsbruck, Austria
| | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, MD, USA
- Center for Human Immunology (CHI), NIH, Bethesda, MD, USA
| | | | - Jon M Wigginton
- Discovery Medicine-Oncology, Bristol-Myers Squibb Company, Princeton, New Jersey, USA
| | - Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
29
|
Hoos A, Britten CM, Huber C, O'Donnell-Tormey J. A methodological framework to enhance the clinical success of cancer immunotherapy. Nat Biotechnol 2011; 29:867-70. [PMID: 21997622 DOI: 10.1038/nbt.2000] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
30
|
Speetjens FM, Zeestraten ECM, Kuppen PJK, Melief CJM, van der Burg SH. Colorectal cancer vaccines in clinical trials. Expert Rev Vaccines 2011; 10:899-921. [PMID: 21692708 DOI: 10.1586/erv.11.63] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This article elucidates current strategies of active immunotherapy for colorectal cancer patients with a focus on T-cell mediated immunotherapy. Poor prognosis of especially stage III and IV colorectal cancer patients emphasizes the need for advanced therapeutic intervention. Here, we refer to clinical trials using either tumor cell-derived vaccines or tumor antigen vaccines with a special interest on safety, induced immune responses, clinical benefit and efforts to improve the clinical impact of these vaccines in the context of colorectal cancer treatment.
Collapse
Affiliation(s)
- Frank M Speetjens
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
31
|
Davis MM, Altman JD, Newell EW. Interrogating the repertoire: broadening the scope of peptide-MHC multimer analysis. Nat Rev Immunol 2011; 11:551-8. [PMID: 21760610 PMCID: PMC3699324 DOI: 10.1038/nri3020] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Labelling antigen-specific T cells with peptide-MHC multimers has provided an invaluable way to monitor T cell-mediated immune responses. A number of recent developments in this technology have made these multimers much easier to make and use in large numbers. Furthermore, enrichment techniques have provided a greatly increased sensitivity that allows the analysis of the naive T cell repertoire directly. Thus, we can expect a flood of new information to emerge in the coming years.
Collapse
Affiliation(s)
- Mark M Davis
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA.
| | | | | |
Collapse
|
32
|
Attig S, Price L, Janetzki S, Kalos M, Pride M, McNeil L, Clay T, Yuan J, Odunsi K, Hoos A, Romero P, Britten CM. A critical assessment for the value of markers to gate-out undesired events in HLA-peptide multimer staining protocols. J Transl Med 2011; 9:108. [PMID: 21745365 PMCID: PMC3148571 DOI: 10.1186/1479-5876-9-108] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 07/11/2011] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The introduction of antibody markers to identify undesired cell populations in flow-cytometry based assays, so called DUMP channel markers, has become a practice in an increasing number of labs performing HLA-peptide multimer assays. However, the impact of the introduction of a DUMP channel in multimer assays has so far not been systematically investigated across a broad variety of protocols. METHODS The Cancer Research Institute's Cancer Immunotherapy Consortium (CRI-CIC) conducted a multimer proficiency panel with a specific focus on the impact of DUMP channel use. The panel design allowed individual laboratories to use their own protocol for thawing, staining, gating, and data analysis. Each experiment was performed twice and in parallel, with and without the application of a dump channel strategy. RESULTS The introduction of a DUMP channel is an effective measure to reduce the amount of non-specific MULTIMER binding to T cells. Beneficial effects for the use of a DUMP channel were observed across a wide range of individual laboratories and for all tested donor-antigen combinations. In 48% of experiments we observed a reduction of the background MULTIMER-binding. In this subgroup of experiments the median background reduction observed after introduction of a DUMP channel was 0.053%. CONCLUSIONS We conclude that appropriate use of a DUMP channel can significantly reduce background staining across a large fraction of protocols and improve the ability to accurately detect and quantify the frequency of antigen-specific T cells by multimer reagents. Thus, use of a DUMP channel may become crucial for detecting low frequency antigen-specific immune responses. Further recommendations on assay performance and data presentation guidelines for publication of MULTIMER experimental data are provided.
Collapse
Affiliation(s)
- Sebastian Attig
- Division of Translational and Experimental Oncology, Department of Internal Medicine III, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Leah Price
- Department of Biostatistics, New York University, New York, NY USA
| | | | - Michael Kalos
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Abramson Family Cancer Research Institute, Philadelphia, PA USA
| | - Michael Pride
- Vaccine Research East and Early Development, Pfizer Inc. Pearl River, NY USA
| | - Lisa McNeil
- Vaccine Research East and Early Development, Pfizer Inc. Pearl River, NY USA
| | - Tim Clay
- Surgery and Immunology, Duke University Medical Center, Durham, NC, USA
| | - Jianda Yuan
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, New York, NY USA
| | - Kunle Odunsi
- Departments of Gynecologic Oncology and Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Axel Hoos
- Bristol-Myers Squibb, Wallingford, CT USA
| | - Pedro Romero
- Translational Tumor Immunology Group, Ludwig Center for Cancer Research of the University of Lausanne, Switzerland
| | - Cedrik M Britten
- Division of Translational and Experimental Oncology, Department of Internal Medicine III, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research & Development, BioNTech AG, Mainz, Germany
| |
Collapse
|
33
|
Butterfield LH, Palucka AK, Britten CM, Dhodapkar MV, Håkansson L, Janetzki S, Kawakami Y, Kleen TO, Lee PP, Maccalli C, Maecker HT, Maino VC, Maio M, Malyguine A, Masucci G, Pawelec G, Potter DM, Rivoltini L, Salazar LG, Schendel DJ, Slingluff CL, Song W, Stroncek DF, Tahara H, Thurin M, Trinchieri G, van Der Burg SH, Whiteside TL, Wigginton JM, Marincola F, Khleif S, Fox BA, Disis ML. Recommendations from the iSBTc-SITC/FDA/NCI Workshop on Immunotherapy Biomarkers. Clin Cancer Res 2011; 17:3064-76. [PMID: 21558394 PMCID: PMC3096674 DOI: 10.1158/1078-0432.ccr-10-2234] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To facilitate development of innovative immunotherapy approaches, especially for treatment concepts exploiting the potential benefits of personalized therapy, there is a need to develop and validate tools to identify patients who can benefit from immunotherapy. Despite substantial effort, we do not yet know which parameters of antitumor immunity to measure and which assays are optimal for those measurements. EXPERIMENTAL DESIGN The iSBTc-SITC (International Society for Biological Therapy of Cancer-Society for Immunotherapy of Cancer), FDA (Food and Drug Administration), and NCI (National Cancer Institute) partnered to address these issues for immunotherapy of cancer. Here, we review the major challenges, give examples of approaches and solutions, and present our recommendations. RESULTS AND CONCLUSIONS Although specific immune parameters and assays are not yet validated, we recommend following standardized (accurate, precise, and reproducible) protocols and use of functional assays for the primary immunologic readouts of a trial; consideration of central laboratories for immune monitoring of large, multi-institutional trials; and standardized testing of several phenotypic and functional potential potency assays specific to any cellular product. When reporting results, the full QA (quality assessment)/QC (quality control) should be conducted and selected examples of truly representative raw data and assay performance characteristics should be included. Finally, to promote broader analysis of multiple aspects of immunity, and gather data on variability, we recommend that in addition to cells and serum, RNA and DNA samples be banked (under standardized conditions) for later testing. We also recommend that sufficient blood be drawn to allow for planned testing of the primary hypothesis being addressed in the trial, and that additional baseline and posttreatment blood is banked for testing novel hypotheses (or generating new hypotheses) that arise in the field.
Collapse
Affiliation(s)
- Lisa H. Butterfield
- Departments of Medicine, Surgery and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - A. Karolina Palucka
- Baylor Institute for Immunology Research, Dallas, TX, USA
- Department of Gene and Cell Medicine, Immunology Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Cedrik M. Britten
- Department of Internal Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Clinical Development, BioNTech AG, Mainz, Germany
| | - Madhav V. Dhodapkar
- Department of Hematology & Immunobiology, Yale University, New Haven, CT, USA
| | | | | | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | | | - Peter P. Lee
- Dept. of Medicine (Hematology), Stanford University School of Medicine, Stanford, CA, USA
| | - Cristina Maccalli
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors, Division of Molecular Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Holden T. Maecker
- Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University Medical School, Stanford, CA, USA
| | | | - Michele Maio
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy and Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico, Aviano, Italy
| | - Anatoli Malyguine
- Applied and Developmental Research Directorate, SAIC-Frederick, Inc. Frederick, MD, USA
| | - Giuseppe Masucci
- Affiliation Dept of Oncology-Pathology, Karolinska Institutet/and University Hospital Coordinator of the NCEV (Nordic network Centrum of Excellence for antitumour Vaccination), Stockholm, Sweden
| | - Graham Pawelec
- Second Department of Internal Medicine, University of Tuebingen Medical School, Tuebingen, Germany
| | - Douglas M. Potter
- Biostatistics Department, Graduate School of Public Health, University of Pittsburgh and Biostatistics Facility, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan Italy
| | - Lupe G. Salazar
- Tumor Vaccine Group, Division of Oncology, University of Washington, Seattle, WA, USA
| | - Dolores J. Schendel
- Institute of Molecular Immunology and Immune Monitoring Group, Helmholth Zentrum München, Germany
| | | | | | - David F. Stroncek
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Hideaki Tahara
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Magdalena Thurin
- Cancer Diagnosis Program, Division of Cancer Treatment and Disagnosis, National Cancer Institute, NIH, Rockville, MD, USA
| | | | - Sjoerd H. van Der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Theresa L. Whiteside
- Immunologic Monitoring and Cellular Products Laboratory, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Jon M. Wigginton
- Discovery Medicine-Clinical Oncology, Bristol-Myers Squibb, Inc., Princeton, NJ, USA
| | - Francesco Marincola
- Dept. of Translational Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Samir Khleif
- Cancer Vaccine Section, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bernard A. Fox
- Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Mary L. Disis
- Tumor Vaccine Group, Division of Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
34
|
Olson WC, Smolkin ME, Farris EM, Fink RJ, Czarkowski AR, Fink JH, Chianese-Bullock KA, Slingluff CL. Shipping blood to a central laboratory in multicenter clinical trials: effect of ambient temperature on specimen temperature, and effects of temperature on mononuclear cell yield, viability and immunologic function. J Transl Med 2011; 9:26. [PMID: 21385453 PMCID: PMC3063218 DOI: 10.1186/1479-5876-9-26] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 03/08/2011] [Indexed: 01/26/2023] Open
Abstract
Background Clinical trials of immunologic therapies provide opportunities to study the cellular and molecular effects of those therapies and may permit identification of biomarkers of response. When the trials are performed at multiple centers, transport and storage of clinical specimens become important variables that may affect lymphocyte viability and function in blood and tissue specimens. The effect of temperature during storage and shipment of peripheral blood on subsequent processing, recovery, and function of lymphocytes is understudied and represents the focus of this study. Methods Peripheral blood samples (n = 285) from patients enrolled in 2 clinical trials of a melanoma vaccine were shipped from clinical centers 250 or 1100 miles to a central laboratory at the sponsoring institution. The yield of peripheral blood mononuclear cells (PBMC) collected before and after cryostorage was correlated with temperatures encountered during shipment. Also, to simulate shipping of whole blood, heparinized blood from healthy donors was collected and stored at 15°C, 22°C, 30°C, or 40°C, for varied intervals before isolation of PBMC. Specimen integrity was assessed by measures of yield, recovery, viability, and function of isolated lymphocytes. Several packaging systems were also evaluated during simulated shipping for the ability to maintain the internal temperature in adverse temperatures over time. Results Blood specimen containers experienced temperatures during shipment ranging from -1 to 35°C. Exposure to temperatures above room temperature (22°C) resulted in greater yields of PBMC. Reduced cell recovery following cryo-preservation as well as decreased viability and immune function were observed in specimens exposed to 15°C or 40°C for greater than 8 hours when compared to storage at 22°C. There was a trend toward improved preservation of blood specimen integrity stored at 30°C prior to processing for all time points tested. Internal temperatures of blood shipping containers were maintained longer in an acceptable range when warm packs were included. Conclusions Blood packages shipped overnight by commercial carrier may encounter extreme seasonal temperatures. Therefore, considerations in the design of shipping containers should include protecting against extreme ambient temperature deviations and maintaining specimen temperature above 22°C or preferably near 30°C.
Collapse
Affiliation(s)
- Walter C Olson
- Human Immune Therapy Center, University of Virginia, Charlottesville, VA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Gilewski T. Targeting the Immune System as a Therapeutic Strategy for Patients with Breast Cancer. CURRENT BREAST CANCER REPORTS 2010. [DOI: 10.1007/s12609-010-0029-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
Chan C, Lin L, Frelinger J, Hérbert V, Gagnon D, Landry C, Sékaly RP, Enzor J, Staats J, Weinhold KJ, Jaimes M, West M. Optimization of a highly standardized carboxyfluorescein succinimidyl ester flow cytometry panel and gating strategy design using discriminative information measure evaluation. Cytometry A 2010; 77:1126-36. [PMID: 21053294 PMCID: PMC3042236 DOI: 10.1002/cyto.a.20987] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The design of a panel to identify target cell subsets in flow cytometry can be difficult when specific markers unique to each cell subset do not exist, and a combination of parameters must be used to identify target cells of interest and exclude irrelevant events. Thus, the ability to objectively measure the contribution of a parameter or group of parameters toward target cell identification independent of any gating strategy could be very helpful for both panel design and gating strategy design. In this article, we propose a discriminative information measure evaluation (DIME) based on statistical mixture modeling; DIME is a numerical measure of the contribution of different parameters towards discriminating a target cell subset from all the others derived from the fitted posterior distribution of a Gaussian mixture model. Informally, DIME measures the "usefulness" of each parameter for identifying a target cell subset. We show how DIME provides an objective basis for inclusion or exclusion of specific parameters in a panel, and how ranked sets of such parameters can be used to optimize gating strategies. An illustrative example of the application of DIME to streamline the gating strategy for a highly standardized carboxyfluorescein succinimidyl ester (CFSE) assay is described.
Collapse
Affiliation(s)
- Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Britten CM, Janetzki S, van der Burg SH, Huber C, Kalos M, Levitsky HI, Maecker HT, Melief CJM, O'Donnell-Tormey J, Odunsi K, Old LJ, Pawelec G, Roep BO, Romero P, Hoos A, Davis MM. Minimal information about T cell assays: the process of reaching the community of T cell immunologists in cancer and beyond. Cancer Immunol Immunother 2010; 60:15-22. [PMID: 21080166 PMCID: PMC3029829 DOI: 10.1007/s00262-010-0940-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 10/23/2010] [Indexed: 11/26/2022]
Abstract
Many assays to evaluate the nature, breadth, and quality of antigen-specific T cell responses are currently applied in human medicine. In most cases, assay-related protocols are developed on an individual laboratory basis, resulting in a large number of different protocols being applied worldwide. Together with the inherent complexity of cellular assays, this leads to unnecessary limitations in the ability to compare results generated across institutions. Over the past few years a number of critical assay parameters have been identified which influence test performance irrespective of protocol, material, and reagents used. Describing these critical factors as an integral part of any published report will both facilitate the comparison of data generated across institutions and lead to improvements in the assays themselves. To this end, the Minimal Information About T Cell Assays (MIATA) project was initiated. The objective of MIATA is to achieve a broad consensus on which T cell assay parameters should be reported in scientific publications and to propose a mechanism for reporting these in a systematic manner. To add maximum value for the scientific community, a step-wise, open, and field-spanning approach has been taken to achieve technical precision, user-friendliness, adequate incorporation of concerns, and high acceptance among peers. Here, we describe the past, present, and future perspectives of the MIATA project. We suggest that the approach taken can be generically applied to projects in which a broad consensus has to be reached among scientists working in fragmented fields, such as immunology. An additional objective of this undertaking is to engage the broader scientific community to comment on MIATA and to become an active participant in the project.
Collapse
Affiliation(s)
- C M Britten
- III. Medical Department, Johannes Gutenberg-University, Mainz, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
BACKGROUND Harnessing the immune response in treating breast cancer would potentially offer a less toxic, more targeted approach to eradicating residual disease. Breast cancer vaccines are being developed to effectively train cytotoxic T cells to recognize and kill transformed cells while sparing normal ones. However, achieving this goal has been problematic due to the ability of established cancers to suppress and evade the immune response. METHODS A review of the literature on vaccines and breast cancer treatment was conducted, specifically addressing strategies currently available, as well as appropriate settings, paradigms for vaccine development and response monitoring, and challenges with immunosuppression. RESULTS Multiple issues need to be addressed in order to optimize the benefits offered by breast cancer vaccines. Primary issues include the following: (1) cancer vaccines will likely work better in a minimal residual disease state, (2) clinical trial design for immunotherapy should incorporate recommendations from expert groups such as the Cancer Vaccine Working Group and use standardized immune response measurements, (3) the presently available cancer vaccine approaches, including dendritic cell-based, tumor-associated antigen peptide-based, and whole cell-based, have various pros and cons, (4) to date, no one approach has been shown to be superior to another, and (5) vaccines will need to be combined with immunoregulatory agents to overcome tumor-related immunosuppression. CONCLUSIONS Combining a properly optimized cancer vaccine with novel immunomodulating agents that overcome tumor-related immunosuppression in a well-designed clinical trial offers the best hope for developing an effective breast cancer vaccine strategy.
Collapse
Affiliation(s)
- Hatem Soliman
- Department of Women's Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
39
|
Hoos A, Eggermont AMM, Janetzki S, Hodi FS, Ibrahim R, Anderson A, Humphrey R, Blumenstein B, Old L, Wolchok J. Improved endpoints for cancer immunotherapy trials. J Natl Cancer Inst 2010; 102:1388-97. [PMID: 20826737 PMCID: PMC2943524 DOI: 10.1093/jnci/djq310] [Citation(s) in RCA: 401] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Unlike chemotherapy, which acts directly on the tumor, cancer immunotherapies exert their effects on the immune system and demonstrate new kinetics that involve building a cellular immune response, followed by changes in tumor burden or patient survival. Thus, adequate design and evaluation of some immunotherapy clinical trials require a new development paradigm that includes reconsideration of established endpoints. Between 2004 and 2009, several initiatives facilitated by the Cancer Immunotherapy Consortium of the Cancer Research Institute and partner organizations systematically evaluated an immunotherapy-focused clinical development paradigm and created the principles for redefining trial endpoints. On this basis, a body of clinical and laboratory data was generated that supports three novel endpoint recommendations. First, cellular immune response assays generate highly variable results. Assay harmonization in multicenter trials may minimize variability and help to establish cellular immune response as a reproducible biomarker, thus allowing investigation of its relationship with clinical outcomes. Second, immunotherapy may induce novel patterns of antitumor response not captured by Response Evaluation Criteria in Solid Tumors or World Health Organization criteria. New immune-related response criteria were defined to more comprehensively capture all response patterns. Third, delayed separation of Kaplan–Meier curves in randomized immunotherapy trials can affect results. Altered statistical models describing hazard ratios as a function of time and recognizing differences before and after separation of curves may allow improved planning of phase III trials. These recommendations may improve our tools for cancer immunotherapy trials and may offer a more realistic and useful model for clinical investigation.
Collapse
Affiliation(s)
- Axel Hoos
- Cancer Immunotherapy Consortium of the Cancer Research Institute, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Following the evidence that T-cell responses are crucial in the control of HIV-1 infection, vaccines targeting T-cell responses were tested in recent clinical trials. However, these vaccines showed a lack of efficacy. This review attempts to define the qualitative and quantitative features that are desirable for T-cell-induced responses by vaccines. We also describe strategies that could lead to achievement of this goal. RECENT FINDINGS Using the yellow fever vaccine as a benchmark of an efficient vaccine, recent studies identified factors of immune protection and more importantly innate immune pathways needed for the establishment of long-term protective adaptive immunity. SUMMARY To prevent or control HIV-1 infection, a vaccine must induce efficient and persistent antigen-specific T cells endowed with mucosal homing capacity. Such cells should have the capability to counteract HIV-1 diversity and its rapid spread from the initial site of infection. To achieve this goal, the activation of a diversified innate immune response is critical. New systems biology approaches will provide more precise correlates of immune protection that will pave the way for new approaches in T-cell-based vaccines.
Collapse
Affiliation(s)
- Hélène Perrin
- Vaccine and Gene Therapy Institute - Florida, Port Saint Lucie, Florida, USA
| | | | | | | |
Collapse
|
41
|
Jaimes MC, Maecker HT, Yan M, Maino VC, Hanley MB, Greer A, Darden JM, D'Souza MP. Quality assurance of intracellular cytokine staining assays: analysis of multiple rounds of proficiency testing. J Immunol Methods 2010; 363:143-57. [PMID: 20727897 DOI: 10.1016/j.jim.2010.08.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 07/13/2010] [Accepted: 08/11/2010] [Indexed: 12/01/2022]
Abstract
When evaluating candidate prophylactic HIV and cancer vaccines, intracellular cytokine staining (ICS) assays that measure the frequency and magnitude of antigen-specific T-cell subsets are one tool to monitor immunogen performance and make product advancement decisions. To assess the inter-laboratory assay variation among multiple laboratories testing vaccine candidates, the NIH/NIAID/DAIDS in collaboration with BD Biosciences implemented an ICS Quality Assurance Program (QAP). Seven rounds of testing have been conducted in which 16 laboratories worldwide participated. In each round, IFN-γ, IL-2 and/or TNF-α responses in CD4+ and CD8+ T-cells to CEF or CMV pp65 peptide mixes were tested using cryopreserved peripheral blood mononuclear cells (PBMC) from CMV seropositive donors. We found that for responses measured above 0.2%, inter-laboratory %CVs were, on average, 35%. No differences in inter-laboratory variation were observed if a 4-color antibody cocktail or a 7-color combination was used. Moreover, the data allowed identification of important sources of variability for flow cytometry-based assays, including: number of collected events, gating strategy and instrument setup and performance. As a consequence, in this multi-site study we were able to define pass and fail criteria for ICS assays, which will be adopted in the subsequent rounds of testing and could be easily extrapolated to QAP for other flow cytometry-based assays.
Collapse
Affiliation(s)
- Maria C Jaimes
- BD Biosciences, 2350 Qume Drive, San Jose, CA 95131, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Madore DV, Meade BD, Rubin F, Deal C, Lynn F, the Meeting Contributors. Utilization of serologic assays to support efficacy of vaccines in nonclinical and clinical trials: meeting at the crossroads. Vaccine 2010; 28:4539-47. [PMID: 20470795 PMCID: PMC2886496 DOI: 10.1016/j.vaccine.2010.04.094] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 04/22/2010] [Accepted: 04/27/2010] [Indexed: 10/19/2022]
Abstract
In May 2009 the National Institute of Allergy and Infectious Diseases hosted a workshop on serologic assays that support vaccine efficacy evaluations. The meeting promoted exchange of ideas among investigators from varying disciplines who are working on anti-infectious agent vaccines at different stages of development. The presentations and discussions at the workshop illustrated the challenges common across various pathogens with recurring themes: (1) A thorough understanding of the science regarding the pathogen and the host response to disease and immunization is fundamental to assay selection. (2) The intended use of the immunoassay data must be clearly defined to ensure appropriate specificity, accuracy, and precision; a laboratory must also commit resources to assure data quality and reliability. (3) During vaccine development, an immunoassay may evolve with respect to quality, purpose, and degree of standardization, and, in some cases, must be changed or replaced as data are accumulated. (4) Collaboration on standardized reagents and methods, harmonization efforts, and multidisciplinary teams facilitate consistent generation of quality data. This report provides guidance for effective development and utilization of immunoassays based on the lessons learned from currently licensed vaccines. Investigators are encouraged to create additional opportunities for scientific exchange, noting that the discussed themes are relevant for immunoassays used for other purposes such as therapeutics and diagnostics.
Collapse
Affiliation(s)
| | | | - Fran Rubin
- National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892
| | - Carolyn Deal
- National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892
| | - Freyja Lynn
- National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892
| | | |
Collapse
Collaborators
Margaret C Bash, Drusilla L Burns, Jan Callahan, James B Dale, Morven S Edwards, Karen L Elkins, Carl E Frasch, Steven W Hildreth, Katrin Jansen, Robert C Kohberger, Michael Kalos, Karen C Meysick, ChrisAnna M Mink, Moon Nahm, Brian D Plikaytis, David S Stephens, Kanta Subbarao, Christopher Taylor, Edward E Walsh,
Collapse
|
43
|
Frelinger J, Ottinger J, Gouttefangeas C, Chan C. Modeling flow cytometry data for cancer vaccine immune monitoring. Cancer Immunol Immunother 2010; 59:1435-41. [PMID: 20563720 PMCID: PMC2892609 DOI: 10.1007/s00262-010-0883-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 06/09/2010] [Indexed: 11/30/2022]
Abstract
Flow cytometry (FCM) is widely used in cancer research for diagnosis, detection of minimal residual disease, as well as immune monitoring and profiling following immunotherapy. In all these applications, the challenge is to detect extremely rare cell subsets while avoiding spurious positive events. To achieve this objective, it helps to be able to analyze FCM data using multiple markers simultaneously, since the additional information provided often helps to minimize the number of false positive and false negative events, hence increasing both sensitivity and specificity. However, with manual gating, at most two markers can be examined in a single dot plot, and a sequential strategy is often used. As the sequential strategy discards events that fall outside preceding gates at each stage, the effectiveness of the strategy is difficult to evaluate without laborious and painstaking back-gating. Model-based analysis is a promising computational technique that works using information from all marker dimensions simultaneously, and offers an alternative approach to flow analysis that can usefully complement manual gating in the design of optimal gating strategies. Results from model-based analysis will be illustrated with examples from FCM assays commonly used in cancer immunotherapy laboratories.
Collapse
Affiliation(s)
- Jacob Frelinger
- Institute for Genome Sciences and Policy, Duke University, Durham, NC 27708, USA
| | | | | | | |
Collapse
|
44
|
Parida SK, Magalhaes I, Dubois P, Janetzki S. Training in immunology of relevance to global health issues in resource poor settings. Eur J Immunol 2010; 40:1228-31. [DOI: 10.1002/eji.201090022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
| | | | - Patrice Dubois
- Immunovacc Consulting, Brussels, Belgium and University of Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
45
|
Speiser DE, Romero P. Molecularly defined vaccines for cancer immunotherapy, and protective T cell immunity. Semin Immunol 2010; 22:144-54. [PMID: 20413326 DOI: 10.1016/j.smim.2010.03.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 03/15/2010] [Indexed: 11/28/2022]
Abstract
Malignant cells are frequently recognized and destroyed by T cells, hence the development of T cell vaccines against established tumors. The challenge is to induce protective type 1 immune responses, with efficient Th1 and CTL activation, and long-term immunological memory. These goals are similar as in many infectious diseases, where successful immune protection is ideally induced with live vaccines. However, large-scale development of live vaccines is prevented by their very limited availability and vector immunogenicity. Synthetic vaccines have multiple advantages. Each of their components (antigens, adjuvants, delivery systems) contributes specifically to induction and maintenance of T cell responses. Here we summarize current experience with vaccines based on proteins and peptide antigens, and discuss approaches for the molecular characterization of clonotypic T cell responses. With carefully designed step-by-step modifications of innovative vaccine formulations, T cell vaccination can be optimized towards the goal of inducing therapeutic immune responses in humans.
Collapse
Affiliation(s)
- Daniel E Speiser
- Clinical Investigation Center, Ludwig Institute for Cancer Research Ltd., Lausanne branch, University of Lausanne, Switzerland.
| | | |
Collapse
|
46
|
Janetzki S, Price L, Britten CM, van der Burg SH, Caterini J, Currier JR, Ferrari G, Gouttefangeas C, Hayes P, Kaempgen E, Lennerz V, Nihlmark K, Souza V, Hoos A. Performance of serum-supplemented and serum-free media in IFNgamma Elispot Assays for human T cells. Cancer Immunol Immunother 2010; 59:609-18. [PMID: 19894047 PMCID: PMC2813531 DOI: 10.1007/s00262-009-0788-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 10/16/2009] [Indexed: 12/26/2022]
Abstract
The choice of serum for supplementation of media for T cell assays and in particular, Elispot has been a major challenge for assay performance, standardization, optimization, and reproducibility. The Assay Working Group of the Cancer Vaccine Consortium (CVC-CRI) has recently identified the choice of serum to be the leading cause for variability and suboptimal performance in large international Elispot proficiency panels. Therefore, a serum task force was initiated to compare the performance of commercially available serum-free media to laboratories' own medium/serum combinations. The objective of this project was to investigate whether a serum-free medium exists that performs as well as lab-own serum/media combinations with regard to antigen-specific responses and background reactivity in Elispot. In this way, a straightforward solution could be provided to address the serum challenge. Eleven laboratories tested peripheral blood mononuclear cells (PBMC) from four donors for their reactivity against two peptide pools, following their own Standard Operating Procedure (SOP). Each laboratory performed five simultaneous experiments with the same SOP, the only difference between the experiments was the medium used. The five media were lab-own serum-supplemented medium, AIM-V, CTL, Optmizer, and X-Vivo. The serum task force results demonstrate compellingly that serum-free media perform as well as qualified medium/serum combinations, independent of the applied SOP. Recovery and viability of cells are largely unaffected by serum-free conditions even after overnight resting. Furthermore, one serum-free medium was identified that appears to enhance antigen-specific IFNgamma-secretion.
Collapse
Affiliation(s)
- Sylvia Janetzki
- Cancer Vaccine Consortium of the Cancer Research Institute, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kalos M. An integrative paradigm to impart quality to correlative science. J Transl Med 2010; 8:26. [PMID: 20233418 PMCID: PMC2848636 DOI: 10.1186/1479-5876-8-26] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Accepted: 03/16/2010] [Indexed: 12/27/2022] Open
Abstract
Correlative studies are a primary mechanism through which insights can be obtained about the bioactivity and potential efficacy of candidate therapeutics evaluated in early-stage clinical trials. Accordingly, well designed and performed early-stage correlative studies have the potential to strongly influence further clinical development of candidate therapeutic agents, and correlative data obtained from early stage trials has the potential to provide important guidance on the design and ultimate successful evaluation of products in later stage trials, particularly in the context of emerging clinical trial paradigms such as adaptive trial design. Historically the majority of early stage trials have not generated meaningful correlative data sets that could guide further clinical development of the products under evaluation. In this review article we will discuss some of the potential limitations with the historical approach to performing correlative studies that might explain at least in part the to-date overall failure of such studies to adequately support clinical trial development, and present emerging thought and approaches related to comprehensiveness and quality that hold the promise to support the development of correlative plans which will provide meaningful correlative data that can effectively guide and support the clinical development path for candidate therapeutic agents.
Collapse
Affiliation(s)
- Michael Kalos
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Abramson Family Cancer Research Institute, Philadelphia, 19104-4283, USA.
| |
Collapse
|
48
|
Casalegno-Garduño R, Schmitt A, Yao J, Wang X, Xu X, Freund M, Schmitt M. Multimer technologies for detection and adoptive transfer of antigen-specific T cells. Cancer Immunol Immunother 2010; 59:195-202. [PMID: 19847424 PMCID: PMC11030699 DOI: 10.1007/s00262-009-0778-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Accepted: 09/24/2009] [Indexed: 10/20/2022]
Abstract
Identification and purification of antigen-specific T cells without altering their functional status are of high scientific and clinical interest. Staining with major histocompatibility complex (MHC)-peptide multimers constitutes a very powerful method to study antigen-specific T-cell subpopulations, allowing their direct visualization and quantification. MHC-peptide multimers, such as dimers, tetramers, pentamers, streptamers, dextramers and octamers have been used to evaluate the frequency of CD8(+) T cells, specific for tumor/leukemia-associated antigens as well as for viral antigens, e.g., CMVpp65 and EBV-EBNA. Moreover, MHC-peptide multimers have been used for rapid and efficient ex vivo isolation and expansion of T cells. A recent development in the field of MHC-peptide multimers led to the purification of CD8(+) T cells specific for leukemia antigens. This might help to select leukemia-specific donor lymphocyte infusions (DLIs), thus allowing dissection of the noxious graft-versus-host disease (GvHD) from beneficial anti-viral and even anti-leukemic effects. This review covers different types of MHC-peptide multimers and their applications, as well as the impact that multimers might have on further development of DLIs.
Collapse
Affiliation(s)
- Rosaely Casalegno-Garduño
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
| | - Anita Schmitt
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
| | - Junxia Yao
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xinchao Wang
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
- Department of Oncology and Hematology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xun Xu
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
- Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Mathias Freund
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
| | - Michael Schmitt
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
| |
Collapse
|
49
|
Mander A, Gouttefangeas C, Ottensmeier C, Welters MJP, Low L, van der Burg SH, Britten CM. Serum is not required for ex vivo IFN-gamma ELISPOT: a collaborative study of different protocols from the European CIMT Immunoguiding Program. Cancer Immunol Immunother 2010; 59:619-27. [PMID: 20052465 PMCID: PMC2813523 DOI: 10.1007/s00262-009-0814-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Accepted: 12/21/2009] [Indexed: 02/04/2023]
Abstract
The Cancer Immunotherapy Immunoguiding Program has conducted an IFN-γ ELISPOT proficiency panel to examine the influence of serum supplementation of test media on assay performance. Sixteen European laboratories analyzed the same PBMC samples using different locally established protocols. Participants generated two simultaneous data sets—one using medium supplemented with serum and one without serum. Performances of the two test conditions were compared by quantifying: (1) the number of viable cells, (2) background spot formation induced in the medium only control and (3) the ability to detect antigen-specific T cell responses. The study demonstrated that the number of viable cells recovered and the overall background spot production were not significantly different between the two conditions. Furthermore, overall laboratory performance was equivalent for the two test conditions; 11 out of 16 laboratories reported equal or greater detection rates using serum-free medium, while 5 laboratories reported decreased detections rates under serum-free conditions. These results show that good performance of the IFN-γ ELISPOT assay can be achieved under serum-free conditions. Optimization of the protocol for serum-free conditions should result in excellent detection rates and eliminate the requirement of serum batch and stability testing, allowing further harmonization of the assay.
Collapse
Affiliation(s)
- A. Mander
- Cancer Sciences Division, Southampton University Hospitals, Southampton, UK
| | - C. Gouttefangeas
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | - C. Ottensmeier
- Cancer Sciences Division, Southampton University Hospitals, Southampton, UK
| | - M. J. P. Welters
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - L. Low
- Cancer Sciences Division, Southampton University Hospitals, Southampton, UK
| | - S. H. van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - C. M. Britten
- Division of Experimental and Translational Oncology, Department of Internal Medicine III, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
50
|
Axelsson-Robertson R, Weichold F, Sizemore D, Wulf M, Skeiky YAW, Sadoff J, Maeurer MJ. Extensive major histocompatibility complex class I binding promiscuity for Mycobacterium tuberculosis TB10.4 peptides and immune dominance of human leucocyte antigen (HLA)-B*0702 and HLA-B*0801 alleles in TB10.4 CD8 T-cell responses. Immunology 2009; 129:496-505. [PMID: 20002212 DOI: 10.1111/j.1365-2567.2009.03201.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The molecular definition of major histocompatibility complex (MHC) class I-presented CD8(+) T-cell epitopes from clinically relevant Mycobacterium tuberculosis (Mtb) target proteins will aid in the rational design of T-cell-based diagnostics of tuberculosis (TB) and the measurement of TB vaccine-take. We used an epitope discovery system, based on recombinant MHC class I molecules that cover the most frequent Caucasian alleles [human leucocyte antigen (HLA)-A*0101, A*0201, A*0301, A*1101, A*2402, B*0702, B*0801 and B*1501], to identify MHC class I-binding peptides from overlapping 9-mer peptides representing the Mtb protein TB10.4. A total of 33 MHC class I-binding epitopes were identified, spread across the entire amino acid sequence, with some clustering at the N- and C-termini of the protein. Binding of individual peptides or closely related peptide species to different MHC class I alleles was frequently observed. For instance, the common motif of xIMYNYPAMx bound to six of eight alleles. Affinity (50% effective dose) and off-rate (half life) analysis of candidate Mtb peptides will help to define the conditions for CD8(+) T-cell interaction with their nominal MHC class I-peptide ligands. Subsequent construction of tetramers allowed us to confirm the recognition of some of the epitopes by CD8(+) T cells from patients with active pulmonary TB. HLA-B alleles served as the dominant MHC class I restricting molecules for anti-Mtb TB10.4-specific CD8(+) T-cell responses measured in CD8(+) T cells from patients with pulmonary TB.
Collapse
|