1
|
Baraniuk JN, Eaton-Fitch N, Marshall-Gradisnik S. Meta-analysis of natural killer cell cytotoxicity in myalgic encephalomyelitis/chronic fatigue syndrome. Front Immunol 2024; 15:1440643. [PMID: 39483457 PMCID: PMC11524851 DOI: 10.3389/fimmu.2024.1440643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/09/2024] [Indexed: 11/03/2024] Open
Abstract
Reduced natural killer (NK) cell cytotoxicity is the most consistent immune finding in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Meta-analysis of the published literature determined the effect size of the decrement in ME/CFS. Databases were screened for papers comparing NK cell cytotoxicity in ME/CFS and healthy controls. A total of 28 papers and 55 effector:target cell ratio (E:T) data points were collected. Cytotoxicity in ME/CFS was significantly reduced to about half of healthy control levels, with an overall Hedges' g of 0.96 (0.75-1.18). Heterogeneity was high but was explained by the range of E:T ratios, different methods, and potential outliers. The outcomes confirm reproducible NK cell dysfunction in ME/CFS and will guide studies using the NK cell model system for pathomechanistic investigations. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024542140.
Collapse
Affiliation(s)
- James N. Baraniuk
- Department of Medicine, Georgetown University, Washington, DC, United States
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
2
|
Eugene-Norbert M, Cuffel A, Riou G, Jean L, Blondel C, Dehayes J, Bisson A, Giverne C, Brotin E, Denoyelle C, Poulain L, Boyer O, Martinet J, Latouche JB. Development of optimized cytotoxicity assays for assessing the antitumor potential of CAR-T cells. J Immunol Methods 2024; 525:113603. [PMID: 38147898 DOI: 10.1016/j.jim.2023.113603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/01/2023] [Accepted: 12/09/2023] [Indexed: 12/28/2023]
Abstract
CAR-T cells are T cells expressing a chimeric antigen receptor (CAR) rendering them capable of killing tumor cells after recognition of a target antigen. CD19 CAR-T cells have revolutionized the treatment of hematological malignancies. Their function is typically assessed by cytotoxicity assays using human allogeneic cell lines expressing the target antigen CD19 such as Nalm-6. However, an alloreactive reaction is observed with these cells, leading to a CD19-independent killing. To address this issue, we developed a fluorescence microscopy-based potency assay using murine target cells to provide an optimized cytotoxicity assay with enhanced specificity towards CD19. Murine NIH/3T3 (3T3) fibroblast-derived cell line and EL4 T-cell lymphoma-derived cell line were used as targets (no xenoreactivity was observed after coculture with human T cells). 3T3 and EL4 cells were engineered to express eGFP (enhanced Green Fluorescent Protein) and CD19 or CD22 using retroviral vectors. CD19 CAR-T cells and non-transduced (NT) control T cells were produced from several donors. After 4 h or 24 h, alloreactive cytotoxicity against CD19+ Nalm-6-GFP cells and CD19- Jurkat-GFP cells was observed with NT or CAR-T cells. In the same conditions, CAR-T but not NT cells specifically killed CD19+ but not CD19- 3T3-GFP or EL4-GFP cells. Both microscope- and flow cytometry-based assays revealed as sensitive as impedance-based assay. Using flow cytometry, we could further determine that CAR-T cells had mostly a stem cell-like memory phenotype after contact with EL4 target cells. Therefore, CD19+ 3T3-GFP or EL4-GFP cells and fluorescence microscopy- or flow cytometry-based assays provide convenient, sensitive and specific tools to evaluate CAR-T cell function with no alloreactivity.
Collapse
Affiliation(s)
- Misa Eugene-Norbert
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Rouen F-76000, France
| | - Alexis Cuffel
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Rouen F-76000, France; Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Department of Immunology and Biotherapy, Rouen University Hospital, Rouen F-76000, France
| | - Gaetan Riou
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Rouen F-76000, France
| | - Laetitia Jean
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Rouen F-76000, France
| | - Clara Blondel
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Rouen F-76000, France
| | - Justine Dehayes
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Department of Immunology and Biotherapy, Rouen University Hospital, Rouen F-76000, France
| | - Aurélie Bisson
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Department of Immunology and Biotherapy, Rouen University Hospital, Rouen F-76000, France
| | - Camille Giverne
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Department of Immunology and Biotherapy, Rouen University Hospital, Rouen F-76000, France
| | - Emilie Brotin
- Université de Caen Normandie, Unité de Service PLATON, ImpedanCELL Core Facility, Caen F-14000, France
| | - Christophe Denoyelle
- Université de Caen Normandie, Unité de Service PLATON, ImpedanCELL Core Facility, Caen F-14000, France; Université de Caen Normandie, Inserm, ANTICIPE UMR (1086), Structure Fédérative 4207 Normandie Oncologie, Normandie Univ, Caen F-14000, France; Comprehensive Cancer Center F. Baclesse, Unicancer, Caen F-14000, France
| | - Laurent Poulain
- Université de Caen Normandie, Inserm, ANTICIPE UMR (1086), Structure Fédérative 4207 Normandie Oncologie, Normandie Univ, Caen F-14000, France; Comprehensive Cancer Center F. Baclesse, Unicancer, Caen F-14000, France
| | - Olivier Boyer
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Rouen F-76000, France; Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Department of Immunology and Biotherapy, Rouen University Hospital, Rouen F-76000, France
| | - Jérémie Martinet
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Rouen F-76000, France; Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Department of Immunology and Biotherapy, Rouen University Hospital, Rouen F-76000, France.
| | - Jean-Baptiste Latouche
- Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Rouen F-76000, France; Univ Rouen Normandie, Inserm UMR1234, FOCIS Center of Excellence PAn'THER, Department of Immunology and Biotherapy, Rouen University Hospital, Rouen F-76000, France
| |
Collapse
|
3
|
Hirosaki H, Maeda Y, Takeyoshi M. Establishment of Cell-Based Assay System for Evaluating Cytotoxic Activity Modulated by the Blockade of PD-1 and PD-L1 Interactions with a Therapeutic Antibody. Immunol Invest 2023; 52:332-342. [PMID: 36731129 DOI: 10.1080/08820139.2023.2174442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Therapeutic antibodies targeting the PD-1/PD-L1 immune checkpoint are widely used in cancer therapy and are under active further development. Historically, the antitumor activity of PD-1/PD-L1 immune checkpoint inhibitors has been evaluated using in vivo and ex vivo test methods; however, a simple in vitro assay method to evaluate antitumor activity accurately is needed for the efficient development of new therapeutic agents. In the present study, we attempted to establish a simple cell-based assay system to evaluate the modulating effect of PD-1/PD-L1 immune checkpoint inhibitors on cytotoxic activity. METHODS We established a new natural killer (NK) cell line stably transfected with the PD-1 and IL-2 genes and a new NK-sensitive target cell line stably transfected with the PD-L1 gene. Then, the assay system was established by co-cultivation of the established cell lines and measurement of the cytotoxic activities using the europium release assay. To confirm the performance of the established assay system, model therapeutic antibodies to block the PD-1/PD-L1 signal, nivolumab and atezolizumab were added to the co-culture system and the modulating effect on the cytotoxic activities were evaluated. RESULTS Nivolumab and atezolizumab clearly showed a modulating effect on cytotoxic activity in a dose-dependent manner in our assay system, whereas a human IgG isotype control antibody did not show any modulating effect on the assay system. CONCLUSION The newly established cell-based assay system can quantitatively evaluate the modulating effect of PD-1/PD-L1 immune checkpoint inhibitors by measuring cytotoxic activity, playing an important role in antitumor effects as innate immunity.
Collapse
Affiliation(s)
- Haruka Hirosaki
- Chemicals Assessment and Research Center, Chemicals Evaluation and Research Institute, Japan
| | - Yosuke Maeda
- Chemicals Assessment and Research Center, Chemicals Evaluation and Research Institute, Japan
| | - Masahiro Takeyoshi
- Chemicals Assessment and Research Center, Chemicals Evaluation and Research Institute, Japan
| |
Collapse
|
4
|
Halliez C, Ibrahim H, Otonkoski T, Mallone R. In vitro beta-cell killing models using immune cells and human pluripotent stem cell-derived islets: Challenges and opportunities. Front Endocrinol (Lausanne) 2023; 13:1076683. [PMID: 36726462 PMCID: PMC9885197 DOI: 10.3389/fendo.2022.1076683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Type 1 diabetes (T1D) is a disease of both autoimmunity and β-cells. The β-cells play an active role in their own demise by mounting defense mechanisms that are insufficient at best, and that can become even deleterious in the long term. This complex crosstalk is important to understanding the physiological defense mechanisms at play in healthy conditions, their alterations in the T1D setting, and therapeutic agents that may boost such mechanisms. Robust protocols to develop stem-cell-derived islets (SC-islets) from human pluripotent stem cells (hPSCs), and islet-reactive cytotoxic CD8+ T-cells from peripheral blood mononuclear cells offer unprecedented opportunities to study this crosstalk. Challenges to develop in vitro β-cell killing models include the cluster morphology of SC-islets, the relatively weak cytotoxicity of most autoimmune T-cells and the variable behavior of in vitro expanded CD8+ T-cells. These challenges may however be highly rewarding in light of the opportunities offered by such models. Herein, we discuss these opportunities including: the β-cell/immune crosstalk in an islet microenvironment; the features that make β-cells more sensitive to autoimmunity; therapeutic agents that may modulate β-cell vulnerability; and the possibility to perform analyses in an autologous setting, i.e., by generating T-cell effectors and SC-islets from the same donor.
Collapse
Affiliation(s)
- Clémentine Halliez
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| | - Hazem Ibrahim
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
- Department of Pediatrics, Helsinki University Hospital, Helsinki, Finland
| | - Roberto Mallone
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| |
Collapse
|
5
|
Dias J, Cadiñanos-Garai A, Roddie C. Release Assays and Potency Assays for CAR T-Cell Interventions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:117-137. [PMID: 37258787 DOI: 10.1007/978-3-031-30040-0_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Chimeric antigen receptor (CAR) T-cells are considered "living drugs" and offer a compelling alternative to conventional anticancer therapies. Briefly, T-cells are redirected, using gene engineering technology, toward a specific cancer cell surface target antigen via a synthetic chimeric antigen receptor (CAR) protein. CARs have a modular design comprising four main structures: an antigen-binding domain, a hinge region, a transmembrane domain, and one or more intracellular signaling domains for T-cell activation. A major challenge in the CAR T-cell manufacturing field is balancing product quality with scalability and cost-effectiveness, especially when transitioning from an academic clinical trial into a marketed product, to be implemented across many collection, manufacturing, and treatment sites. Achieving product consistency while circumnavigating the intrinsic variability associated with autologous products is an additional barrier. To overcome these limitations, a robust understanding of the product and its biological actions is crucial to establish a target product profile with a defined list of critical quality attributes to be assessed for each batch prior to product certification. Additional challenges arise as the field progresses, such as new safety considerations associated with the use of allogenic T-cells and genome editing tools. In this chapter, we will discuss the release and potency assays required for CAR T-cell manufacturing, covering their relevance, current challenges, and future perspectives.
Collapse
Affiliation(s)
- Juliana Dias
- UCL Cancer Institute, University College London, London, UK.
- Royal Free Hospital London, NHS Foundation Trust, London, UK.
| | - Amaia Cadiñanos-Garai
- USC/CHLA Cell Therapy Program, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, USA
| | - Claire Roddie
- UCL Cancer Institute, University College London, London, UK
- Department of Haematology, UCL Hospital, London, UK
| |
Collapse
|
6
|
Garvin D, Stecha P, Gilden J, Wang J, Grailer J, Hartnett J, Fan F, Cong M, Cheng ZJ. Determining ADCC Activity of Antibody-Based Therapeutic Molecules using Two Bioluminescent Reporter-Based Bioassays. Curr Protoc 2021; 1:e296. [PMID: 34787960 DOI: 10.1002/cpz1.296] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibody Fc effector function is one of the main mechanisms of action (MoA) for therapeutic monoclonal antibodies. Measurement of antibody-dependent cellular cytotoxicity (ADCC) is critical for understanding the Fc effector function during monoclonal antibody development. This article covers two cell-based ADCC bioassays which can quantitatively measure the antibody potency in ADCC. Basic Protocol 1 describes the ADCC reporter bioassay using engineered ADCC effector cells which measures the FcγRIIIa-mediated luciferase reporter activation upon the binding of antibody-coated target cells. Basic Protocol 2 describes the PBMC ADCC bioassay using primary peripheral blood mononuclear cells (PBMC) as effector cells and engineered HiBiT target cells in an assay that measures the release of HiBiT from target cells upon antibody-mediated target lysis. Optimization of several key assay parameters including cell handling, effector:target (E:T) ratios, assay plate, and plate reader requirement, and how these parameters impact assay performance are discussed. © 2021 Promega Corporation. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: ADCC reporter bioassay using engineered ADCC bioassay effector cells Basic Protocol 2: PBMC ADCC bioassay using primary PBMC and engineered HiBiT target cells.
Collapse
Affiliation(s)
| | | | | | - Jun Wang
- Promega Corporation, Madison, Wisconsin
| | | | | | - Frank Fan
- Promega Corporation, Madison, Wisconsin
| | - Mei Cong
- Promega Corporation, Madison, Wisconsin
| | | |
Collapse
|
7
|
Penny SA, Abelin JG, Malaker SA, Myers PT, Saeed AZ, Steadman LG, Bai DL, Ward ST, Shabanowitz J, Hunt DF, Cobbold M. Tumor Infiltrating Lymphocytes Target HLA-I Phosphopeptides Derived From Cancer Signaling in Colorectal Cancer. Front Immunol 2021; 12:723566. [PMID: 34504498 PMCID: PMC8421858 DOI: 10.3389/fimmu.2021.723566] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/27/2021] [Indexed: 12/21/2022] Open
Abstract
There is a pressing need for novel immunotherapeutic targets in colorectal cancer (CRC). Cytotoxic T cell infiltration is well established as a key prognostic indicator in CRC, and it is known that these tumor infiltrating lymphocytes (TILs) target and kill tumor cells. However, the specific antigens that drive these CD8+ T cell responses have not been well characterized. Recently, phosphopeptides have emerged as strong candidates for tumor-specific antigens, as dysregulated signaling in cancer leads to increased and aberrant protein phosphorylation. Here, we identify 120 HLA-I phosphopeptides from primary CRC tumors, CRC liver metastases and CRC cell lines using mass spectrometry and assess the tumor-resident immunity against these posttranslationally modified tumor antigens. Several CRC tumor-specific phosphopeptides were presented by multiple patients’ tumors in our cohort (21% to 40%), and many have previously been identified on other malignancies (58% of HLA-A*02 CRC phosphopeptides). These shared antigens derived from mitogenic signaling pathways, including p53, Wnt and MAPK, and are therefore markers of malignancy. The identification of public tumor antigens will allow for the development of broadly applicable targeted therapeutics. Through analysis of TIL cytokine responses to these phosphopeptides, we have established that they are already playing a key role in tumor-resident immunity. Multifunctional CD8+ TILs from primary and metastatic tumors recognized the HLA-I phosphopeptides presented by their originating tumor. Furthermore, TILs taken from other CRC patients’ tumors targeted two of these phosphopeptides. In another cohort of CRC patients, the same HLA-I phosphopeptides induced higher peripheral T cell responses than they did in healthy donors, suggesting that these immune responses are specifically activated in CRC patients. Collectively, these results establish HLA-I phosphopeptides as targets of the tumor-resident immunity in CRC, and highlight their potential as candidates for future immunotherapeutic strategies.
Collapse
Affiliation(s)
- Sarah A Penny
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Jennifer G Abelin
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Stacy A Malaker
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Paisley T Myers
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Abu Z Saeed
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Lora G Steadman
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Dina L Bai
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Stephen T Ward
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom.,Department of Colorectal Surgery, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Donald F Hunt
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States.,Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Mark Cobbold
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom.,Center for Cancer Immunology, Massachusetts General Hospital, Charlestown, MA, United States
| |
Collapse
|
8
|
Comparative analysis of assays to measure CAR T-cell-mediated cytotoxicity. Nat Protoc 2021; 16:1331-1342. [PMID: 33589826 DOI: 10.1038/s41596-020-00467-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023]
Abstract
The antitumor efficacy of genetically engineered 'living drugs', including chimeric antigen receptor and T-cell receptor T cells, is influenced by their activation, proliferation, inhibition, and exhaustion. A sensitive and reproducible cytotoxicity assay that collectively reflects these functions is an essential requirement for translation of these cellular therapeutic agents. Here, we compare various in vitro cytotoxicity assays (including chromium release, bioluminescence, impedance, and flow cytometry) with respect to their experimental setup, appropriate uses, advantages, and disadvantages, and measures to overcome their limitations. We also highlight the US Food and Drug Administration (FDA) directives for a potency assay for release of clinical cell therapy products. In addition, we discuss advanced assays of repeated antigen exposure and simultaneous testing of combinations of immune effector cells, immunomodulatory antibodies, and targets with variable antigen expression. This review article should help to equip investigators with the necessary knowledge to select appropriate cytotoxicity assays to test the efficacy of immunotherapeutic agents alone or in combination.
Collapse
|
9
|
Lee HY, Register A, Shim J, Contreras E, Wu Q, Jiang G. Characterization of a single reporter-gene potency assay for T-cell-dependent bispecific molecules. MAbs 2019; 11:1245-1253. [PMID: 31348721 PMCID: PMC6748617 DOI: 10.1080/19420862.2019.1640548] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 01/07/2023] Open
Abstract
T-cell-dependent bispecific antibodies (TDBs) are promising cancer immunotherapies that recruit patients' T cells to kill cancer cells. There are many TDBs in clinical trials, demonstrating their widely recognized therapeutic potential. However, their complex, multi-step mechanism of action (MoA), which includes bispecific antigen binding, T-cell activation, and target-cell killing, presents unique challenges for biological characterization and potency assay selection. Here, we describe the development of a single reporter-gene potency assay for a TDB (TDB1) that is MoA reflective and sensitive to binding of both antigens. Our reporter-gene assay measures T-cell activation using Jurkat cells engineered to express luciferase under the control of an NFkB response element. The potencies of select samples were measured both by this assay and by a flow-cytometry-based cell-killing assay using human lymphocytes as effector cells. Correlating the two sets of potency results clearly establishes our reporter-gene assay as MoA reflective. Furthermore, correlating potencies for the same panel of samples against binding data measured by binding assays for each individual arm demonstrates that the reporter-gene potency assay reflects dual-antigen binding and can detect changes in affinity for either arm. This work demonstrates that one reporter-gene assay can be used to measure the potency of TDB1 while capturing key aspects of its MoA, thus serving as a useful case study of selection and justification of reporter-gene potency assays for TDBs. Furthermore, our strategy of correlating reporter-gene potency, target-cell killing, and antigen binding for each individual arm serves as a useful example of a thorough, holistic approach to biological characterization for TDBs that can be applied to other bispecific molecules.
Collapse
Affiliation(s)
- Ho Young Lee
- Biological Technologies, Department of Analytical Development and Quality Control, Genentech, A member of the Roche group, South San Francisco, CA, USA
| | - Ames Register
- Biological Technologies, Department of Analytical Development and Quality Control, Genentech, A member of the Roche group, South San Francisco, CA, USA
| | - Jeongsup Shim
- Biological Technologies, Department of Analytical Development and Quality Control, Genentech, A member of the Roche group, South San Francisco, CA, USA
| | - Edward Contreras
- Biological Technologies, Department of Analytical Development and Quality Control, Genentech, A member of the Roche group, South San Francisco, CA, USA
| | - Qiang Wu
- Biological Technologies, Department of Analytical Development and Quality Control, Genentech, A member of the Roche group, South San Francisco, CA, USA
| | - Guoying Jiang
- Biological Technologies, Department of Analytical Development and Quality Control, Genentech, A member of the Roche group, South San Francisco, CA, USA
| |
Collapse
|
10
|
Tagod MSO, Mizuta S, Sakai Y, Iwasaki M, Shiraishi K, Senju H, Mukae H, Morita CT, Tanaka Y. Determination of human γδ T cell-mediated cytotoxicity using a non-radioactive assay system. J Immunol Methods 2019; 466:32-40. [PMID: 30654042 DOI: 10.1016/j.jim.2019.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/29/2018] [Accepted: 01/11/2019] [Indexed: 12/20/2022]
Abstract
The adoptive transfer of immune effector cells, such as CD8+ killer αβ T cells, γδ T cells, NK (natural killer) cells, and genetically-modified T cells, has been receiving increasing attention. It is essential to determine cellular cytotoxicity so as to monitor the function and quality of ex vivo-expanded immune effector cells before infusion. The most common method is the [51Cr]-sodium chromate release assay. It is, however, preferable to avoid the use of radioactive materials in clinical laboratories. In order to establish a non-radioactive alternative to the standard radioactive assay, we previously synthesized a chelate-forming prodrug (BM-HT) and demonstrated that a combination of BM-HT and europium (Eu3+) was useful to determine NK cell-mediated cytotoxicity. In the present study, we examined whether or not this improved assay system could be used to determine the cellular cytotoxicity exhibited by Vγ2Vδ2+ γδ T cells. In addition, we compared Eu3+ and terbium (Tb3+) in the measurement of cellular cytotoxicity. Our assay system using BM-HT could be used successfully for the analysis of both γδ T cell receptor (TCR)- and CD16-mediated cytotoxicity. When the intensity of fluorescence was compared between Eu3+ and Tb3+, Tb3+ chelate was more sensitive than Eu3+ chelate, suggesting that the detection system using Tb3+ is superior to Eu3+ when tumor cells are not efficiently labeled with BM-HT. The method established herein is expected to promote the development of novel adoptive cell therapies for cancer.
Collapse
Affiliation(s)
- Mohammed S O Tagod
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Infectious Diseases, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Satoshi Mizuta
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yuki Sakai
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Masashi Iwasaki
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshidakonoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Kengo Shiraishi
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Hiroaki Senju
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Respiratory Medicine, Nagasaki University School of Medicine, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University School of Medicine, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Craig T Morita
- Department of Internal Medicine and the Interdisciplinary Graduate Program in Immunology, Iowa City Veterans Affairs Health Care System, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
| | - Yoshimasa Tanaka
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Infectious Diseases, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshidakonoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501, Japan.
| |
Collapse
|
11
|
Hayek S, Bekaddour N, Besson L, Alves de Sousa R, Pietrancosta N, Viel S, Smith N, Jacob Y, Nisole S, Mandal R, Wishart DS, Walzer T, Herbeuval JP, Vidalain PO. Identification of Primary Natural Killer Cell Modulators by Chemical Library Screening with a Luciferase-Based Functional Assay. SLAS DISCOVERY 2018; 24:25-37. [PMID: 30184441 DOI: 10.1177/2472555218797078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Natural killer (NK) cells are essential players of the innate immune response that secrete cytolytic factors and cytokines such as IFN-γ when contacting virus-infected or tumor cells. They represent prime targets in immunotherapy as defects in NK cell functions are hallmarks of many pathological conditions, such as cancer and chronic infections. The functional screening of chemical libraries or biologics would greatly help identify new modulators of NK cell activity, but commonly used methods such as flow cytometry are not easily scalable to high-throughput settings. Here we describe an efficient assay to measure the natural cytotoxicity of primary NK cells where the bioluminescent enzyme NanoLuc is constitutively expressed in the cytoplasm of target cells and is released in co-culture supernatants when lysis occurs. We fully characterized this assay using either purified NK cells or total peripheral blood mononuclear cells (PBMCs), including some patient samples, as effector cells. A pilot screen was also performed on a library of 782 metabolites, xenobiotics, and common drugs, which identified dextrometorphan and diphenhydramine as novel NK cell inhibitors. Finally, this assay was further improved by developing a dual-reporter cell line to simultaneously measure NK cell cytotoxicity and IFN-γ secretion in a single well, extending the potential of this system.
Collapse
Affiliation(s)
- Simon Hayek
- 1 Chimie & Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS, UMR8601, Paris, France
| | - Nassima Bekaddour
- 1 Chimie & Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS, UMR8601, Paris, France
| | - Laurie Besson
- 2 Centre International de Recherche en Infectiologie, CIRI, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, University of Lyon, Lyon, France.,3 Laboratoire d'Immunologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre-Bénite, France
| | - Rodolphe Alves de Sousa
- 1 Chimie & Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS, UMR8601, Paris, France
| | - Nicolas Pietrancosta
- 1 Chimie & Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS, UMR8601, Paris, France
| | - Sébastien Viel
- 2 Centre International de Recherche en Infectiologie, CIRI, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, University of Lyon, Lyon, France.,3 Laboratoire d'Immunologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre-Bénite, France
| | - Nikaia Smith
- 1 Chimie & Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS, UMR8601, Paris, France
| | - Yves Jacob
- 4 CNRS, UMR3569, Unité de Génétique Moléculaire des Virus à ARN, Institut Pasteur, Université Paris Diderot, Paris, France
| | - Sébastien Nisole
- 5 Institut de Recherche en Infectiologie de Montpellier, CNRS, UMR9004, Université de Montpellier, Montpellier, France
| | - Rupasri Mandal
- 6 Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, Alberta, Canada
| | - David S Wishart
- 6 Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, Alberta, Canada
| | - Thierry Walzer
- 2 Centre International de Recherche en Infectiologie, CIRI, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Jean-Philippe Herbeuval
- 1 Chimie & Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS, UMR8601, Paris, France
| | - Pierre-Olivier Vidalain
- 1 Chimie & Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS, UMR8601, Paris, France
| |
Collapse
|
12
|
Sakai Y, Mizuta S, Kumagai A, Tagod MSO, Senju H, Nakamura T, Morita CT, Tanaka Y. Live Cell Labeling with Terpyridine Derivative Proligands to Measure Cytotoxicity Mediated by Immune Cells. ChemMedChem 2017; 12:2006-2013. [PMID: 29110403 DOI: 10.1002/cmdc.201700626] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 10/30/2017] [Indexed: 12/31/2022]
Abstract
Immunotherapy using immune checkpoint inhibitors and CAR-T cells has revolutionized treatment for patients with malignant tumors. However, measuring tumor cell cytotoxicity mediated by immune effector cells in clinical laboratories has been difficult due to the requirement for radioactive substances. In this study, a series of novel terpyridine derivative proligands were synthesized, and a non-radioactive cellular cytotoxicity assay using the newly synthesized compounds was developed for use in preclinical and clinical studies for cancer immunotherapy. Once internalized into target cells, the compounds are hydrolyzed by esterases, resulting in the intracellular accumulation of the negatively charged terpyridine derivatives. When the labeled target cells are recognized and killed by immune effector cells, the integrity of the cell membrane is disrupted, and the terpyridine derivatives are released. Upon combining the culture supernatant with europium (Eu3+ ), the cytotoxicity of immune effector cells for the target cells can be quantitatively determined by measuring the intensity of the Eu3+ /ligand-derived time-resolved fluorescence. Thus, the assay developed in this study would facilitate the development of novel cancer immunotherapies.
Collapse
Affiliation(s)
- Yuki Sakai
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Satoshi Mizuta
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Asuka Kumagai
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Mohammed S O Tagod
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Hiroaki Senju
- Second Department of Internal Medicine, Nagasaki University Hospital, Nagasaki University, 1-7-1, Sakamoto, Nagsaki, 852-8501, Japan
| | - Tatsufumi Nakamura
- Department of Social Work, Faculty of Human and Social Studies, Nagasaki International University, 2825-7 Huis Ten Bosch, Sasebo, Nagasaki, 859-3298, Japan
| | - Craig T Morita
- Department of Internal Medicine and the Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Veterans Affairs Health Care System, 601 Highway 6 West, Research, 151, Iowa City, IA, 52246, USA
| | - Yoshimasa Tanaka
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| |
Collapse
|
13
|
Kandarian F, Sunga GM, Arango-Saenz D, Rossetti M. A Flow Cytometry-Based Cytotoxicity Assay for the Assessment of Human NK Cell Activity. J Vis Exp 2017. [PMID: 28829424 DOI: 10.3791/56191] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Within the innate immune system, effector lymphocytes known as natural killer (NK) cells play an essential role in host defense against aberrant cells, specifically eliminating tumoral and virally infected cells. Approximately 30 known monogenic defects, together with a host of other pathological conditions, cause either functional or classic NK cell deficiency, manifesting in reduced or absent cytotoxic activity. Historically, cytotoxicity has been investigated with radioactive methods, which are cumbersome, expensive and potentially hazardous. This article describes a streamlined, clinically applicable flow cytometry-based method to quantify NK cell cytotoxic activity. In this assay, peripheral blood mononuclear cells (PBMCs) or purified NK cell preparations are co-incubated at different ratios with a target tumor cell line known to be sensitive to NK cell-mediated cytotoxicity (NKCC). The target cells are pre-labeled with a fluorescent dye to allow their discrimination from the effector cells (NK cells). After the incubation period, killed target cells are identified by a nucleic acid stain, which specifically permeates dead cells. This method is amenable to both diagnostic and research applications and, thanks to the multi-parameter capabilities of flow cytometry, has the added advantage of potentially enabling a deeper analysis of NK cell phenotype and function.
Collapse
Affiliation(s)
- Fadi Kandarian
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Gemalene M Sunga
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Diana Arango-Saenz
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Maura Rossetti
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles;
| |
Collapse
|
14
|
Surrogate CD16-expressing effector cell lines for determining the bioactivity of therapeutic monoclonal antibodies. J Pharm Biomed Anal 2017; 143:188-198. [PMID: 28605680 DOI: 10.1016/j.jpba.2017.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/01/2017] [Accepted: 06/03/2017] [Indexed: 12/12/2022]
Abstract
Traditional antibody dependent cellular cytotoxicity (ADCC) assays use donor derived natural killer (NK) or peripheral blood mononuclear cells, but donor genetic variability and the technically challenging nature of the assay means that alternative in vitro assay formats are required. We explored the utility of two reporter gene cell lines, the J2 and J9, as surrogate effector cells for ADCC assays. Both express the ADCC relevant Fcγ receptor CD16, crosslinking of which leads to firefly luciferase expression. For anti-CD20 rituximab and anti-HER2 trastuzumab (both IgG1 monoclonal antibodies, mAbs) a dose dependent firefly luciferase response was observed exclusively in the presence of their respective targets, representing the molecular interaction which potentiates ADCC activity. Importantly, both surrogate effector and NK cell based assays gave statistically similar values for rituximab ADCC activity. Increased engagement with target cell bound mAbs was determined to be cytotoxic for the J2 and J9 cell lines at the assay end point (at which luciferase expression is measured). However, use of the J9 cells containing the constitutively expressed renilla luciferase gene enabled data normalisation and corrected for fluctuations in both cell number and viability providing an advantage over currently available surrogate effector cell-lines. Abrogated ADCC activity with IgG4 mAbs, but enhanced activity with an IgG1 non-fucosylated mAb, was seen with the J9 cell line, as expected. Additionally, two rituximab products (biosimilars in development) with similar binding by flow cytometry, N-glycan profiles using HPLC and CD16 binding by surface plasmon resonance showed comparable ADCC activity to Mabthera. The ADCC activity of another anti-CD20 mAb, ofatumumab, reported only with primary cell based assays to date was also measured. This is the first report of a dual reporter gene based ADCC assay.
Collapse
|
15
|
Chung S, Nguyen V, Lin YL, Kamen L, Song A. Thaw-and-use target cells pre-labeled with calcein AM for antibody-dependent cell-mediated cytotoxicity assays. J Immunol Methods 2017; 447:37-46. [PMID: 28434980 DOI: 10.1016/j.jim.2017.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 04/04/2017] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
Abstract
In vitro antibody-dependent cell-mediated cytotoxicity (ADCC) assays are routinely performed to support the research and development of therapeutic antibodies. In ADCC assays, target cells bound by the antibodies are lysed by activated effector cells following interactions between the Fc region of the bound antibody and Fcγ receptors on effector cells. Target cell lysis is typically measured by quantification of released endogenous enzymes, e.g., lactate dehydrogenase, or measurement of released exogenous labels, e.g., 51Cr, europium or calcein. ADCC assays based on the detection of exogenous labels released from lysed target cells generally show higher sensitivity and require shorter incubation times. However, target cells are usually labeled immediately prior to assay, which inadvertently introduces additional assay variations due to differences in target cell conditions and labeling/handling processes. In this report, we describe the use of thaw-and-use pre-labeled target cells for ADCC assays. Thaw-and-use target cells in our experiments were pre-labeled with the fluorescent dye calcein AM, cryopreserved in single-use aliquots and used directly in assays after thawing. Upon thaw, the pre-labeled cells displayed viability and label retention comparable to freshly labeled cells, responded to ADCC mediated by both peripheral blood mononuclear cells and engineered natural killer cells, performed stably for at least 3 years and provided favorable precision and accuracy to ADCC assays. Implementation of thaw-and-use pre-labeled target cells in ADCC assays can help to alleviate both cell culture and dye labeling derived variability, increase the flexibility of assay scheduling and improve assay consistency and robustness.
Collapse
Affiliation(s)
- Shan Chung
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, United States.
| | - Van Nguyen
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, United States
| | - Yuwen Linda Lin
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, United States
| | - Lynn Kamen
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, United States
| | - An Song
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, United States
| |
Collapse
|
16
|
Luciferase mRNA Transfection of Antigen Presenting Cells Permits Sensitive Nonradioactive Measurement of Cellular and Humoral Cytotoxicity. J Immunol Res 2016; 2016:9540975. [PMID: 27057556 PMCID: PMC4736447 DOI: 10.1155/2016/9540975] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/10/2015] [Indexed: 11/25/2022] Open
Abstract
Immunotherapy is rapidly evolving as an effective treatment option for many cancers. With the emerging fields of cancer vaccines and adoptive cell transfer therapies, there is an increasing demand for high-throughput in vitro cytotoxicity assays that efficiently analyze immune effector functions. The gold standard 51Cr-release assay is very accurate but has the major disadvantage of being radioactive. We reveal the development of a versatile and nonradioactive firefly luciferase in vitro transcribed (IVT) RNA-based assay. Demonstrating high efficiency, consistency, and excellent target cell viability, our optimized luciferase IVT RNA is used to transfect dividing and nondividing primary antigen presenting cells. Together with the long-lasting expression and minimal background, the direct measurement of intracellular luciferase activity of living cells allows for the monitoring of killing kinetics and displays paramount sensitivity. The ability to cotransfect the IVT RNA of the luciferase reporter and the antigen of interest into the antigen presenting cells and its simple read-out procedure render the assay high-throughput in nature. Results generated were comparable to the 51Cr release and further confirmed the assay's ability to measure antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity. The assay's combined simplicity, practicality, and efficiency tailor it for the analysis of antigen-specific cellular and humoral effector functions during the development of novel immunotherapies.
Collapse
|
17
|
Cheng ZJ, Garvin D, Paguio A, Moravec R, Engel L, Fan F, Surowy T. Development of a robust reporter-based ADCC assay with frozen, thaw-and-use cells to measure Fc effector function of therapeutic antibodies. J Immunol Methods 2014; 414:69-81. [DOI: 10.1016/j.jim.2014.07.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 11/16/2022]
|
18
|
Wang Z, Hall MD, Rewers-Felkins KA, Quinlin IS, Wright SE. Dendritic cells enhance the activity of human MUC1-stimulated mononuclear cells against breast cancer. Oncoimmunology 2014; 2:e23335. [PMID: 23526065 PMCID: PMC3601184 DOI: 10.4161/onci.23335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Dendritic cells (DCs) are among the most potent antigen-presenting cells (APCs), stimulating peripheral blood mononuclear cells (PBMCs) to generate antigen-specific cytotoxic T lymphocytes (CTLs). The objectives of this study were to determine if interleukin (IL)-4 is beneficial or detrimental for the generation of human DCs in vitro and to understand whether DCs generated in vitro in the presence or absence of IL-4 stimulate the killing of adenocarcinoma cells by CTLs in vivo. Mucin 1 (MUC1), a glycoprotein found on the surface of adenocarcinoma cells was used to load DCs. MUC1-loaded DCs generated in the absence of IL-4 were superior to their counterparts produced with IL-4 in stimulating PBMCs to kill human breast cancer MCF-7 cells in vitro. A corollary in vivo protection experiment was performed by injecting immunodeficient NOD-SCID mice with MCF-7 cells s.c. and MUC1-loaded CTLs, PBMCs, or DCs generated in the absence of IL-4, i.p. Mice that received CTLs and MUC1-loaded DCs on days 0, 2, 4, 9, 14 and 19 were completely protected against the development of MCF-7-derived tumors, while other schedules conferred lower protection. Therefore, tumor antigen-loaded DCs enhance the efficacy of adoptive CTL transfer, and should thus be considered for combinatorial immunotherapeutic regimens.
Collapse
Affiliation(s)
- Zhenyao Wang
- Departments of Internal Medicine and Biomedical Sciences; Texas Tech University Health Sciences Center Schools of Medicine and Pharmacy; Amarillo, TX USA ; Department of Life, Earth and Environmental Sciences; West Texas A & M University; Canyon, TX USA
| | | | | | | | | |
Collapse
|
19
|
Wright SE, Rewers-Felkins KA, Chowdhury NI, Ahmed J, Srivastava SK, Lockwood-Cooke PR. Tucaresol down-modulation of MUC1-stimulated human mononuclear cells. Immunol Invest 2013; 43:160-9. [DOI: 10.3109/08820139.2013.860161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
20
|
Abonnenc M, Borgatti M, Fabbri E, Gavioli R, Fortini C, Destro F, Altomare L, Manaresi N, Medoro G, Romani A, Tartagni M, Lo Monaco E, Giacomini P, Guerrieri R, Gambari R. Lysis-on-Chip of Single Target Cells following Forced Interaction with CTLs or NK Cells on a Dielectrophoresis-Based Array. THE JOURNAL OF IMMUNOLOGY 2013; 191:3545-52. [DOI: 10.4049/jimmunol.1300890] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
21
|
Kellner C, Derer S, Valerius T, Peipp M. Boosting ADCC and CDC activity by Fc engineering and evaluation of antibody effector functions. Methods 2013; 65:105-13. [PMID: 23851282 DOI: 10.1016/j.ymeth.2013.06.036] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 06/26/2013] [Accepted: 06/27/2013] [Indexed: 01/18/2023] Open
Abstract
In recent years, therapy with monoclonal antibodies has become standard of care in various clinical applications. Despite obvious clinical activity, not all patients respond and benefit from this generally well tolerated treatment option. Therefore, rational optimization of antibody therapy represents a major area of interest in translational research. Animal models and clinical data suggested important roles of Fc-mediated effector mechanisms such as antibody dependent cell-mediated cytotoxicity (ADCC) or complement dependent cytotoxicity (CDC) in antibody therapy. These novel insights into the mechanisms of action mediated by monoclonal antibodies inspired the development of different engineering approaches to enhance/optimize antibodies' effector functions. Fc-engineering approaches by altering the Fc-bound glycosylation profile or by exchanging amino acids in the protein backbone have been intensively studied. Here, advanced and emerging technologies in Fc-engineering resulting in altered ADCC and CDC activity are summarized and experimental strategies to evaluate antibodies' effector functions are discussed.
Collapse
Affiliation(s)
- Christian Kellner
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Stefanie Derer
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University Kiel, Germany.
| |
Collapse
|
22
|
Hidaka M, Nakamura M, Ohmichi Y, Itoh J, Fukuzawa K, Masuko T, Yagi H. Involvement of intestinal intraepithelial lymphocytes in turnover of intestinal epithelial cells: Morphological and functional alterations due to daily administration of FK506. Cell Immunol 2012; 279:124-33. [DOI: 10.1016/j.cellimm.2012.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 08/29/2012] [Accepted: 10/02/2012] [Indexed: 12/21/2022]
|
23
|
Paillard C, Halle P, Tchirkov A, Confland C, Veyrat-Masson R, Quainon F, Perreira B, Rochette E, Pfeiffer M, Lang P, Deméocq F, Kanold J. NK cytotoxicity and alloreactivity against neuroblastoma cell lines in vitro: Comparison of Europium fluorometry assay and quantification by RT-PCR. J Immunol Methods 2012; 380:56-64. [DOI: 10.1016/j.jim.2012.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 02/13/2012] [Accepted: 03/29/2012] [Indexed: 10/28/2022]
|
24
|
Abstract
The objective was to evaluate the toxicity and feasibility of intraperitoneal infusion of tumor-specific cytotoxic T lymphocytes (CTL) as therapy for recurrent ovarian cancer, and to determine if repetitive cycles of CTL generation and infusion measurably increases the host's ovarian cancer immune response. In this study, 7 subjects with recurrent ovarian cancer confined to the peritoneal cavity underwent up to 4 cycles, each cycle beginning with a leukapheresis for collection of precursor lymphocytes, which were stimulated in vitro with mucin 1, a tumor-specific antigen found commonly in ovarian cancer cells. The resulting new CTL for each cycle were reintroduced into the host by intraperitoneal infusion. Immunologic parameters (killer cells, cytokine production, memory T lymphocytes, and natural killer cells) were studied. Toxicity, CA-125, and survival data were also evaluated. The tumor marker CA-125 was nonstatistically significantly reduced after the first month of immunotherapy. However, after that it rose. Killer cells, cytokine production, and memory T lymphocytes increased after the first cycle of stimulation, but plateaued or reduced thereafter. The percent of natural killer cells inversely correlated with other immune parameters. Median survival was 11.5 months. One subject is free of disease since December, 2000. Multiple cycles, beyond 1 cycle, of T-cell stimulation followed by adoptive T-cell infusion, may not enhance the in vivo immune response.
Collapse
|
25
|
Valiathan R, Lewis JE, Melillo AB, Leonard S, Ali KH, Asthana D. Evaluation of a Flow Cytometry-Based Assay for Natural Killer Cell Activity in Clinical Settings. Scand J Immunol 2012; 75:455-62. [DOI: 10.1111/j.1365-3083.2011.02667.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
26
|
Pascal V, Laffleur B, Cogné M. Class-specific effector functions of therapeutic antibodies. Methods Mol Biol 2012; 901:295-317. [PMID: 22723109 DOI: 10.1007/978-1-61779-931-0_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Physiology usually combines polyclonal antibodies of multiple classes in a single humoral response. Beyond their common ability to bind antigens, these various classes of human immunoglobulins carry specific functions which can each serve specific goals. In many cases, the function of a monoclonal therapeutic antibody may thus be modulated according to the class of its constant domains. Depending on the immunoglobulin class, different functional assays will be used in order to evaluate the functional activity of a monoclonal antibody.
Collapse
Affiliation(s)
- Virginie Pascal
- CNRS UMR6101, Contrôle des Réponses Immunes B et Lymphoproliférations, Université de Limoges, Limoges, France
| | | | | |
Collapse
|
27
|
Zaritskaya L, Shurin MR, Sayers TJ, Malyguine AM. New flow cytometric assays for monitoring cell-mediated cytotoxicity. Expert Rev Vaccines 2010; 9:601-16. [PMID: 20518716 PMCID: PMC2911950 DOI: 10.1586/erv.10.49] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The exact immunologic responses after vaccination that result in effective antitumor immunity have not yet been fully elucidated and the data from ex vivo T-cell assays have not yet defined adequate surrogate markers for clinical efficacy. A more detailed knowledge of the specific immune responses that correlate with positive clinical outcomes should help to develop better or novel strategies to effectively activate the immune system against tumors. Furthermore, clinically relevant material is often limited and, thus, precludes the ability to perform multiple assays. The two main assays currently used to monitor lymphocyte-mediated cytoxicity in cancer patients are the (51)Cr-release assay and IFN-gamma ELISpot assay. The former has a number of disadvantages, including low sensitivity, poor labeling and high spontaneous release of isotope from some tumor target cells. Additional problems with the (51)Cr-release assay include difficulty in obtaining autologous tumor targets, and biohazard and disposal problems for the isotope. The ELISpot assays do not directly measure cytotoxic activity and are, therefore, a surrogate marker of cyotoxic capacity of effector T cells. Furthermore, they do not assess cytotoxicity mediated by the production of the TNF family of death ligands by the cytotoxic cells. Therefore, assays that allow for the simultaneous measurement of several parameters may be more advantageous for clinical monitoring. In this respect, multifactor flow cytometry-based assays are a valid addition to the currently available immunologic monitoring assays. Use of these assays will enable detection and enumeration of tumor-specific cytotoxic T lymphocytes and their specific effector functions and any correlations with clinical responses. Comprehensive, multifactor analysis of effector cell responses after vaccination may help to detect factors that determine the success or failure of a vaccine and its immunological potency.
Collapse
Affiliation(s)
- Liubov Zaritskaya
- Applied and Developmental Research Support Program, SAIC-Frederick,
Inc., National Cancer Institute at Frederick, Frederick, MD, USA
| | - Michael R Shurin
- Departments of Pathology and Immunology, University of Pittsburgh
Medical Center, Pittsburgh, PA, USA
| | - Thomas J Sayers
- Cancer and Inflammation Program, SAIC-Frederick, Inc., National
Cancer Institute at Frederick, Frederick, MD, USA
| | - Anatoli M Malyguine
- Applied and Developmental Research Support Program, SAIC-Frederick,
Inc., National Cancer Institute at Frederick, Frederick, MD, USA
| |
Collapse
|
28
|
Measurement of cytotoxic T lymphocyte activity of human cytomegalovirus seropositive individuals by a highly sensitive coupled luminescent method. Med Microbiol Immunol 2009; 198:257-62. [PMID: 19813020 DOI: 10.1007/s00430-009-0126-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Indexed: 10/20/2022]
Abstract
A coupled luminescent method (CLM) based on glyceraldehyde-3-phosphate dehydrogenase released from injured target cells was used to evaluate the cytotoxicity of antigen-specific HLA class I-restricted CTLs. In contrast to established methods, CLM does not require the pretreatment of target cells with radioactive or toxic labeling substances. CTLs from healthy HLA-A2 positive donors were stimulated by autologous dendritic cells (DCs) pulsed with HLA-A2 restricted HCMV-pp65 nonamer peptides. HLA-A2 positive T2 cells or autologous monocytes pulsed with HCMV-pp65 nonamer peptide served as target cells. Lysis was detected only in HCMV-pp65-pulsed target cells incubated with CTLs from seropositive donors stimulated by HCMV-pp65-pulsed DCs. After 3 days, stimulation 38% of T2 cells and 17% of monocytes were lysed at an effector to target ratio of 8:1. In conclusion, CLM represents a highly sensitive, fast, material-saving and non-toxic/non-radioactive method for the measurement of antigen-specific CTL cytotoxic activity.
Collapse
|
29
|
Kim GG, Donnenberg VS, Donnenberg AD, Gooding W, Whiteside TL. A novel multiparametric flow cytometry-based cytotoxicity assay simultaneously immunophenotypes effector cells: comparisons to a 4 h 51Cr-release assay. J Immunol Methods 2007; 325:51-66. [PMID: 17617419 PMCID: PMC2040258 DOI: 10.1016/j.jim.2007.05.013] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 05/09/2007] [Accepted: 05/30/2007] [Indexed: 11/16/2022]
Abstract
Natural killer (NK) cell-or T cell-mediated cytotoxicity traditionally is measured in 4-16 h (51)Cr-release assays (CRA). A new four-color flow cytometry-based cytotoxicity assay (FCC) was developed to simultaneously measure NK cell cytotoxicity and NK cell phenotype (CD3(-)CD16(+)CD56(+)). Target cells, K562 or Daudi, were labeled with Cell Tracker Orange (CTO) prior to the addition of effector cells. Following co-incubation, 7 amino-actinomycin D (7-AAD) was added to measure death of target cells. The phenotype of effectors, viability of targets, the formation of tumor-effector cell conjugates and absolute numbers of all cells were measured based on light scatter (FSC/SSC), double discrimination of the fluorescence peak integral and height, and fluorescence intensity. Kinetic studies (0.5 and 1 to 4 h) at different effector to target (E:T) cell ratios (50, 25, 12, and 6) confirmed that the 3 h incubation was optimal. The FCC assay is more sensitive than the CRA, has a coefficient of variation (CV) 8-13% and reliably measures NK cell-or lymphokine-activated killer (LAK) cell-mediated killing of target cells in normal controls and subjects with cancer. The FCC assay can be used to study a range of phenotypic attributes, in addition to lytic activity of various subsets of effector cells, without radioactive tracers and thus, it is relatively inexpensive. The FCC assay has a potential for providing information about molecular interactions underlying target cell lysis and thus becoming a major tool for studies of disease pathogenesis as well as development of novel immune therapies.
Collapse
MESH Headings
- Antigens, CD/analysis
- Antigens, Differentiation, T-Lymphocyte/analysis
- CD3 Complex/analysis
- CD56 Antigen/analysis
- Cell Line, Tumor
- Chromium Radioisotopes/metabolism
- Cytotoxicity Tests, Immunologic/methods
- Dactinomycin/analogs & derivatives
- Dactinomycin/chemistry
- Flow Cytometry/methods
- Fluorescent Dyes/chemistry
- GPI-Linked Proteins
- Granzymes/analysis
- Humans
- Immunophenotyping/methods
- K562 Cells
- Killer Cells, Lymphokine-Activated/chemistry
- Killer Cells, Lymphokine-Activated/immunology
- Killer Cells, Lymphokine-Activated/metabolism
- Killer Cells, Natural/chemistry
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Kinetics
- Lectins, C-Type
- Leukocytes, Mononuclear/immunology
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/pathology
- Receptors, IgG/analysis
- Reproducibility of Results
- T-Lymphocyte Subsets/chemistry
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes, Cytotoxic/chemistry
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Time Factors
Collapse
Affiliation(s)
- G G Kim
- University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
30
|
Nagorsen D, Scheibenbogen C, Thiel E, Keilholz U. Immunological monitoring of cancer vaccine therapy. Expert Opin Biol Ther 2005; 4:1677-84. [PMID: 15461579 DOI: 10.1517/14712598.4.10.1677] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Immunological treatment of malignant diseases in humans aiming at the induction and proliferation of antigen-specific T cells has made rapid progress in recent years. A growing number of tumour-associated antigens, potentially synergistic combinations with adjuvants, and various routes of application provide new opportunities for cancer vaccination. Therefore, a highly accurate assessment of vaccine-induced T cell responses is required. Three T cell assays (tetramers, intracellular cytokine flow cytometry and ELISPOT assay) have emerged as first-line methods for monitoring T cell induction during vaccination. These assays are relatively easy to perform, reliable, sensitive and allow an ex vivo T cell analysis at the single cell level. Although at this stage assays are not a defined surrogate marker for clinical efficacy, they already provide information concerning the immunological potency of a given vaccine. In particular, comparing immune responses under various treatment conditions will help to develop more clinically efficient tumour vaccination. Novel assays, such as CD107 staining, human leukocyte antigen/green fluorescent protein-antigen-presenting cells or microarrays, and assays determining functions, such as proliferation assays, are beginning to complement first-line monitoring assays.
Collapse
Affiliation(s)
- Dirk Nagorsen
- Charité, Campus Benjamin Franklin, Medizinische Klinik III, Hindenburgdamm 30, 12200 Berlin, Germany.
| | | | | | | |
Collapse
|
31
|
Onon TS, Kitchener HC, Duggan-Keen M, Stern PL. No alteration in NK function or zeta chain expression in NK and T cells of cervical cancer patients. Gynecol Oncol 2003; 89:120-8. [PMID: 12694665 DOI: 10.1016/s0090-8258(03)00009-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To investigate in vitro natural killer (NK) cell activity and expression of signal-transducing zeta chains in patients with cervical cancer. PATIENTS AND METHODS Experiments were performed with frozen lymphocytes from patients at all disease stages and from healthy controls. Thawed NK were activated by overnight incubation in interferon-gamma (IFN-gamma); activity against two target cell lines was assessed by 4-h (51)Cr release assay. Targets chosen were K562, an erythroleukemic cell line, and a cervical carcinoma cell line designated 808. T and NK cell zeta chain expression was measured by flow cytometry. RESULTS Patients' NK were found to be as cytotoxic as those of normal controls against cell lines K562 and 808. Patient T and NK cells did not show significant down-regulation of the zeta chain. CONCLUSIONS We have found no evidence to suggest that loss of zeta chains is a mechanism for immunocompromise in patients with cervical carcinoma. IFN-recoverable patient NK activity is not reduced compared to matched controls. This may be clinically relevant since NK are active against cells exhibiting class I human leukocyte antigen (HLA) down-regulation and many cervical cancers show loss of HLA.
Collapse
Affiliation(s)
- Toli S Onon
- Department of Immunology, Paterson Institute for Cancer Research, Christie Hospital, Manchester, United Kingdom.
| | | | | | | |
Collapse
|
32
|
Neri S, Mariani E, Meneghetti A, Cattini L, Facchini A. Calcein-acetyoxymethyl cytotoxicity assay: standardization of a method allowing additional analyses on recovered effector cells and supernatants. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2001; 8:1131-5. [PMID: 11687452 PMCID: PMC96238 DOI: 10.1128/cdli.8.6.1131-1135.2001] [Citation(s) in RCA: 234] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cytotoxicity assays provide an in vitro evaluation of the lytic activity of NK and T cells against tumors or transformed cells. However, none of these methods allow the recovery of cells or supernatants after the assay. We standardized a microcytotoxicity test using calcein-acetoxymethyl (calcein-AM) dye that requires very small quantities of cells while maintaining the same sensitivity as the traditional (51)Cr assay. The assay is applicable to resting as well as activated human effector cells and uses different targets such as human cell lines that are adherent or growing in suspension and resistant or sensitive. The most important feature of the method is the possibility of recovering cells and supernatants for additional analyses such as phenotyping and evaluation of soluble factors.
Collapse
Affiliation(s)
- S Neri
- Dipartimento di Medicina Interna e Gastroenterologia, University of Bologna, Bologna, Italy
| | | | | | | | | |
Collapse
|
33
|
Wright SE, Kilinski L, Talib S, Lowe KE, Burnside JS, Wu JY, Dolby N, Dombrowski KE, Lebkowski JS, Philip R. Cytotoxic T lymphocytes from humans with adenocarcinomas stimulated by native MUC1 mucin and a mucin peptide mutated at a glycosylation site. J Immunother 2000; 23:2-10. [PMID: 10687132 DOI: 10.1097/00002371-200001000-00002] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
MUC1 mucin peptides stimulated cytotoxic T lymphocytes (CTL) from humans with adenocarcinomas. Peripheral blood mononuclear cells, tumor-draining lymph node cells, or tumor-infiltrating lymphocytes were stimulated using mono-nuclear cells from humans with adenocarcinomas of breast or ovary, respectively, using (a) a native MUC1 mucin tandem repeat peptide of 20 amino acids (MUC1-mtr1) plus recombinant human interleukin-2 (IL-2), (b) the mutated (T3N) MUC1-mtr1 plus IL-2, or (c) immobilized anti-CD3 plus IL-2, or (d) IL-2 alone. The CTL stimulated by each of these four conditions were predominately CD4+. However, the CTL stimulated by either the native MUC1-mtr1 or (T3N) MUC1-mtr1 showed 5-10 times greater cytotoxicity of a breast cancer cell line that expresses MUC1 compared to CTL stimulated by either anti-CD3 + IL-2 or IL-2 alone. Each incubation condition generated CTL with different variable beta gene families of T-cell receptors, implying an oligoclonal expansion of a limited CTL repertoire for each. Thus, peptide-stimulated T cells showed expression of cytotoxic cells, which was not induced by nonspecific (anti-CD3 or IL-2) stimulation.
Collapse
Affiliation(s)
- S E Wright
- Department of Veterans Affairs Medical Center, Amarillo, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|