1
|
Liang W, Li K, Gao H, Li K, Zhang J, Zhang Q, Jiao X, Yang J, Wei X. Full T-cell activation and function in teleosts require collaboration of first and co-stimulatory signals. Zool Res 2024; 45:13-24. [PMID: 38114429 PMCID: PMC10839663 DOI: 10.24272/j.issn.2095-8137.2023.053] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/08/2023] [Indexed: 12/21/2023] Open
Abstract
Mammalian T-cell responses require synergism between the first signal and co-stimulatory signal. However, whether and how dual signaling regulates the T-cell response in early vertebrates remains unknown. In the present study, we discovered that the Nile tilapia ( Oreochromis niloticus) encodes key components of the LAT signalosome, namely, LAT, ITK, GRB2, VAV1, SLP-76, GADS, and PLC-γ1. These components are evolutionarily conserved, and CD3ε mAb-induced T-cell activation markedly increased their expression. Additionally, at least ITK, GRB2, and VAV1 were found to interact with LAT for signalosome formation. Downstream of the first signal, the NF-κB, MAPK/ERK, and PI3K-AKT pathways were activated upon CD3ε mAb stimulation. Furthermore, treatment of lymphocytes with CD28 mAbs triggered the AKT-mTORC1 pathway downstream of the co-stimulatory signal. Combined CD3ε and CD28 mAb stimulation enhanced ERK1/2 and S6 phosphorylation and elevated NFAT1, c-Fos, IL-2, CD122, and CD44 expression, thereby signifying T-cell activation. Moreover, rather than relying on the first or co-stimulatory signal alone, both signals were required for T-cell proliferation. Full T-cell activation was accompanied by marked apoptosis and cytotoxic responses. These findings suggest that tilapia relies on dual signaling to maintain an optimal T-cell response, providing a novel perspective for understanding the evolution of the adaptive immune system.
Collapse
Affiliation(s)
- Wei Liang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Haiyou Gao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Kunming Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiansong Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qian Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xinying Jiao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai 200241, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266237, China. E-mail:
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai 200241, China. E-mail:
| |
Collapse
|
2
|
Heitmann JS, Pfluegler M, Jung G, Salih HR. Bispecific Antibodies in Prostate Cancer Therapy: Current Status and Perspectives. Cancers (Basel) 2021; 13:549. [PMID: 33535627 PMCID: PMC7867165 DOI: 10.3390/cancers13030549] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/29/2022] Open
Abstract
Prostate carcinoma (PC) is the second most common cancer in men. When the disease becomes unresponsive to androgen deprivation therapy, the remaining treatment options are of limited benefit. Despite intense efforts, none of the T cell-based immunotherapeutic strategies that meanwhile have become a cornerstone for treatment of other malignancies is established in PC. This refers to immune checkpoint inhibition (CI), which generally reinforces T cell immunity as well as chimeric antigen receptor T (CAR-T) cells and bispecific antibodies (bsAbs) that stimulate the T cell receptor/CD3-complex and mobilize T cells in a targeted manner. In general, compared to CAR-T cells, bsAb would have the advantage of being an "off the shelf" reagent associated with less preparative effort, but at present, despite enormous efforts, neither CAR-T cells nor bsAbs are successful in solid tumors. Here, we focus on the various bispecific constructs that are presently in development for treatment of PC, and discuss underlying concepts and the state of clinical evaluation as well as future perspectives.
Collapse
Affiliation(s)
- Jonas S. Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (J.S.H.); (M.P.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
| | - Martin Pfluegler
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (J.S.H.); (M.P.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| | - Gundram Jung
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, 72076 Tübingen, Germany
| | - Helmut R. Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (J.S.H.); (M.P.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
| |
Collapse
|
3
|
Differentiation into an Effector Memory Phenotype Potentiates HIV-1 Latency Reversal in CD4 + T Cells. J Virol 2019; 93:JVI.00969-19. [PMID: 31578289 PMCID: PMC6880164 DOI: 10.1128/jvi.00969-19] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/15/2019] [Indexed: 12/12/2022] Open
Abstract
By performing phenotypic analysis of latency reversal in CD4+ T cells from virally suppressed individuals, we identify the TEM subset as the largest contributor to the inducible HIV reservoir. Differential responses of memory CD4+ T cell subsets to latency-reversing agents (LRAs) demonstrate that HIV gene expression is associated with heightened expression of transcriptional pathways associated with differentiation, acquisition of effector function, and cell cycle entry. In vitro modeling of the latent HIV reservoir in memory CD4+ T cell subsets identify LRAs that reverse latency with ranges of efficiency and specificity. We found that therapeutic induction of latency reversal is associated with upregulation of identical sets of TEM-associated genes and cell surface markers shown to be associated with latency reversal in our ex vivo and in vitro models. Together, these data support the idea that the effector memory phenotype supports HIV latency reversal in CD4+ T cells. During antiretroviral therapy (ART), human immunodeficiency virus type 1 (HIV-1) persists as a latent reservoir in CD4+ T cell subsets in central memory (TCM), transitional memory (TTM), and effector memory (TEM) CD4+ T cells. We have identified differences in mechanisms underlying latency and responses to latency-reversing agents (LRAs) in ex vivo CD4+ memory T cells from virally suppressed HIV-infected individuals and in an in vitro primary cell model of HIV-1 latency. Our ex vivo and in vitro results demonstrate the association of transcriptional pathways of T cell differentiation, acquisition of effector function, and cell cycle entry in response to LRAs. Analyses of memory cell subsets showed that effector memory pathways and cell surface markers of activation and proliferation in the TEM subset are predictive of higher frequencies of cells carrying an inducible reservoir. Transcriptional profiling also demonstrated that the epigenetic machinery (known to control latency and reactivation) in the TEM subset is associated with frequencies of cells with HIV-integrated DNA and inducible HIV multispliced RNA. TCM cells were triggered to differentiate into TEM cells when they were exposed to LRAs, and this increase of TEM subset frequencies upon LRA stimulation was positively associated with higher numbers of p24+ cells. Together, these data highlight differences in underlying biological latency control in different memory CD4+ T cell subsets which harbor latent HIV in vivo and support a role for differentiation into a TEM phenotype in facilitating latency reversal. IMPORTANCE By performing phenotypic analysis of latency reversal in CD4+ T cells from virally suppressed individuals, we identify the TEM subset as the largest contributor to the inducible HIV reservoir. Differential responses of memory CD4+ T cell subsets to latency-reversing agents (LRAs) demonstrate that HIV gene expression is associated with heightened expression of transcriptional pathways associated with differentiation, acquisition of effector function, and cell cycle entry. In vitro modeling of the latent HIV reservoir in memory CD4+ T cell subsets identify LRAs that reverse latency with ranges of efficiency and specificity. We found that therapeutic induction of latency reversal is associated with upregulation of identical sets of TEM-associated genes and cell surface markers shown to be associated with latency reversal in our ex vivo and in vitro models. Together, these data support the idea that the effector memory phenotype supports HIV latency reversal in CD4+ T cells.
Collapse
|
4
|
Abstract
Cancer immunotherapy is a highly active area in translational medicine where the challenges and rewards of developing new drugs "from bench to bedside" become particularly visible. Here, we comment on both, the scientific and non-scientific hurdles of this translational process using the example of bispecific antibodies (bsAbs) and chimeric antigen receptor (CAR) T cells, two closely related strategies for antibody-guided recruitment of T cells against cancer. Both exert impressive therapeutic activity and were recently approved for treatment of B-cell malignancies. We discuss how the efficacy of these auspicious therapeutic tools may be further improved, in particular against solid tumors, but we also address another critical issue: Since both approaches were already introduced in the 1980s, why did it take almost thirty years until they became clinically available?
Collapse
Affiliation(s)
- Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - Gundram Jung
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| |
Collapse
|
5
|
Osswald A, Hedrich V, Sommergruber W. 3D-3 Tumor Models in Drug Discovery for Analysis of Immune Cell Infiltration. Methods Mol Biol 2019; 1953:151-162. [PMID: 30912021 DOI: 10.1007/978-1-4939-9145-7_10] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The cross talk between tumor cells and other cells present in the tumor microenvironment such as stromal and immune cells highly influences the behavior and progression of disease. Understanding the underlying mechanisms of interaction is a prerequisite to develop new treatment strategies and to prevent or at least reduce therapy failure in the future. Specific reactivation of the patient's immune system is one of the major goals today. However, standard two-dimensional (2D) cell culture techniques lack the necessary complexity to address related questions. Novel three-dimensional (3D) in vitro models-embedded in a matrix or encapsulated in alginate-recapitulate the in vivo situation much better. Cross talk between different cell types can be studied starting from co-cultures. As cancer immune modulation is becoming a major research topic, 3D in vitro models represent an important tool to address immune regulatory/modulatory questions for T, NK, and other cells of the immune system. The 3D systems consisting of tumor cells, fibroblasts, and immune cells (3D-3) already proved as a reliable tool for us. For instance, we made use of those models to study the molecular mechanisms of the cross talk of non-small cell lung cancer (NSCLC) and fibroblasts, to unveil macrophage plasticity in the tumor microenvironment and to mirror drug responses in vivo. Generation of those 3D models and how to use them to study immune cell infiltration and activation will be described in the present book chapter.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Bioreactors
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Cells, Immobilized/drug effects
- Cells, Immobilized/immunology
- Cells, Immobilized/pathology
- Coculture Techniques/methods
- Drug Discovery/methods
- Drug Screening Assays, Antitumor/methods
- Fibroblasts/drug effects
- Fibroblasts/immunology
- Fibroblasts/pathology
- Humans
- Immunity, Cellular/drug effects
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/immunology
- Spheroids, Cellular/pathology
- Tumor Cells, Cultured
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
| | - Viola Hedrich
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | |
Collapse
|
6
|
Co-signal Molecules in T-Cell Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:3-23. [DOI: 10.1007/978-981-32-9717-3_1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
7
|
Jackson SR, Yuan J, Teague RM. Targeting CD8+ T-cell tolerance for cancer immunotherapy. Immunotherapy 2015; 6:833-52. [PMID: 25290416 DOI: 10.2217/imt.14.51] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In the final issue of Science in 2013, the American Association of Science recognized progress in the field of cancer immunotherapy as the 'Breakthrough of the Year.' The achievements were actually twofold, owing to the early success of genetically engineered chimeric antigen receptors (CAR) and to the mounting clinical triumphs achieved with checkpoint blockade antibodies. While fundamentally very different, the common thread of these independent strategies is the ability to prevent or overcome mechanisms of CD8(+) T-cell tolerance for improved tumor immunity. Here we discuss how circumventing T-cell tolerance has provided experimental insights that have guided the field of clinical cancer immunotherapy to a place where real breakthroughs can finally be claimed.
Collapse
Affiliation(s)
- Stephanie R Jackson
- Saint Louis University School of Medicine, Department of Molecular Microbiology & Immunology, 1100 South Grand Blvd, St Louis, MO 63104, USA
| | | | | |
Collapse
|
8
|
Durben M, Schmiedel D, Hofmann M, Vogt F, Nübling T, Pyz E, Bühring HJ, Rammensee HG, Salih HR, Große-Hovest L, Jung G. Characterization of a bispecific FLT3 X CD3 antibody in an improved, recombinant format for the treatment of leukemia. Mol Ther 2015; 23:648-55. [PMID: 25578618 DOI: 10.1038/mt.2015.2] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 12/19/2014] [Indexed: 01/08/2023] Open
Abstract
FLT3 is a receptor-tyrosine-kinase that is expressed on leukemic cells of the myeloid and lymphoid lineage rather specifically. We here report on the construction and selection of bispecific FLT3 X CD3 antibodies in a new recombinant format, termed Fabsc, that resembles the normal antibody structure more closely than the well-established bispecific single chain (bssc)-format. Our preferred antibody, which emerged from an initial selection procedure utilizing different FLT3- and CD3-antibodies, contains the FLT3-antibody 4G8 and the CD3-antibody UCHT1. The 4G8 X UCHT1 Fabsc-antibody was found to be superior to a bssc-antibody with identical specificities with respect to (i) affinity to the target antigen FLT3, (ii) production yield by transfected cells, and (iii) the diminished formation of aggregates. T-cell activation in the presence and absence of cultured leukemic cells and killing of these cells was comparable for both molecules. In addition, the 4G8 X UCHT1 Fabsc-antibody was found to induce T-cell activation and efficient killing of leukemic blasts in primary peripheral blood mononuclear cell (PBMC) cultures of acute myeloid leukemia (AML) patients. In these experiments, the bispecific molecule was clearly superior to an Fc-optimized monospecific FLT3-antibody described previously, indicating that within PBMC of AML patients the recruitment of T cells is more effective than that of natural killer cells.
Collapse
Affiliation(s)
- Michael Durben
- 1] Department of Immunology, Eberhard Karls Universität Tübingen, Tübingen, Germany [2] Present address: Synimmune GmbH, Auf der Morgenstelle 15, Tübingen, Germany
| | - Dominik Schmiedel
- Department of Immunology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Martin Hofmann
- 1] Department of Immunology, Eberhard Karls Universität Tübingen, Tübingen, Germany [2] Present address: Synimmune GmbH, Auf der Morgenstelle 15, Tübingen, Germany
| | - Fabian Vogt
- 1] Department of Immunology, Eberhard Karls Universität Tübingen, Tübingen, Germany [2] German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tina Nübling
- Department of Internal Medicine 2, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Elwira Pyz
- Department of Immunology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Hans-Jörg Bühring
- Department of Internal Medicine 2, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Hans-Georg Rammensee
- 1] Department of Immunology, Eberhard Karls Universität Tübingen, Tübingen, Germany [2] German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Helmut R Salih
- 1] Department of Internal Medicine 2, Eberhard Karls Universität Tübingen, Tübingen, Germany [2] Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ludger Große-Hovest
- 1] Department of Immunology, Eberhard Karls Universität Tübingen, Tübingen, Germany [2] Present address: Synimmune GmbH, Auf der Morgenstelle 15, Tübingen, Germany
| | - Gundram Jung
- 1] Department of Immunology, Eberhard Karls Universität Tübingen, Tübingen, Germany [2] German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
9
|
Renner C, Pfreundschuh M. Status of Bispecific Monoclonal Antibodies for Cancer Therapy. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/bf03259313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
10
|
Riethmüller G. Symmetry breaking: bispecific antibodies, the beginnings, and 50 years on. CANCER IMMUNITY 2012; 12:12. [PMID: 22896757 PMCID: PMC3380354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Affiliation(s)
- Gert Riethmüller
- Institute for Immunology, Ludwig-Maximilians-Universität, München, Germany.
| |
Collapse
|
11
|
Thompson S, Dessi J, Self CH. The construction and in vitro testing of photo-activatable cancer targeting folated anti-CD3 conjugates. Biochem Biophys Res Commun 2007; 366:526-31. [PMID: 18068664 DOI: 10.1016/j.bbrc.2007.11.158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2007] [Accepted: 11/30/2007] [Indexed: 10/22/2022]
Abstract
The construction and in vitro testing of a photo-activatable anti-tumour immuno-regulatory antibody is described. In this 'cloaked' folated anti-CD3 antibody conjugate, the folate portion of the conjugate is free to bind to folate receptor expressing cancer cells, whilst the anti-CD3 activity is effectively rendered inert by a coating of photo-labile 2-nitrobenzyl groups. On irradiation with UV-A light the activity of the anti-CD3 antibody is restored, not only when it is required, but more importantly, only where it is required. The conjugate can then attract killer T-cells to the surface of the tumour cells and kill them. Unirradiated normal tissues, to which the conjugate has been targeted by specific and non-specific binding, remain unharmed. We believe that these 'photo-switchable' conjugates could be used to markedly improve the targeting of the immune response to folate receptor (FR) expressing ovarian and breast cancers whilst minimising the side effects in the rest of the body.
Collapse
Affiliation(s)
- Stephen Thompson
- Diagnostic and Therapeutic Technologies, School of Clinical and Laboratory Sciences, University of Newcastle Upon Tyne, The Medical School, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| | | | | |
Collapse
|
12
|
Schilbach K, Kerst G, Walter S, Eyrich M, Wernet D, Handgretinger R, Xie W, Rammensee HG, Müller I, Bühring HJ, Niethammer D. Cytotoxic minor histocompatibility antigen HA-1–specific CD8+ effector memory T cells: artificial APCs pave the way for clinical application by potent primary in vitro induction. Blood 2005; 106:144-9. [PMID: 15731181 DOI: 10.1182/blood-2004-07-2940] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Induction of cytotoxic T lymphocytes (CTLs) for treatment of relapsed leukemia after allogeneic stem-cell transplantation is hindered by the laborious and time-consuming procedure of generating dendritic cells for antigen presentation. Artificial antigen-presenting cells (aAPCs) offer the advantage of being readily available in sufficient numbers, thus allowing for a highly standardized in vitro induction of CTLs. We generated aAPCs coated with anti-CD28 antibody (Ab) and either high-density (HD) or low-density (LD) major histocompatibility complex (MHC) class I molecules loaded with HA-1H, a nonapeptide derived from the hematopoiesis-restricted minor histocompatibility antigen HA-1. HD- and LD-aAPCs potently induced HA-1H–specific CD8+ CTLs from untouched CD8+ T cells of healthy donors. CTLs were subsequently purified by magnetic-activated cell sorting. HD- as well as LD-aAPC–induced CTLs exerted high HA-1H–specific cytotoxicity, resembled Tc1 effector memory cells, survived a long time in vitro, and were expanded by a factor varying between 8.2 × 104 and 51 × 104. The T-cell receptor (TCR) repertoire of HA-1H tetramer–positive CTLs was oligoclonal with a prominent usage of Vβ6. The TCR repertoire of tetramer-positive CTLs was distinct from and more restricted than that of tetramer-negative cells. These findings indicate that aAPCs are attractive tools for the ex vivo generation of HA-1H–specific CTLs suitable for immunotherapy of relapsed leukemia.
Collapse
Affiliation(s)
- Karin Schilbach
- University Children's Hospital Tübingen, Hoppe-Seyler-Str 1, D-72076 Tübingen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Walter S, Herrgen L, Schoor O, Jung G, Wernet D, Bühring HJ, Rammensee HG, Stevanović S. Cutting edge: predetermined avidity of human CD8 T cells expanded on calibrated MHC/anti-CD28-coated microspheres. THE JOURNAL OF IMMUNOLOGY 2004; 171:4974-8. [PMID: 14607891 DOI: 10.4049/jimmunol.171.10.4974] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cytotoxic CD8 T cells are key effectors in the immunotherapy of malignant and viral diseases. However, the lack of efficient methods for their in vitro priming and expansion has become a bottleneck to the development of vaccines and adoptive transfer strategies. Synthetic artificial APCs (aAPCs) are now emerging as an attractive tool for eliciting and expanding CTL responses. We show that, by controlling the MHC density on aAPCs, high- or low-avidity tumor-directed human CTL lines can be raised effectively in vitro if costimulation via CD28 and IL-12 is provided. Compared with low-avidity CTL lines, high-avidity CTLs need 100- to 1000-fold less peptide for activation, bind more MHC tetramers, and, as expected, are superior in recognizing tumor cell lines expressing Ag. We believe that the possibility to raise Ag-specific T cells with predetermined avidity will be crucial for the future use of aAPCs in immunotherapeutical settings.
Collapse
Affiliation(s)
- Steffen Walter
- Department of Immunology, Institute for Cell Biology, Division of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Kovacs B, Maus MV, Riley JL, Derimanov GS, Koretzky GA, June CH, Finkel TH. Human CD8+ T cells do not require the polarization of lipid rafts for activation and proliferation. Proc Natl Acad Sci U S A 2002; 99:15006-11. [PMID: 12419850 PMCID: PMC137535 DOI: 10.1073/pnas.232058599] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Lipid rafts are important signaling platforms in T cells. Little is known about their properties in human CD8(+) T cells. We studied polarization of lipid rafts by digital immunofluorescence microscopy in primary human T cells, using beads coated with anti-CD3 and anti-CD28 mAbs (CD3/28 beads). Unlike CD4(+) T cells, CD8(+) T cells did not polarize lipid rafts when stimulated with CD3/28 beads, when the anti-CD28 antibody was substituted with B7.2Ig, or if an anti-CD8 antibody was added to the CD3/28 beads. This phenomenon was also observed in human antigen-specific CD8(+) T cells. On stimulation with CD3/28 beads, the T cell antigen receptor clustered at the cell/bead contact area in both CD4(+) and CD8(+) T cells. Examination of lipid rafts isolated by sucrose density gradient centrifugation revealed the constitutive expression of p(56)Lck in the raft fractions of unstimulated CD8(+) T cells, whereas p(56)Lck was recruited to the raft fraction of CD4(+) T cells only after stimulation with CD3/28 beads. Stimulation with CD3/28 beads induced marked calcium flux, recruitment of PKC-theta and F-actin to the cell/bead contact site, and similar proliferation patterns in CD4(+) and CD8(+) T cells. Thus, polarization of lipid rafts is not essential for early signal transduction events or proliferation of human CD8(+) lymphocytes. It is possible that the lower stringency of CD8(+) T cell activation obviates a requirement for raft polarization.
Collapse
Affiliation(s)
- Birgit Kovacs
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Brandl M, Grosse-Hovest L, Holler E, Kolb HJ, Jung G. Bispecific antibody fragments with CD20 X CD28 specificity allow effective autologous and allogeneic T-cell activation against malignant cells in peripheral blood and bone marrow cultures from patients with B-cell lineage leukemia and lymphoma. Exp Hematol 1999; 27:1264-70. [PMID: 10428503 DOI: 10.1016/s0301-472x(99)00072-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Bispecific antibodies directed against tumor-associated target antigens and to surface receptors mediating T-cell activation, such as the TCR/CD3 complex and the costimulatory receptor CD28, are capable of mediating T-cell activation resulting in tumor cell killing. In this study, we used the B-cell-associated antigens CD19 and CD20 as target structures on human leukemic cells. We found that a combination of bispecific antibody fragments (bsFab2) with target x CD3 and target x CD28 specificity induces vigorous autologous T-cell activation and killing of malignant cells in peripheral blood and bone marrow cultures from patients with chronic lymphocytic leukemia and follicular lymphoma. The bsFab2 targeting CD20 were considerably more effective than those binding to CD19. The colony-forming capacity of treated bone marrow was impaired due to large amounts of tumor necrosis factor alpha produced during bsFab2-induced T-cell activation. Neutralizing tumor necrosis factor alpha antibodies were found to reverse this negative effect without affecting T-cell activation and tumor cell killing. CD20 x CD28 bsFab2, when used alone rather than in combination, markedly improved the recognition of leukemic cells by allogeneic T cells. Therefore, these reagents may be capable of enhancing the immunogenicity of leukemic cells in general and, in particular, of increasing the antileukemic activity of allogeneic donor buffy coat cells in relapsed bone marrow transplanted patients.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Antibodies, Neoplasm/immunology
- Antibody Specificity
- Antigens, CD19/immunology
- Antigens, CD20/immunology
- Antigens, Neoplasm/immunology
- Bone Marrow/pathology
- CD28 Antigens/immunology
- Cytotoxicity, Immunologic
- Humans
- Immunoglobulin Fab Fragments/immunology
- Leukemia, B-Cell/immunology
- Leukemia, B-Cell/pathology
- Lymphocyte Activation/drug effects
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Receptor-CD3 Complex, Antigen, T-Cell/immunology
- T-Lymphocytes/immunology
- Tumor Cells, Cultured/immunology
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- M Brandl
- Department of Hematology and Medical Oncology, Klinikum Grosshadern, University of Munich, Germany
| | | | | | | | | |
Collapse
|
16
|
Abstract
Immunotherapy is a powerful anti-cancer treatment modality. However, despite numerous encouraging results obtained in pre-clinical studies, a definite breakthrough towards an established clinical treatment modality has as yet not occurred. Antibodies against tumor antigens have been shown to localise at the site of the tumor, but inadequate triggering of immune effector mechanisms have thwarted clinical efficacy thus far. Cellular immunotherapy has been hampered by limitations such as lack of specificity, down-regulation of major histocompatibility complex (MHC)-expression or Fas ligand up-regulation on tumor cells. This review focuses on the use of bispecific antibodies (BsAbs) for immunotherapy of cancer. Using BsAbs, it is possible to take advantage of the highly specific binding characteristics of antibodies and combine these with the powerful effector functions of cytotoxic immune effector cells. BsAbs share two different, monoclonal antibody-derived, antigen-recognizing moieties within one molecule. By dual binding, BsAbs reactive with a trigger molecule on an immune effector cell on the one hand and a surface antigen on a tumor target cell on the other are thus able to functionally focus the lytic activity of the immune effector cell towards the target cell. Over the last few years, the concept of BsAb-mediated tumor cell killing has been studied extensively both in preclinical models and in a number of phase I clinical trials. Promising pre-clinical results have been reported using tumor models in which diverse immune effector cell populations have been used. Despite this pre-clinical in vivo efficacy, the first clinical trials indicate that we are still not in a position to successfully treat human malignancies. This review discusses the production of BsAbs, the choice of trigger molecules in combination with potential effector cells and the preclinical models that have led to the current use of BsAbs in experimental clinical trials. It has become clear that appropriate immune cell activation and establishing a favourable effector-to-target cell ratio will have direct impact on the efficacy of the therapeutic approaches using BsAbs. New directions are discussed, i.e. finding appropriate dosage schemes by which immune effector cells become redirected without inducing hyporesponsiveness, defining possibilities for combining different immune effector cell populations and creating an in situ tumor environment that allows maximal tumoricidal activity
Collapse
|
17
|
Renner C, Held G, Ohnesorge S, Bauer S, Gerlach K, Pfitzenmeier JP, Pfreundschuh M. Role of naive and memory T cells in tumor cell lysis mediated by bi-specific antibodies. Immunobiology 1997; 197:122-32. [PMID: 9241536 DOI: 10.1016/s0171-2985(97)80062-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Bispecific monoclonal antibodies (Bi-mAb) with specificity for a tumor associated antigen and the CD3 or CD28 antigen on T lymphocytes, respectively, induce activation of resting T lymphocytes and target-specific tumor cell lysis. Former studies had confirmed that T cells expressing the CD45RO "memory" antigen at high levels were the most potent effectors of Bi-mAb-mediated cytotoxicity when compared to their "naive" counterparts expressing the CD45RA antigen. Further analysis of the T cell subpopulations revealed that within the memory T cell pool, CD8+ T cells were the effector cell, population with strongest cytolytic activity. The cytolytic activity was correlated with the expression level of perforin and granzymes B mRNA. Ca2+ complexing agents, which abrogate perforin activity, reduced necrosis, while inhibition of granzyme activity in effector or target-cells had a similar effect on apoptosis. These results confirm the crucial role perforin and granzymes play in target-cell lysis and explain why CD8+CD45RO+ T cells activated by combined CD3 and CD28 antigen triggering represent the T cell pool with highest cytolytic potential.
Collapse
Affiliation(s)
- C Renner
- Department of Hematology and Oncology, University of Saarland, Homburg/Saar, Germany
| | | | | | | | | | | | | |
Collapse
|
18
|
Hayden MS, Grosmaire LS, Norris NA, Gilliland LK, Winberg G, Tritschler D, Tsu TT, Linsley PS, Mittler RS, Senter PD, Fell HP, Ledbetter JA. Costimulation by CD28 sFv expressed on the tumor cell surface or as a soluble bispecific molecule targeted to the L6 carcinoma antigen. TISSUE ANTIGENS 1996; 48:242-54. [PMID: 8946677 DOI: 10.1111/j.1399-0039.1996.tb02642.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Interaction of the CD80 (B7-1) and CD86 (B7-2) molecules on antigen presenting cells with the receptors CD28 and CTLA-4 on T cells generates signals important in the regulation of immune responses. Because this receptor system involves multiple receptor-ligand interactions, determining the function for individual receptors has been difficult. One approach is the use of antibodies and their derivatives with singular specificity as substitute ligands to explore the activities of these molecules. We have constructed recombinant mono- and bi-specific sFv molecules specific for the CD28 receptor that are capable of binding and generating costimulatory signals to activate T cells. We demonstrate that these soluble molecules are capable of higher levels of costimulation than soluble CD80Ig at equivalent concentrations. We also constructed artificial adhesion receptors on the cell surface using two different CD28-specific sFvIgs fused to the CD80 cytoplasmic and transmembrane domains. In this report, we compared costimulation by a soluble bispecific (alpha CD28-alpha L6) single chain sFvIg fusion protein to that generated by L6 antigen positive (L6+) H3347 tumor cells transduced with cell surface expressed forms of alpha CD28 sFv's. We show that the bispecific protein can target potent CD28 costimulatory activity to L6+ tumor cells in vitro. We also show that transfection of the cell surface forms of the two different CD28 sFvIgs into H3347 tumor cells allows them to generate significant costimulatory signals to activated T cells. Finally, we demonstrate that tumor cell presentation of either the soluble bispecific or transduced cell surface sFv generate similar costimulatory effects resulting in T cell activation.
Collapse
Affiliation(s)
- M S Hayden
- Bristol-Myers Squibb Pharmaceutical Research Institute, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Chapoval AI, Nelson H, Thibault C, Penna C, Dean P. Bifunctional antibody retargeting in vivo-activated T lymphocytes: simplifying clinical application. JOURNAL OF HEMATOTHERAPY 1995; 4:571-7. [PMID: 8846018 DOI: 10.1089/scd.1.1995.4.571] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
For antitumor x anti-CD3 bifunctional antibody (BFA) therapy to be clinically relevant in solid tumors, activated lymphocytes must be present within tumors. Toward that end, three uniquely different in vivo activation approaches were investigated in a p97 human antigen expressing syngeneic murine melanoma model. beta-Glucan (200 micrograms), staphylococcal enterotoxin B (SEB) (50 micrograms), and F(ab')2 BFA (10 micrograms) were tested for their ability to activate lymphocytes, neutralize pulmonary metastases, and treat established tumors. Systemic activation, measured as the ability of splenocytes to lyse tumor cells in vitro in the presence of BFA, was enhanced by the in vivo administration of SEB but not by beta-glucan or F(ab')2 BFA. Despite lacking a systemic effect, F(ab')2 BFA increased both direct and BFA-mediated cytotoxicity in fresh tumor infiltrating lymphocytes. beta-Glucan did not increase systemic or intratumor T cell activation. However, it significantly enhanced the ability of splenocytes to lyse NK-sensitive YAC-1 cells. When tested in a pulmonary metastases model, all three forms of immune modulation combined with F(ab')2 BFA significantly reduced the number of metastases. BFA were more effective at tumor neutralization when combined with SEB compared with adoptively transferred, in vitro-activated splenocytes. These studies demonstrate that immune modulators when combined with F(ab')2 BFA can provide effective antitumor therapy. Several clinical obstacles may be overcome by the application of these reagents.
Collapse
Affiliation(s)
- A I Chapoval
- Mayo Clinic and Foundation, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
20
|
Sanna PP, De Logu A, Williamson RA, Samson ME, Altieri DC, Bloom FE, Burton DR. Rapid assay of phage-derived recombinant human fabs as bispecific antibodies. BIO/TECHNOLOGY (NATURE PUBLISHING COMPANY) 1995; 13:1221-4. [PMID: 9636296 DOI: 10.1038/nbt1195-1221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Specific anti-tumor and anti-viral activities can be conferred on lymphocytic and myeloid effector cells by retargeting them with bispecific antibodies. These are antibodies which possess an anti-target binding region and a region capable of binding specific effector cell surface markers. For the rapid evaluation of recombinant human Fabs as bispecific antibodies, we have constructed a vector that allows for the conversion of Fabs into protein A fusion proteins. These can be used to generate bispecific antibodies when complexed to appropriate anti-effector cell immunoglobulins. As a model system, a protein A fusion derivative of a human recombinant anti-herpes simplex virus (HSV) Fab was constructed and complexed to OKT3, a T cell-activating antibody specific for CD3. This complex reduced HSV-2 yields in infected cells by about three logs relative to controls when incubated on HSV-2-infected cell monolayers in the presence of IL-2-activated lymphocytes. The system described allows for the rapid evaluation of recombinant human Fabs as bispecific antibodies for therapeutic applications. In addition, Fab-protein A fusion proteins can be used in ELISA and other immuno-assays with increased sensitivity.
Collapse
Affiliation(s)
- P P Sanna
- Department of Neuropharmacology, Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Kroesen BJ, Helfrich W, Bakker A, Wubbena AS, Bakker H, Kal HB, The TH, de Leij L. Reduction of EGP-2-positive pulmonary metastases by bispecific-antibody-redirected T cells in an immunocompetent rat model. Int J Cancer 1995; 61:812-8. [PMID: 7790116 DOI: 10.1002/ijc.2910610612] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Effectiveness of bispecific-monoclonal-antibody (BsMAb)-mediated cellular anti-tumour activity was evaluated in vitro and in vivo in relation to the additional need for T-cell activation in a new immunocompetent rat tumour model. L37 tumour cells, derived from a squamous-cell carcinoma of the lung of Wag/Rij rats, were transfected with the cDNA coding for the human 38-kDa transmembrane pan-carcinoma-associated antigen EGP-2. Intravenous inoculation of EGP-2-positive L37 cells resulted in a rapid outgrowth of EGP-2-positive tumour nodules in the lungs. A BsMAb BIS-19, recognizing EGP-2 on the transfected tumour cells and the T-cell receptor of the rat, was made and allowed specific lysis of EGP-2-transfected L37 tumour cells by activated rat T lymphocytes in vitro. In vivo T-cell activation, assessed by up-regulation of IL-2-receptor expression, could be induced by daily injection of rat rIL-2. Intravenous treatment of tumour-bearing EGP-2-positive L37 tumour with BIS-19 together with rat rIL-2 resulted in almost complete disappearance of established tumour. In contrast, animals treated with BIS-19 alone, IL-2 alone or a combination of anti-EGP-2, anti-TcR and IL-2 showed much less or no tumour reduction. These results show effectiveness of systemic treatment with BsMAbs to induce anti-tumour activity in established tumours. Immune activation prior to or during treatment with BsMAbs, as achieved with IL-2, appears to be a prerequisite for successful treatment.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/pharmacology
- Antibodies, Monoclonal/pharmacology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/secondary
- Carcinoma, Squamous Cell/therapy
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Cytotoxicity Tests, Immunologic
- Epithelial Cell Adhesion Molecule
- Female
- Flow Cytometry
- Immunocompetence
- Immunoenzyme Techniques
- Immunotherapy
- Interleukin-2/pharmacology
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Lymphocyte Activation
- Rats
- Rats, Inbred Strains
- Receptors, Antigen, T-Cell/immunology
- Recombinant Proteins/pharmacology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/physiology
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- B J Kroesen
- Department of Clinical Immunology, University Hospital Groningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Many strategies for experimental immunotherapy of cancers aim at inducing and expanding tumor-specific cytotoxic T-lymphocytes. One of the most promising approaches uses bispecific monoclonal antibodies (Bi-MAbs) which are able to accumulate and activate human effector cells at the tumor site. Human resting peripheral NK- or T cells targeted by appropriate Bi-MAbs to tumor cells expressing a tumor-associated antigen display multiple signs of activation including proliferation, cytokine secretion, upregulation of cytotoxic peptides and enzymes and induce an efficient tumor cell lysis in vivo. Moreover, tumor-bearing SCID which were treated by effector cell-triggering Bi-MAbs and human peripheral blood lymphocytes had complete regressions of established tumors and most or all animals were cured by human NK-cell or T-cell cytotoxicity, respectively. Local tumor site-specific activation of T-cells was demonstrated, and enhanced granzyme and perforin expression together with the results of inhibition experiments suggest both mechanisms as the major contributors to the cytolytic machinery of Bi-MAb-mediated T-cell cytotoxicity in vivo. The encouraging results of this approach, which is able to cure animals with even advanced disseminated tumors, together with the local site-specific effector cell activation, which suggests minimal side effects, warrant the clinical evaluation of this Bi-MAb approach. As lymphocytes from tumor patients can be adequately activated by the respective Bi-MAbs, the clinical application of Bi-MAbs promises to become a safe, efficient and simple approach which should be readily applicable to the treatment of human malignancies that cannot be cured by standard regimens.
Collapse
Affiliation(s)
- C Renner
- Med. Klinik I, University of the Saarland, Homburg/Sar, Germany
| | | |
Collapse
|
24
|
Abstract
Bispecific monoclonal antibodies (bsAbs) that recognize CD3 with one arm and a tumor associated antigen with the other arm can retarget T-cells toward tumor cells in an MHC independent manner, thereby combining the specificity of monoclonal antibodies with the power of the cellular immune system. B-cell malignancies are particularly attractive as targets for anti-CD3-based bsAb therapy because of their sensitivity to other forms of antibody therapy, and the extent to which B-cells and T-cells communicate at the molecular level. BsAbs that recognize CD3 and a number of antigens on malignant B-cells have been shown in vitro to be capable of retargeting T-cells. In animal models of B-cell malignancy, bsAb can eliminate tumor loads that are resistant to unmodified monoclonal antibody therapy. Ongoing early clinical trials in advanced B-cell lymphoma indicate CD3-based bsAbs have significant biologic effects, and suggest they have anti-tumor activity as well. A number of significant questions relating to bsAb therapy of B-cell malignancies remain. It is unclear what role both endogenously produced and exogenously administered cytokines are likely to play. Further exploration of whether bsAb can induce T-cells to target to tumor will also be required before the true promise of this novel form of immunotherapy can be determined.
Collapse
Affiliation(s)
- G J Weiner
- Department of Internal Medicine, University of Iowa, Iowa City 52242, USA
| | | |
Collapse
|
25
|
Wolf H, Müller Y, Salmen S, Wilmanns W, Jung G. Induction of anergy in resting human T lymphocytes by immobilized anti-CD3 antibodies. Eur J Immunol 1994; 24:1410-7. [PMID: 8206102 DOI: 10.1002/eji.1830240626] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
How the T cell receptor (TcR)/CD3 complex mediates not only the induction of T cell activation but also suppressive effects like T cell anergy or apoptosis is not well understood. Here we describe a series of preincubation and restimulation experiments which demonstrate that primary stimulation of resting, unseparated human T cells with mitogenic doses of immobilized anti-CD3 antibodies induces hyporesponsiveness upon restimulation of the cells. Various costimuli can prevent this type of anergy to a variable degree if present during the preincubation period, phorbol 12-myristate 13-acetate (PMA) being the most and anti-CD4 antibody the least effective. If employed together with anti-CD3 antibody during the restimulation phase of the assay, interleukin (IL)-2, IL-4 and anti-CD28 antibody break anergy almost completely. Proliferation induced by a submitogenic dose of anti-CD3 antibody supplemented by costimulatory signals (anti-CD2, anti-CD4, anti-CD28, IL-2, IL-4 or PMA) does not result in hyporesponsiveness. Taken together, these results support a modified view of the two-signal model for T cell activation according to which anergy induction in resting T cells occurs if primary proliferation is induced by high density triggering of the TcR/CD3 complex in the absence of accessory signals. We discuss possible implications of these findings for the induction of peripheral tolerance.
Collapse
Affiliation(s)
- H Wolf
- Labor für Rationale Immuntherapie, Medizinische Klinik III der Universität München, FRG
| | | | | | | | | |
Collapse
|
26
|
Renner C, Jung W, Sahin U, Denfeld R, Pohl C, Trümper L, Hartmann F, Diehl V, van Lier R, Pfreundschuh M. Cure of xenografted human tumors by bispecific monoclonal antibodies and human T cells. Science 1994; 264:833-5. [PMID: 8171337 DOI: 10.1126/science.8171337] [Citation(s) in RCA: 119] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Tumor immunotherapy should increase both the number of T cells that kill the tumor and the likelihood that those cells are activated at the tumor site. Bispecific monoclonal antibodies (Bi-mAbs) were designed that bound to a Hodgkin's tumor-associated antigen (CD30) on the tumor and to either CD3 or CD28 on the T cell. Immunodeficient mice were cured of established human tumors when mice were treated with both the CD3-CD30 and the CD28-CD30 Bi-mAbs and then given human peripheral blood lymphocytes that had been incubated with the CD3-CD30 Bi-mAb and cells that expressed CD30. The enrichment of human T cells within the tumor and the fact that established tumors can be cured may indicate in situ activation of both the T cell receptor and the costimulatory pathway.
Collapse
Affiliation(s)
- C Renner
- Medizinische Klinik und Poliklinik, Universität des Saarlandes, Homburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Pohl C, Denfeld R, Renner C, Jung W, Bohlen H, Sahin U, Hombach A, van Lier R, Schwonzen M, Diehl V. CD30-antigen-specific targeting and activation of T cells via murine bispecific monoclonal antibodies against CD3 and CD28: potential use for the treatment of Hodgkin's lymphoma. Int J Cancer 1993; 54:820-7. [PMID: 7686889 DOI: 10.1002/ijc.2910540517] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cross-linking of specific tumor antigens with the T-cell-associated CD3 and CD28 antigens can increase IL-2 secretion, proliferation and antigen-specific cytotoxicity in resting T cells. This cross-linking can be achieved effectively by bispecific monoclonal antibodies (BiMAb) with specificity for both the tumor antigen and CD3 or CD28 antigen, respectively. To take advantage of the enhanced activation of CD3 pre-activated T cells by additional activation via the CD28 antigen, BiMAb OKT3/HRS-3 with reactivity to both CD3 and the Hodgkin's-lymphoma-associated CD30 antigen and the BiMAb 15E8/HRS-3 with reactivity to both CD28 and CD30 antigen were generated by hybridoma fusion. Resting T cells, represented by Jurkat cells (CD3+/CD28+) were specifically activated to produce IL-2 by co-cultivation with an EBV-transformed B-cell line (LAZ509, CD30+/CD19+) only in the presence of the CD30/CD28 cross-linking BiMAb and an additional cross-linking anti-CD3/CD19 BiMAb (OKT3/6A4). Neither the cross-linking BiMAbs alone nor any combination of the monospecific parental MAbs induced a comparable IL-2 production by Jurkat cells in the presence of LAZ509. In addition, using a combination of these BiMAbs, an antigen-dependent cytotoxicity was induced by targeting APC-depleted peripheral blood lymphocytes to CD30+ L540 cells. T cells, previously specifically activated by CD3/CD30 in the presence of CD30 antigen, were cytotoxic to CD30+ cell lines only after incubation with BiMAb anti-CD28/CD30. Neither of the BiMAbs nor any of the parental antibodies induced a comparable effect. Our results indicate that such BiMAbs may offer a new approach for specific immunotherapy of Hodgkin's lymphoma, which takes advantage of cytokine secretion and cytotoxicity of activated T cells.
Collapse
Affiliation(s)
- C Pohl
- Klinik I für Innere Medizin, Universität zu Köln, Cologne, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Reid I, Lundy J, Monson J, Nelson H, Ramsay P, Ilstrup D, Donohue J. Heteroconjugated antibodies enhance lymphocyte-mediated tumour cell lysis in vitro and in vivo. Br J Surg 1992; 79:628-32. [PMID: 1386550 DOI: 10.1002/bjs.1800790710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Covalent linkage of an antitumour antibody specific for a tumour cell surface antigen to an antilymphocyte antibody specific for the T lymphocyte receptor complex produces a heteroconjugated antibody that can activate and redirect cytotoxic T lymphocytes to lyse tumour cells. The ability of an antilymphocyte-antitumour heteroconjugate (500A2 x 96.5) to direct the lysis of murine melanoma cells by cultured murine lymphocytes was tested in vitro using a 4-h chromium release assay and in vivo with a tumour neutralization assay. In vitro, the addition of heteroconjugated antibody significantly increased tumour lysis by murine C3H/HeN lymphocytes (median specific lysis 82.7 per cent with lymphocytes plus heteroconjugate versus 9.5 per cent for lymphocytes alone, P less than 0.001). In vivo, treatment with heteroconjugated antibody plus lymphocytes significantly reduced the development of pulmonary metastases after intravenous tumour administration (median number of pulmonary metastases 28.5 for combined treatment versus 250 for heteroconjugate or lymphocytes alone, P less than 0.001).
Collapse
Affiliation(s)
- I Reid
- Department of Surgery, Mayo Clinic, Rochester, Minnesota 55905
| | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Chen Q, Sun P, Prigione I, Xie H, Ferrini S. Targeting of «T» Lymphocytes against Human Hepatoma Cells by a Bispecific Monoclonal Antibody: Role of Different Lymphocyte Subsets. TUMORI JOURNAL 1992; 78:79-86. [PMID: 1326141 DOI: 10.1177/030089169207800204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In an attempt to construct bispecific monoclonal antibodies (bimAbs) able to target cytotoxic T lymphocytes against human hepatoma cells, an HGPRT-deficient mutant of the Hepama-6 hybridoma, which produces an antihuman-hepatoma mAb, was directly fused with splenocytes from Balb/C mice immunized by a polyclonal cytotoxic T-cell line. Hybrid hybridomas were selected in HAT medium, and their supernatants were directly screened for the ability to induce IL-2-cultured cytotoxic T lymphocytes to kill hepatoma cells in a 51Cr-release assay. The selected hybrid hybridoma, termed DQ-33, secretes a bimAb, which reacts with a CD3-associated determinant. When resting peripheral-blood lymphocytes were used as effector cells, virtually no cytolytic activity could be induced by DQ-33, whereas phytohemagglutinin-activated lymphocytes that had been expanded in vitro in IL-2-containing medium could be efficiently targeted against hepatoma cells. Targeting by DQ-33 bimAb was analyzed on different subsets of IL-2-cultured lymphocytes. It was evident that CD+4–8+ TCRα/β+ and CD3+4–8-TCRγ/δ+ lymphocytes were efficiently induced by bimAb to lyse human hepatoma cells, whereas no induction of cytolysis could be observed when CD3 + 4+8-TCRα/β+ cells were used as effectors. DQ-33 bimAb was also able to induce lymphokine secretion (IL-2, GM-CSF and TNF-α) by all the different subsets of lymphocytes analyzed in the presence of target cells expressing the relevant antigen, independent of the expression of cytolytic activity.
Collapse
Affiliation(s)
- Q Chen
- Shanghai Institute of Cell Biology, Chinese Academy of Science
| | | | | | | | | |
Collapse
|
31
|
Azuma M, Cayabyab M, Buck D, Phillips JH, Lanier LL. CD28 interaction with B7 costimulates primary allogeneic proliferative responses and cytotoxicity mediated by small, resting T lymphocytes. J Exp Med 1992; 175:353-60. [PMID: 1370679 PMCID: PMC2119127 DOI: 10.1084/jem.175.2.353] [Citation(s) in RCA: 266] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Engagement of the CD3/T cell antigen receptor complex on small, resting T cells is insufficient to trigger cell-mediated cytotoxicity or to induce a proliferative response. In the present study, we have used genetic transfection to demonstrate that interaction of the B7-BB1 B cell activation antigen with the CD28 T cell differentiation antigen costimulates cell-mediated cytotoxicity and proliferation initiated by either anti-CD2 or anti-CD3 monoclonal antibody (mAb). Moreover, a B7- negative Burkitt's lymphoma cell line that fails to stimulate an allogeneic mixed lymphocyte response is rendered a potent stimulator after transfection with B7. The mixed leukocyte reaction proliferative response against the B7 transfectant is inhibited by either anti-CD28 or B7 mAb. We also demonstrate that freshly isolated small, resting human T cells can mediate anti-CD3 or anti-CD2 mAb-redirected cytotoxicity against a murine Fc receptor-bearing mastocytoma transfected with human B7. These preexisting cytotoxic T lymphocytes in peripheral blood are present in both the CD4 and CD8 subsets, but are preferentially within the CD45RO+ "memory" population. While small, resting T cells apparently require costimulation by CD28/B7 interactions, this requirement is lost after T cell activation. Anti- CD3 initiates a cytotoxic response mediated by in vitro cultured T cell clones in the absence of B7 ligand. The existence of functional cytolytic T cells in the small, resting T cell population may be advantageous in facilitating rapid responses to immune challenge.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Surface/immunology
- B-Lymphocytes/immunology
- B7-1 Antigen
- CD2 Antigens
- CD28 Antigens
- CD3 Complex
- Cytotoxicity, Immunologic/immunology
- Flow Cytometry
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred BALB C
- Receptors, Antigen, T-Cell/immunology
- Receptors, Immunologic/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- M Azuma
- Department of Immunology, DNAX Research Institute for Molecular and Cellular Biology, Palo Alto, California 94304
| | | | | | | | | |
Collapse
|
32
|
von Fliedner V, Miescher S, Gérain J, Gallati H, Barras C, Heumann D, Cerottini JC. Production of tumor necrosis factor-alpha by naive or memory T lymphocytes activated via CD28. Cell Immunol 1992; 139:198-207. [PMID: 1309488 DOI: 10.1016/0008-8749(92)90112-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
While it is well established that activated T cells can produce tumor necrosis factor alpha (TNF-alpha), it is less clear whether this function is confined to a given subset, e.g., memory cells. To approach this question, we investigated the production of TNF-alpha by human peripheral blood T lymphocytes activated with anti-CD28 mAb since this activation pathway is known to potentiate cytokine production. Under the culture conditions used, the amount of TNF-alpha produced was markedly enhanced compared to that obtained after activation with immobilized anti-CD3 mAb. The enhancement of TNF-alpha production was already apparent after incubation of T cells for 6 hr. Up to 5 ng/ml of TNF-alpha was measured on Day 2 in supernatants of cultures of 10(4) T lymphocytes. To determine the source of the cells producing high amounts of TNF-alpha, T lymphocytes were separated into two subpopulations, namely naive cells (expressing the CD45RA isoform) and memory cells (expressing the CD45RO isoform). While both subpopulations proliferated equally well after stimulation with anti-CD28 mAb, up to 90% of the TNF-alpha produced under these conditions originated from memory T cells. These results thus document that T cell activation via CD28 results in a marked increase in TNF-alpha production without affecting the functional disparity that exists between naive and memory T cells.
Collapse
Affiliation(s)
- V von Fliedner
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | | | | | | | | | | | | |
Collapse
|
33
|
Dohlsten M, Hedlund G, Akerblom E, Lando PA, Kalland T. Monoclonal antibody-targeted superantigens: a different class of anti-tumor agents. Proc Natl Acad Sci U S A 1991; 88:9287-91. [PMID: 1924393 PMCID: PMC52699 DOI: 10.1073/pnas.88.20.9287] [Citation(s) in RCA: 81] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The bacterial superantigen staphylococcal enterotoxin (SE) A (SEA) directs cytotoxic T lymphocytes (CTLs) expressing particular sequences of the T-cell receptor (TCR) beta chain to lyse tumor cells expressing major histocompatibility complex (MHC) class II molecules, which serve as receptors for SEs. We now report that chemical conjugates of SEA and the colon carcinoma-reactive monoclonal antibodies (mAbs) C215 or C242 mediate T cell-dependent destruction of colon carcinoma cells lacking MHC class II molecules. SEA was covalently linked to the mAbs C215 and C242 via a PEG-based hydrophilic spacer. The C215-SEA conjugate targeted CD4+ as well as CD8+ CTLs to lyse a panel of colon carcinoma cells lacking MHC class II molecules. T-cell recognition of mAb-SEA conjugates was SEA specific, since SEB-selective T-cell lines with potent cytotoxic activity towards Raji cells coated with SEB did not respond to the C215-SEA conjugate. Unconjugated SEA did not induce T-cell lysis of MHC class II- colon carcinoma cells but efficiently directed CTLs against MHC class II+ Raji cells and certain interferon-treated MHC class II+ colon carcinoma cells. These results suggest that SEA-mAb conjugates retain the SEA-related selectivity for certain TCR beta-chain variable region (V beta) sequences but, in contrast to unconjugated SEA, mediate the TCR interaction in a MHC class II-independent manner. The cytotoxic activity mediated by C215-SEA and C242-SEA conjugates was blocked by excess of C215 mAb and C242 mAb, respectively, showing that the specificity in the targeting of mAb-SEA conjugates is defined by the antigen reactivity of the mAb. These results demonstrate that bacterial superantigens may be successfully conjugated to mAb with preserved T cell-activating capacity. The circumvention of MHC class II binding of SEs by conjugation to mAb suggests that such conjugates may find general application as antitumor agents, taking advantage of the extreme T cell-activating potency of superantigens.
Collapse
Affiliation(s)
- M Dohlsten
- Kabi Pharmacia Therapeutics AB, Lund, Sweden
| | | | | | | | | |
Collapse
|
34
|
Jung G, Freimann U, Von Marschall Z, Reisfeld RA, Wilmanns W. Target cell-induced T cell activation with bi- and trispecific antibody fragments. Eur J Immunol 1991; 21:2431-5. [PMID: 1655465 DOI: 10.1002/eji.1830211020] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Previously we proposed a concept for tumor immunotherapy in which two different bispecific antibody conjugates, an anti-target x anti-CD3 and an anti-target x anti-CD28 conjugate, induce the activation of resting human T cells upon binding to the respective tumor target cells. After in vivo application of these reagents, this model of a "target cell-induced T cell activation" envisages the destruction of target cells by in situ activated T cells. Obviously however, for in vivo application, the use of Fc-free antibody fragments is mandatory to prevent binding of the conjugates to Fc receptor-positive cells which would lead to Fc-mediated T cell activation. Here we report a simplification of published procedures for the generation of bispecific Fab-hybrid fragments, univalent for each antigen. We demonstrate that an anti-target x anti-CD3/anti-target x anti-CD28 combination of such hybrids, as well as an identical combination of covalently coupled F(ab')2 fragments, mediate "target cell-induced T cell activation" in an in vitro test system. Thus, these reagents may be capable of inducing an in situ activation of human T cells upon systemic in vivo application according to the concept outlined above. A trispecific conjugate with anti-target, anti-CD3- and anti-CD28 specificity appears to be unsuitable for this purpose because it activates resting T cells in soluble form without requiring immobilization through binding via its anti-target portion.
Collapse
Affiliation(s)
- G Jung
- Medizinische Klinik III, Universität München, FRG
| | | | | | | | | |
Collapse
|
35
|
Tutt A, Greenman J, Stevenson GT, Glennie MJ. Bispecific F(ab'gamma)3 antibody derivatives for redirecting unprimed cytotoxic T cells. Eur J Immunol 1991; 21:1351-8. [PMID: 1828423 DOI: 10.1002/eji.1830210604] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Bispecific F(ab'gamma)3 derivatives have been constructed by selective coupling of monoclonal antibody Fab'gamma fragments, using the cross-linker o-phenylenedimaleimide attached to hinge-region sulfhydryl groups. All the derivatives were designed to redirect the cytotoxic activity of circulating human T lymphocytes, via their CD2, CD3 or CD5 molecules, against 51Cr-labeled chicken red blood cells (CRBC) or the human lymphoblastoid cell line, Namalwa. The composition of F(ab'gamma)3 molecules was controlled by the selection of Fab'gamma for initial attachment of cross-linker, allowing production of derivatives with either two anti-effector and one anti-target Fab'gamma arms or vice versa. Bispecific F(ab'gamma)3 derivatives, recruiting peripheral blood lymphocytes (PBL) via CD3 as effectors against CRBC, gave titers in redirected cellular cytotoxicity assays which were consistently 25-100 times higher than those given by an equivalent F(ab'gamma)2 reagent, with full activity being retained down to antibody concentrations of 1 ng/ml. The allocation of valencies--two anti-target/one anti-effector or the converse--was not important in determining the increase in titer. No significant lysis was seen when recruiting PBL via CD2 or CD5. It was notable that these CD3-specific derivatives yielded good tumor-cell lysis when using fresh, unprimed PBL as effectors. The increased activity over a bispecific F(ab'gamma)2 was apparent both in titer and the maximum level of lysis achieved. Blocking studies with excess Fab'gamma suggested that the potency of F(ab'gamma)3 derivatives lie in the strength of bridging between effector and target cells. This high avidity contact could favor metabolic activation in either the target or effector cell, or could increase the efficiency of lytic machinery already triggered.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Neoplasm
- CD3 Complex
- Cytotoxicity, Immunologic
- Glycoproteins/immunology
- Immunoglobulin Fab Fragments/immunology
- Immunoglobulin G/immunology
- Mice
- Mice, Inbred BALB C
- Receptors, Antigen, T-Cell/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Tetraspanins
Collapse
Affiliation(s)
- A Tutt
- Tenovus Research Laboratory, General Hospital, Southampton, GB
| | | | | | | |
Collapse
|
36
|
Unconjugated Monoclonal Antibodies as Anticancer Agents. Immunol Allergy Clin North Am 1991. [DOI: 10.1016/s0889-8561(22)00323-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
37
|
Mansfield PF, Rosenblum MG, Murray JL, Itoh K. Augmentation of interleukin-2-induced activation of human melanoma tumor-infiltrating lymphocytes by heteroconjugate antibody. Cancer Immunol Immunother 1991; 33:247-54. [PMID: 1829394 PMCID: PMC11038763 DOI: 10.1007/bf01744944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/1991] [Accepted: 03/06/1991] [Indexed: 12/29/2022]
Abstract
Heteroconjugate (HC) antibody (anti-CD3 mAb x anti-p97 melanoma mAb) or monomeric anti-CD3 mAb by itself did not induce proliferation of uncultured melanoma tumor-infiltrating lymphocytes (TILs). They also failed to induce IL-2 production in uncultured TILs, although anti-CD3 mAb, but not HC antibody, stimulated IL-2 production in peripheral blood mononuclear cells (PBMCs). Sequential treatment of uncultured TILs from p97-antigen-positive (p97+) melanomas with HC antibody, followed by washing and incubation with interleukin-2 (IL-2), induced significantly higher proliferation than incubation with IL-2 alone. HC antibody pretreatment led to significantly greater results than with anti-CD3 mAb at a 1 ng/ml level in IL-2-induced proliferation of TILs from p97+ melanomas, similar to those with anti-CD3 mAb at a level of 100 ng/ml. HC antibody (1 ng/ml) pretreatment did not enhance IL-2-induced proliferation of either TILs from p97- melanomas or PBMCs, while anti-CD3 mAb enhanced the proliferation of TILs from some p97- melanomas and PBMCs. Regardless of the pretreatment of uncultured TILs with HC antibody or anti-CD3 mAb, IL-2-activated TILs were cytotoxic primarily only to autologous tumor cells, and their phenotypes remained the same. Thus, HC antibody can augment IL-2-induced activation of TILs only from p97+ melanomas, without altering their pattern of cytotoxicity or phenotype. The findings were consistent with observations at the clonal level. In contrast to anti-CD3 mAb, HC pretreatment of uncultured TILs from only p97+ melanoma prior to limiting-dilution analysis increased the number of proliferating TIL clones, including autologous tumor-specific cytotoxic T lymphocyte clones. These results suggest that use of HC antibody in vivo would be more advantageous than anti-CD3 mAb, with regard to augmentation of IL-2-induced TIL activation.
Collapse
Affiliation(s)
- P F Mansfield
- Department of General Surgery, University of Texas, M. D. Anderson Cancer Center, Houston 77030
| | | | | | | |
Collapse
|
38
|
Möller SA, Reisfeld RA. Bispecific-monoclonal-antibody-directed lysis of ovarian carcinoma cells by activated human T lymphocytes. Cancer Immunol Immunother 1991; 33:210-6. [PMID: 1647871 PMCID: PMC11038555 DOI: 10.1007/bf01744939] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/1990] [Accepted: 02/15/1991] [Indexed: 12/28/2022]
Abstract
Two different bispecific hybrid antibodies were established by fusing a hybridoma producing monoclonal antibody (mAb) against the pancarcinoma antigen KS1/4 with either of the two hybridomas OKT3 and 9.3, secreting antibodies reactive with the T cell determinants CD3 and CD28, respectively. The KS1/4 antibody reacts with a 40-kDa cell-surface glycoprotein antigen that is expressed on the surface of a variety of adenocarcinoma cells, including ovarian carcinoma. The ability of the bispecific antibodies 9.3 x KS1/4 and OKT3 x KS1/4 to direct peripheral blood mononuclear cells (PBMC) specifically against OVCAR-3 ovarian carcinoma target cells was measured in a 4-h 51Cr-release assay. The bispecific antibodies were four to six times more potent in killing the OVCAR-3 target cells when compared to their parental antibodies either alone or in combination. A dose-dependent response was observed in the 10-10,000 ng/ml range. The specificity of the targeting was demonstrated by the complete inhibition of cytotoxic activity following pre-incubation of tumor target cells with the parental mAb and by the lack of killing of KS1/4-negative target cell lines. An evaluation of the efficacy of PBMC from ovarian cancer patients as effector cells revealed that their specific cytotoxicity against OVCAR-3 cells was enhanced severalfold by bispecific antibodies as compared to parental antibodies. Furthermore, stimulation of PBMC with immobilized CD3 and interleukin-2 for 4 days resulted in an enhanced directed killing of human ovarian carcinoma cells by human T effector cells and the bispecific antibodies.
Collapse
Affiliation(s)
- S A Möller
- Department of Immunology, Scripps Clinic and Research Foundation, La Jolla, CA 92037
| | | |
Collapse
|
39
|
Abstract
Human proteins with identified effects on host responses to malignant cells have been established as effective therapeutic techniques in cancer. Lymphokines, products of activated cells of the immune system, have pleiotropic biochemical and cellular effects. These include stimulation of immune effector cell proliferation, augmentation of cytotoxicity of immune effector cells for tumor cell targets, enhancement in antigen-recognition potential by monocytes, and modulation of tumor-associated antigen expression on neoplastic cells. Interferons (IFN) and interleukin-2 (IL-2), purified to homogeneity, can induce regression of metastatic malignancy. Recombinant DNA technology has facilitated large-scale production of these and other lymphokines and cytokines. It has also made possible analyses of physical structures of the molecules themselves and has enabled creation of mutated molecules with specific, desired substitutions in their amino acid sequence. Monoclonal antibodies, directed at tumor-associated antigens, can augment antibody-dependent cell-mediated cytotoxicity and can selectively deliver cytotoxic techniques to malignant cells. Molecules that modify the host resistance to malignant disease also have potential to augment effectiveness of other cancer treatment techniques. Lymphokines, cytokines, and monoclonal antibodies, all products of biotechnology, have resulted in fulfillment of the promise of the immune system for inhibition of growth of human malignancy.
Collapse
Affiliation(s)
- E C Borden
- Department of Human Oncology, University of Wisconsin Clinical Cancer Center, Madison 53792
| | | |
Collapse
|
40
|
Van Dijk J, Tsuruo T, Segal DM, Bolhuis RL, Colognola R, van de Griend RJ, Fleuren GJ, Warnaar SO. Bispecific antibodies reactive with the multidrug-resistance-related glycoprotein and CD3 induce lysis of multidrug-resistant tumor cells. Int J Cancer 1989; 44:738-43. [PMID: 2793246 DOI: 10.1002/ijc.2910440431] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We describe the lysis of multidrug-resistant (MDR) tumor cells by various lymphocytic effector cells, retargeted with bispecific antibodies (heteroconjugates). The Ab-heteroconjugate used was prepared by chemically cross-linking the OKT3 monoclonal antibody (MAb) reactive with CD3 antigen on T lymphocytes, with the MRK16 MAb, which recognizes the MDR-associated P-glycoprotein. Cloned TCR alpha beta/CD3+ T lymphocytes, OKT3-activated peripheral-blood mononuclear cells and peripheral-blood mononuclear blood lymphocytes, stimulated with allogeneic irradiated cells in a mixed lymphocyte culture, could be induced to lyse MDR ovarian tumor cells in the presence of Ab-heteroconjugate CD3/MRK16, whereas the drug-sensitive parental tumor cells lacking the P-glycoprotein were not lysed by these retargeted effector cells. Cloned TCR gamma delta/CD3+ T lymphocytes showed a high MHC-unrestricted lysis of MDR tumor cells. Addition of Ab-heteroconjugate CD3/MRK16 could therefore not enhance target-cell lysis. Melanoma tumor cells transfected with the mdr-I gene which codes for the P-glycoprotein were also efficiently lysed by Ab-heteroconjugate retargeted cloned TCR alpha beta/CD3+ T cells. Tumor cell lines derived from organs known to express the P-glycoprotein also were lysable by the retargeted effector cells.
Collapse
Affiliation(s)
- J Van Dijk
- Department of Pathology, State University Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Nitta T, Yagita H, Azuma T, Sato K, Okumura K. Bispecific F (ab')2 monomer prepared with anti-CD3 and anti-tumor monoclonal antibodies is most potent in induction of cytolysis of human T cells. Eur J Immunol 1989; 19:1437-41. [PMID: 2528460 DOI: 10.1002/eji.1830190814] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Induction of cytolytic activity of peripheral blood mononuclear cells towards target cells was studied by preparing bispecific F(ab')2 which was composed of two Fab' fragments, one of which was derived from anti-CD3 monoclonal antibody and the other from anti-tumor monoclonal antibody. After reduction of the interchange disulfide bonds of these fragments by dithiothreitol, a thiol-disulfide interchain reagent, 5,5'-dithiobis-2-nitrobenzoic acid, was added to convert the free SH groups of one of the Fab' fragments to mixed disulfide derivatives. These were then coupled with the other Fab' fragment bearing free SH groups, producing a bispecific hybrid F(ab')2 monomer of high yield. The bispecific F(ab')2 monomers were able to render nonactivated peripheral blood mononuclear cells cytotoxic against natural killer-resistant tumor cell lines at doses as low as 1 microgram/ml. The polymeric forms of the F(ab')2 fragments prepared by use of cross-linking reagents such as N-succinimidyl-3-(2-pyridyldithiol)-propionate (SPDP) or S-acetylmercaptosuccinic acid anhydride (SAMSA) were less efficient for induction of cytolytic activity than the monomeric one. It may be feasible to use bispecific F(ab')2 monomers in cancer immunotherapy because of the ease of preparation, as well as the efficiency in inducing cytolytic activity and their high tissue permeability due to their small size (100-110 kDa). In addition, redirected cytolytic activity towards peripheral blood mononuclear cells by reticuloendothelial system cells, resulting from linking these two types of cells through Fc-Fc receptor interactions, does not occur.
Collapse
Affiliation(s)
- T Nitta
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | |
Collapse
|
42
|
van Dijk J, Warnaar SO, van Eendenburg JD, Thienpont M, Braakman E, Boot JH, Fleuren GJ, Bolhuis RL. Induction of tumor-cell lysis by bi-specific monoclonal antibodies recognizing renal-cell carcinoma and CD3 antigen. Int J Cancer 1989; 43:344-9. [PMID: 2521843 DOI: 10.1002/ijc.2910430230] [Citation(s) in RCA: 59] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Bi-specific monoclonal antibodies (MAbs) were developed by somatic hybridization of 2 mouse hybridomas, one producing MAb against the G250 renal-cell carcinoma (RCC)-associated antigen and the other against the T-cell antigen CD3 (OKT3). The dual specificity of the hybrid MAb produced by these so-called quadromas was analyzed by immunohistochemistry on tissue sections and by cytotoxicity assays with relevant target and effector cells. The bi-specific MAb could induce TCR alpha beta/CD3+ and TCR gamma delta/CD3+ cloned lymphocytes to kill RCC cells. A noteworthy finding was that the TCR alpha beta and gamma delta lymphocyte clones showed different triggering abilities. The specificity of target-cell lysis by the cytotoxic T cells (CTL) was dictated by the specificity of the G250 MAb. Control bi-specific MAb, recognizing a cell-surface structure not involved in T-cell activation, did not induce lysis. Several IgG subclass switch variants of the G250 hybridoma, i.e., IgG1, 2a, 2b and IgE, were used for somatic hybridization with the OKT3 hybridoma (IgG2a). Except for IgE, all IgG subclass combinations could equally induce cytolysis. Induction of cytolysis was inhibited only by excess OKT3 MAb. Comparison of 2 bi-specific MAb preparations of the same combination (IgG2a/1), produced by 2 quadromas derived from the same parental hybridomas after identical purification procedures, produced different amounts of bispecific MAb.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Monoclonal/therapeutic use
- Antibody Specificity
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Neoplasm/immunology
- CD3 Complex
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/therapy
- Humans
- Kidney Neoplasms/immunology
- Kidney Neoplasms/therapy
- Mice
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- J van Dijk
- Department of Pathology, State University Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Pupa SM, Canevari S, Fontanelli R, Ménard S, Mezzanzanica D, Lanzavecchia A, Colnaghi MI. Activation of mononuclear cells to be used for hybrid monoclonal antibody-induced lysis of human ovarian carcinoma cells. Int J Cancer 1988; 42:455-9. [PMID: 2971021 DOI: 10.1002/ijc.2910420324] [Citation(s) in RCA: 34] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Recently we reported that cytotoxic T-cell clones can be retargeted to unrelated tumor cells by bispecific monoclonal antibodies (MAbs), anti-CD3 and anti-ovarian carcinoma (alpha OC/TR) (Mezzanzanica et al., 1988). In the perspective of in vivo tumor immunotherapy, as an alternative to cytotoxic T lymphocytes (CTL) from T-cell clones, since human peripheral blood mononuclear cells (PBMCs) without stimulation were quite ineffective, a suitable in vitro activation method was developed to render PBMCs lytic for relevant targets in the presence of the bispecific hybrid MAb alpha OC/TR. This activation protocol was applied to PBMCs from 9 healthy donors (HD) and 6 ovarian carcinoma patients (P) and to tumor-associated lymphocytes (TAL) from 4 ovarian carcinoma P. The method consisted of in vitro stimulation with phytohemagglutinin (PHA) for 2 days, followed by culture with a low dose of recombinant human interleukin-2 (rIL-2) for 6 to 10 days. The antibody-mediated lysis of CTL from HD PBMCs was found to be specifically directed against cells expressing the relevant ovarian tumor antigen when different tumor cell lines and short-term cultures of tumor and normal cells were tested. The antibody-mediated lysis of CTL from P PBMCs or TAL was efficient both on autologous and allogeneic ovarian tumor cells, whereas no reactivity with autologous normal cells was observed and LAK activity was only evident in 1 out of 4 cases. The hybrid antibody induced cytotoxic activity of CTL from P was, however, lower than that of CTL from HD.
Collapse
Affiliation(s)
- S M Pupa
- Division of Experimental Oncology E, Istituto Nazionale Tumori, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
44
|
Liu MA, Nussbaum SR, Eisen HN. Hormone conjugated with antibody to CD3 mediates cytotoxic T cell lysis of human melanoma cells. Science 1988; 239:395-8. [PMID: 3257303 DOI: 10.1126/science.3257303] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cytotoxic T lymphocytes can be activated by antibodies to their antigen-specific receptor complex (TCR-CD3) to destroy target cells, regardless of the specificity of the cytotoxic T cells. A novel hormone-antibody conjugate, consisting of an analog of melanocyte-stimulating hormone chemically coupled to a monoclonal antibody to CD3, the invariant component of the T cell receptor complex, was used to target human melanoma cells for destruction by human cytotoxic T lymphocytes that bear no specificity for the tumor cells. As targeting components of such anti-CD3 conjugates, hormones or growth factors are expected to prove more effective than antibodies to tumor-associated antigens in focusing the destructive activity of cytotoxic T cells on tumor target cells.
Collapse
MESH Headings
- Antibodies, Monoclonal/therapeutic use
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Neoplasm/immunology
- Cytotoxicity, Immunologic
- Humans
- Lymphocyte Activation
- Melanocyte-Stimulating Hormones/analogs & derivatives
- Melanocyte-Stimulating Hormones/metabolism
- Melanocyte-Stimulating Hormones/therapeutic use
- Melanoma/therapy
- Receptors, Antigen, T-Cell/immunology
- Receptors, Fc/immunology
- Receptors, Pituitary Hormone/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- M A Liu
- Center for Cancer Research, Massachusetts Institute of Technology, Cambridge 02139
| | | | | |
Collapse
|