1
|
Nelson AD, Wang L, Laffey KG, Becher LRE, Parks CA, Hoffmann MM, Galeano BK, Mangalam A, Teixeiro E, White TA, Schrum AG, Cannon JF, Gil D. Rigid crosslinking of the CD3 complex leads to superior T cell stimulation. Front Immunol 2024; 15:1434463. [PMID: 39281668 PMCID: PMC11392757 DOI: 10.3389/fimmu.2024.1434463] [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: 05/17/2024] [Accepted: 08/07/2024] [Indexed: 09/18/2024] Open
Abstract
Functionally bivalent non-covalent Fab dimers (Bi-Fabs) specific for the TCR/CD3 complex promote CD3 signaling on T cells. While comparing functional responses to stimulation with Bi-Fab, F(ab')2 or mAb specific for the same CD3 epitope, we observed fratricide requiring anti-CD3 bridging of adjacent T cells. Surprisingly, anti-CD3 Bi-Fab ranked first in fratricide potency, followed by anti-CD3 F(ab')2 and anti-CD3 mAb. Low resolution structural studies revealed anti-CD3 Bi-Fabs and F(ab')2 adopt similar global shapes with CD3-binding sites oriented outward. However, under molecular dynamic simulations, anti-CD3 Bi-Fabs crosslinked CD3 more rigidly than F(ab')2. Furthermore, molecular modelling of Bi-Fab and F(ab')2 binding to CD3 predicted crosslinking of T cell antigen receptors located in opposing plasma membrane domains, a feature fitting with T cell fratricide observed. Thus, increasing rigidity of Fab-CD3 crosslinking between opposing effector-target pairs may result in stronger T cell effector function. These findings could guide improving clinical performance of bi-specific anti-CD3 drugs.
Collapse
Affiliation(s)
- Alfreda D Nelson
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Liangyu Wang
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
| | - Kimberly G Laffey
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
| | - Laura R E Becher
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Christopher A Parks
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Michele M Hoffmann
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Belinda K Galeano
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Ashutosh Mangalam
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Emma Teixeiro
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
| | - Tommi A White
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Adam G Schrum
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
- Department of Biomedical, Biological and Medical Engineering, College of Engineering, University of Missouri, Columbia, MO, United States
| | - John F Cannon
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
| | - Diana Gil
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
- Department of Biomedical, Biological and Medical Engineering, College of Engineering, University of Missouri, Columbia, MO, United States
| |
Collapse
|
2
|
Peptide-HLA-based immunotherapeutics platforms for direct modulation of antigen-specific T cells. Sci Rep 2021; 11:19220. [PMID: 34584159 PMCID: PMC8479091 DOI: 10.1038/s41598-021-98716-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/06/2021] [Indexed: 11/20/2022] Open
Abstract
Targeted pharmacologic activation of antigen-specific (AgS) T cells may bypass limitations inherent in current T cell-based cancer therapies. We describe two immunotherapeutics platforms for selective delivery of costimulatory ligands and peptide-HLA (pHLA) to AgS T cells. We engineered and deployed on these platforms an affinity-attenuated variant of interleukin-2, which selectively expands oligoclonal and polyfunctional AgS T cells in vitro and synergizes with CD80 signals for superior proliferation versus peptide stimulation.
Collapse
|
3
|
Rath JA, Arber C. Engineering Strategies to Enhance TCR-Based Adoptive T Cell Therapy. Cells 2020; 9:E1485. [PMID: 32570906 PMCID: PMC7349724 DOI: 10.3390/cells9061485] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/13/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
T cell receptor (TCR)-based adoptive T cell therapies (ACT) hold great promise for the treatment of cancer, as TCRs can cover a broad range of target antigens. Here we summarize basic, translational and clinical results that provide insight into the challenges and opportunities of TCR-based ACT. We review the characteristics of target antigens and conventional αβ-TCRs, and provide a summary of published clinical trials with TCR-transgenic T cell therapies. We discuss how synthetic biology and innovative engineering strategies are poised to provide solutions for overcoming current limitations, that include functional avidity, MHC restriction, and most importantly, the tumor microenvironment. We also highlight the impact of precision genome editing on the next iteration of TCR-transgenic T cell therapies, and the discovery of novel immune engineering targets. We are convinced that some of these innovations will enable the field to move TCR gene therapy to the next level.
Collapse
MESH Headings
- Biomedical Engineering
- Cell Engineering
- Cell- and Tissue-Based Therapy/adverse effects
- Cell- and Tissue-Based Therapy/methods
- Cell- and Tissue-Based Therapy/trends
- Gene Editing
- Genetic Therapy
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Lymphocyte Activation
- Molecular Targeted Therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Safety
- Synthetic Biology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Translational Research, Biomedical
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
| | - Caroline Arber
- Department of oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, 1015 Lausanne, Switzerland;
| |
Collapse
|
4
|
Tjandra KC, Thordarson P. Multivalency in Drug Delivery–When Is It Too Much of a Good Thing? Bioconjug Chem 2019; 30:503-514. [DOI: 10.1021/acs.bioconjchem.8b00804] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Kristel C. Tjandra
- School of Chemistry, the Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Pall Thordarson
- School of Chemistry, the Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
5
|
Delcassian D, Sattler S, Dunlop IE. T cell immunoengineering with advanced biomaterials. Integr Biol (Camb) 2017; 9:211-222. [PMID: 28252135 PMCID: PMC6034443 DOI: 10.1039/c6ib00233a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 02/15/2017] [Indexed: 12/25/2022]
Abstract
Recent advances in biomaterials design offer the potential to actively control immune cell activation and behaviour. Many human diseases, such as infections, cancer, and autoimmune disorders, are partly mediated by inappropriate or insufficient activation of the immune system. T cells play a central role in the host immune response to these diseases, and so constitute a promising cell type for manipulation. In vivo, T cells are stimulated by antigen presenting cells (APC), therefore to design immunoengineering biomaterials that control T cell behaviour, artificial interfaces that mimic the natural APC-T cell interaction are required. This review draws together research in the design and fabrication of such biomaterial interfaces, and highlights efforts to elucidate key parameters in T cell activation, such as substrate mechanical properties and spatial organization of receptors, illustrating how they can be manipulated by bioengineering approaches to alter T cell function.
Collapse
Affiliation(s)
- Derfogail Delcassian
- School of Pharmacy, University of Nottingham, NG7 2RD, UK. and Koch Institute for Integrative Cancer Research, MIT, Massachusetts, 02139, USA
| | - Susanne Sattler
- Imperial College London National Heart and Lung Institute, Du Cane Road, W12 0NN, London, UK
| | - Iain E Dunlop
- Department of Materials, Imperial College London, SW7 2AZ, UK.
| |
Collapse
|
6
|
Hoffmann MM, Molina-Mendiola C, Nelson AD, Parks CA, Reyes EE, Hansen MJ, Rajagopalan G, Pease LR, Schrum AG, Gil D. Co-potentiation of antigen recognition: A mechanism to boost weak T cell responses and provide immunotherapy in vivo. SCIENCE ADVANCES 2015; 1:e1500415. [PMID: 26601285 PMCID: PMC4646799 DOI: 10.1126/sciadv.1500415] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/24/2015] [Indexed: 06/05/2023]
Abstract
Adaptive immunity is mediated by antigen receptors that can induce weak or strong immune responses depending on the nature of the antigen that is bound. In T lymphocytes, antigen recognition triggers signal transduction by clustering T cell receptor (TCR)/CD3 multiprotein complexes. In addition, it hypothesized that biophysical changes induced in TCR/CD3 that accompany receptor engagement may contribute to signal intensity. Nonclustering monovalent TCR/CD3 engagement is functionally inert despite the fact that it may induce changes in conformational arrangement or in the flexibility of receptor subunits. We report that the intrinsically inert monovalent engagement of TCR/CD3 can specifically enhance physiologic T cell responses to weak antigens in vitro and in vivo without stimulating antigen-unengaged T cells and without interrupting T cell responses to strong antigens, an effect that we term as "co-potentiation." We identified Mono-7D6-Fab, which biophysically altered TCR/CD3 when bound and functionally enhanced immune reactivity to several weak antigens in vitro, including a gp100-derived peptide associated with melanoma. In vivo, Mono-7D6-Fab induced T cell antigen-dependent therapeutic responses against melanoma lung metastases, an effect that synergized with other anti-melanoma immunotherapies to significantly improve outcome and survival. We conclude that Mono-7D6-Fab directly co-potentiated TCR/CD3 engagement by weak antigens and that such concept can be translated into an immunotherapeutic design. The co-potentiation principle may be applicable to other receptors that could be regulated by otherwise inert compounds whose latent potency is only invoked in concert with specific physiologic ligands.
Collapse
Affiliation(s)
- Michele M. Hoffmann
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | - Carlos Molina-Mendiola
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
- Department of Statistics, Polytechnic University of Catalonia, Barcelona 08034, Spain
| | - Alfreda D. Nelson
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | - Christopher A. Parks
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | - Edwin E. Reyes
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | - Michael J. Hansen
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | - Govindarajan Rajagopalan
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | - Larry R. Pease
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | - Adam G. Schrum
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | - Diana Gil
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
7
|
Castro M, van Santen HM, Férez M, Alarcón B, Lythe G, Molina-París C. Receptor Pre-Clustering and T cell Responses: Insights into Molecular Mechanisms. Front Immunol 2014; 5:132. [PMID: 24817867 PMCID: PMC4012210 DOI: 10.3389/fimmu.2014.00132] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 03/15/2014] [Indexed: 11/13/2022] Open
Abstract
T cell activation, initiated by T cell receptor (TCR) mediated recognition of pathogen-derived peptides presented by major histocompatibility complex class I or II molecules (pMHC), shows exquisite specificity and sensitivity, even though the TCR-pMHC binding interaction is of low affinity. Recent experimental work suggests that TCR pre-clustering may be a mechanism via which T cells can achieve such high sensitivity. The unresolved stoichiometry of the TCR makes TCR-pMHC binding and TCR triggering, an open question. We formulate a mathematical model to characterize the pre-clustering of T cell receptors (TCRs) on the surface of T cells, motivated by the experimentally observed distribution of TCR clusters on the surface of naive and memory T cells. We extend a recently introduced stochastic criterion to compute the timescales of T cell responses, assuming that ligand-induced cross-linked TCR is the minimum signaling unit. We derive an approximate formula for the mean time to signal initiation. Our results show that pre-clustering reduces the mean activation time. However, additional mechanisms favoring the existence of clusters are required to explain the difference between naive and memory T cell responses. We discuss the biological implications of our results, and both the compatibility and complementarity of our approach with other existing mathematical models.
Collapse
Affiliation(s)
- Mario Castro
- Grupo de Dinámica No-Lineal and Grupo Interdisciplinar de Sistemas Complejos (GISC), Escuela Técnica Superior de Ingeniería (ICAI), Universidad Pontificia Comillas , Madrid , Spain
| | - Hisse M van Santen
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid , Madrid , Spain
| | - María Férez
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid , Madrid , Spain
| | - Balbino Alarcón
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid , Madrid , Spain
| | - Grant Lythe
- Department of Applied Mathematics, School of Mathematics, University of Leeds , Leeds , UK
| | - Carmen Molina-París
- Department of Applied Mathematics, School of Mathematics, University of Leeds , Leeds , UK
| |
Collapse
|
8
|
Corgnac S, Perret R, Derré L, Zhang L, Stirnemann K, Zauderer M, Speiser DE, Mach JP, Romero P, Donda A. CD1d-antibody fusion proteins target iNKT cells to the tumor and trigger long-term therapeutic responses. Cancer Immunol Immunother 2012; 62:747-60. [PMID: 23242316 PMCID: PMC3624007 DOI: 10.1007/s00262-012-1381-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 11/22/2012] [Indexed: 12/22/2022]
Abstract
Despite the well-established antitumor activity of CD1d-restricted invariant natural killer T lymphocytes (iNKT), their use for cancer therapy has remained challenging. This appears to be due to their strong but short-lived activation followed by long-term anergy after a single administration of the CD1d agonist ligand alpha-galactosylceramide (αGC). As a promising alternative, we obtained sustained mouse iNKT cell responses associated with prolonged antitumor effects through repeated administrations of tumor-targeted recombinant sCD1d-antitumor scFv fusion proteins loaded with αGC. Here, we demonstrate that CD1d fusion proteins bound to tumor cells via the antibody fragment specific for a tumor-associated antigen, efficiently activate human iNKT cell lines leading to potent tumor cell lysis. The importance of CD1d tumor targeting was confirmed in tumor-bearing mice in which only the specific tumor-targeted CD1d fusion protein resulted in tumor inhibition of well-established aggressive tumor grafts. The therapeutic efficacy correlated with the repeated activation of iNKT and natural killer cells marked by their release of TH1 cytokines, despite the up-regulation of the co-inhibitory receptor PD-1. Our results demonstrate the superiority of providing the superagonist αGC loaded on recombinant CD1d proteins and support the use of αGC/sCD1d-antitumor fusion proteins to secure a sustained human and mouse iNKT cell activation, while targeting their cytotoxic activity and cytokine release to the tumor site.
Collapse
Affiliation(s)
- Stéphanie Corgnac
- Ludwig Center for Cancer Research, University of Lausanne, 1066, Epalinges, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Huang J, Meyer C, Zhu C. T cell antigen recognition at the cell membrane. Mol Immunol 2012; 52:155-64. [PMID: 22683645 DOI: 10.1016/j.molimm.2012.05.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 04/27/2012] [Accepted: 05/01/2012] [Indexed: 11/16/2022]
Abstract
T cell antigen receptors (TCRs) on the surface of T cells bind specifically to particular peptide bound major histocompatibility complexes (pMHCs) presented on the surface of antigen presenting cells (APCs). This interaction is a key event in T cell antigen recognition and activation. Most studies have used surface plasmon resonance (SPR) to measure the in vitro binding kinetics of TCR-pMHC interactions in solution using purified proteins. However, these measurements are not physiologically precise, as both TCRs and pMHCs are membrane-associated molecules which are regulated by their cellular environments. Recently, single-molecule förster resonance energy transfer (FRET) and single-molecule mechanical assays were used to measure the in situ binding kinetics of TCR-pMHC interactions on the surface of live T cells. These studies have provided exciting insights into the biochemical basis of T cell antigen recognition and suggest that TCRs serially engage with a small number of antigens with very fast kinetics in order to maximize TCR signaling and sensitivity.
Collapse
Affiliation(s)
- Jun Huang
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
10
|
Boyle S, Kolin DL, Bieler JG, Schneck JP, Wiseman PW, Edidin M. Quantum dot fluorescence characterizes the nanoscale organization of T cell receptors for antigen. Biophys J 2012; 101:L57-9. [PMID: 22261075 DOI: 10.1016/j.bpj.2011.10.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 09/19/2011] [Accepted: 10/13/2011] [Indexed: 11/20/2022] Open
Abstract
Changes in the clustering of surface receptors modulate cell responses to ligands. Hence, global measures of receptor clustering can be useful for characterizing cell states. Using T cell receptor for antigen as an example, we show that k-space image correlation spectroscopy of quantum dots blinking detects T cell receptor clusters on a scale of tens of nanometers and reports changes in clustering after T cell activation. Our results offer a general approach to the global analysis of lateral organization and receptor clustering in single cells, and can thus be applied when the cell type of interest is rare.
Collapse
Affiliation(s)
- Sarah Boyle
- Biology Department, The Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|
11
|
Shen ZT, Brehm MA, Daniels KA, Sigalov AB, Selin LK, Welsh RM, Stern LJ. Bi-specific MHC heterodimers for characterization of cross-reactive T cells. J Biol Chem 2010; 285:33144-33153. [PMID: 20729210 PMCID: PMC2963422 DOI: 10.1074/jbc.m110.141051] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 07/25/2010] [Indexed: 11/06/2022] Open
Abstract
T cell cross-reactivity describes the phenomenon whereby a single T cell can recognize two or more different peptide antigens presented in complex with MHC proteins. Cross-reactive T cells have previously been characterized at the population level by cytokine secretion and MHC tetramer staining assays, but single-cell analysis is difficult or impossible using these methods. In this study, we describe development of a novel peptide-MHC heterodimer specific for cross-reactive T cells. MHC-peptide monomers were independently conjugated to hydrazide or aldehyde-containing cross-linkers using thiol-maleimide coupling at cysteine residues introduced into recombinant MHC heavy chain proteins. Hydrazone formation provided bi-specific MHC heterodimers carrying two different peptides. Using this approach we prepared heterodimers of the murine class I MHC protein H-2K(b) carrying peptides from lymphocytic choriomeningitis virus and vaccinia virus, and used these to identify cross-reactive CD8+ T cells recognizing both lymphocytic choriomeningitis virus and vaccinia virus antigens. A similar strategy could be used to develop reagents to analyze cross-reactive T cell responses in humans.
Collapse
Affiliation(s)
- Zu T Shen
- From the Department of Pathology, Worcester, Massachusetts 01655
| | | | - Keith A Daniels
- From the Department of Pathology, Worcester, Massachusetts 01655
| | | | - Liisa K Selin
- From the Department of Pathology, Worcester, Massachusetts 01655
| | - Raymond M Welsh
- From the Department of Pathology, Worcester, Massachusetts 01655
| | - Lawrence J Stern
- From the Department of Pathology, Worcester, Massachusetts 01655; Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01655.
| |
Collapse
|
12
|
Valitutti S, Coombs D, Dupré L. The space and time frames of T cell activation at the immunological synapse. FEBS Lett 2010; 584:4851-7. [PMID: 20940018 DOI: 10.1016/j.febslet.2010.10.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 10/06/2010] [Accepted: 10/06/2010] [Indexed: 01/17/2023]
Abstract
The selective recognition of antigenic peptides by T cells requires the spatio/temporal integration of a panoply of molecular triggers. The space frame of T cell antigen receptors (TCR) interaction with peptide/MHC complexes (pMHC) displayed by antigen presenting cells is delineated by the micrometer-scale area of the immunological synapse. The time frame of T cell stimulation is governed by a series of short TCR-pMHC interactions that are integrated into sustained signaling leading to productive activation. We discuss here how approaching antigen recognition from the time and space angles is key to the comprehension of the puzzling process of T cell activation.
Collapse
Affiliation(s)
- Salvatore Valitutti
- INSERM, U563, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France.
| | | | | |
Collapse
|
13
|
Junghans RP. Strategy escalation: an emerging paradigm for safe clinical development of T cell gene therapies. J Transl Med 2010; 8:55. [PMID: 20537174 PMCID: PMC2904270 DOI: 10.1186/1479-5876-8-55] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 06/10/2010] [Indexed: 11/10/2022] Open
Abstract
Gene therapy techniques are being applied to modify T cells with chimeric antigen receptors (CARs) for therapeutic ends. The versatility of this platform has spawned multiple options for their application with new permutations in strategies continually being invented, a testimony to the creative energies of many investigators. The field is rapidly expanding with immense potential for impact against diverse cancers. But this rapid expansion, like the Big Bang, comes with a somewhat chaotic evolution of its therapeutic universe that can also be dangerous, as seen by recently publicized deaths. Time-honored methods for new drug testing embodied in Dose Escalation that were suitable for traditional inert agents are now inadequate for these novel "living drugs". In the following, I propose an approach to escalating risk for patient exposures with these new immuno-gene therapy agents, termed Strategy Escalation, that accounts for the molecular and biological features of the modified cells and the methods of their administration. This proposal is offered not as a prescriptive but as a discussion framework that investigators may wish to consider in configuring their intended clinical applications.
Collapse
Affiliation(s)
- Richard Paul Junghans
- Department of Surgery, Boston University School of Medicine, Roger Williams Medical Center, Providence, RI 02908, USA.
| |
Collapse
|
14
|
Juang J, Ebert PJR, Feng D, Garcia KC, Krogsgaard M, Davis MM. Peptide-MHC heterodimers show that thymic positive selection requires a more restricted set of self-peptides than negative selection. ACTA ACUST UNITED AC 2010; 207:1223-34. [PMID: 20457759 PMCID: PMC2882826 DOI: 10.1084/jem.20092170] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
T cell selection and maturation in the thymus depends on the interactions between T cell receptors (TCRs) and different self-peptide-major histocompatibility complex (pMHC) molecules. We show that the affinity of the OT-I TCR for its endogenous positively selecting ligands, Catnb-H-2Kb and Cappa1-H-2Kb, is significantly lower than for previously reported positively selecting altered peptide ligands. To understand how these extremely weak endogenous ligands produce signals in maturing thymocytes, we generated soluble monomeric and dimeric peptide-H-2Kb ligands. Soluble monomeric ovalbumin (OVA)-Kb molecules elicited no detectable signaling in OT-I thymocytes, whereas heterodimers of OVA-Kb paired with positively selecting or nonselecting endogenous peptides, but not an engineered null peptide, induced deletion. In contrast, dimer-induced positive selection was much more sensitive to the identity of the partner peptide. Catnb-Kb-Catnb-Kb homodimers, but not heterodimers of Catnb-Kb paired with a nonselecting peptide-Kb, induced positive selection, even though both ligands bind the OT-I TCR with detectable affinity. Thus, both positive and negative selection can be driven by dimeric but not monomeric ligands. In addition, positive selection has much more stringent requirements for the partner self-pMHC.
Collapse
Affiliation(s)
- Jeremy Juang
- The Department of Microbiology and Immunology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
15
|
Ebert PJR, Li QJ, Huppa JB, Davis MM. Functional development of the T cell receptor for antigen. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 92:65-100. [PMID: 20800817 PMCID: PMC4887107 DOI: 10.1016/s1877-1173(10)92004-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
For over three decades now, the T cell receptor (TCR) for antigen has not ceased to challenge the imaginations of cellular and molecular immunologists alike. T cell antigen recognition transcends every aspect of adaptive immunity: it shapes the T cell repertoire in the thymus and directs T cell-mediated effector functions in the periphery, where it is also central to the induction of peripheral tolerance. Yet, despite its central position, there remain many questions unresolved: how can one TCR be specific for one particular peptide-major histocompatibility complex (pMHC) ligand while also binding other pMHC ligands with an immunologically relevant affinity? And how can a T cell's extreme specificity (alterations of single methyl groups in their ligand can abrogate a response) and sensitivity (single agonist ligands on a cell surface are sufficient to trigger a measurable response) emerge from TCR-ligand interactions that are so low in affinity? Solving these questions is intimately tied to a fundamental understanding of molecular recognition dynamics within the many different contexts of various T cell-antigen presenting cell (APC) contacts: from the thymic APCs that shape the TCR repertoire and guide functional differentiation of developing T cells to the peripheral APCs that support homeostasis and provoke antigen responses in naïve, effector, memory, and regulatory T cells. Here, we discuss our recent findings relating to T cell antigen recognition and how this leads to the thymic development of foreign-antigen-responsive alphabetaT cells.
Collapse
Affiliation(s)
- Peter J R Ebert
- The Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | | | | | | |
Collapse
|
16
|
Chervin AS, Stone JD, Holler PD, Bai A, Chen J, Eisen HN, Kranz DM. The impact of TCR-binding properties and antigen presentation format on T cell responsiveness. THE JOURNAL OF IMMUNOLOGY 2009; 183:1166-78. [PMID: 19553539 DOI: 10.4049/jimmunol.0900054] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TCR interactions with cognate peptide-MHC (pepMHC) ligands are generally low affinity. This feature, together with the requirement for CD8/CD4 participation, has made it difficult to dissect relationships between TCR-binding parameters and T cell activation. Interpretations are further complicated when comparing different pepMHC, because these can vary greatly in stability. To examine the relationships between TCR-binding properties and T cell responses, in this study we characterized the interactions and activities mediated by a panel of TCRs that differed widely in their binding to the same pepMHC. Monovalent binding of soluble TCR was characterized by surface plasmon resonance, and T cell hybridomas that expressed these TCR, with or without CD8 coexpression, were tested for their binding of monomeric and oligomeric forms of the pepMHC and for subsequent responses (IL-2 release). The binding threshold for eliciting this response in the absence of CD8 (K(D) = 600 nM) exhibited a relatively sharp cutoff between full activity and no activity, consistent with a switchlike response to pepMHC on APCs. However, when the pepMHC was immobilized (plate bound), T cells with the lowest affinity TCRs (e.g., K(D) = 30 microM) responded, even in the absence of CD8, indicating that these TCR are signaling competent. Surprisingly, even cells that expressed high-affinity (K(D) = 16 nM) TCRs along with CD8 were unresponsive to oligomers in solution. The findings suggest that to drive downstream T cell responses, pepMHC must be presented in a form that supports formation of appropriate supramolecular clusters.
Collapse
Affiliation(s)
- Adam S Chervin
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Stone JD, Chervin AS, Kranz DM. T-cell receptor binding affinities and kinetics: impact on T-cell activity and specificity. Immunology 2009; 126:165-76. [PMID: 19125887 DOI: 10.1111/j.1365-2567.2008.03015.x] [Citation(s) in RCA: 271] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The interaction between the T-cell receptor (TCR) and its peptide-major histocompatibility complex (pepMHC) ligand plays a critical role in determining the activity and specificity of the T cell. The binding properties associated with these interactions have now been studied in many systems, providing a framework for a mechanistic understanding of the initial events that govern T-cell function. There have been various other reviews that have described the structural and biochemical features of TCR : pepMHC interactions. Here we provide an overview of four areas that directly impact our understanding of T-cell function, as viewed from the perspective of the TCR : pepMHC interaction: (1) relationships between T-cell activity and TCR : pepMHC binding parameters, (2) TCR affinity, avidity and clustering, (3) influence of coreceptors on pepMHC binding by TCRs and T-cell activity, and (4) impact of TCR binding affinity on antigenic peptide specificity.
Collapse
Affiliation(s)
- Jennifer D Stone
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | | | |
Collapse
|
18
|
Abstract
The interaction between the T cell receptor (TCR) and a peptide-loaded major histocompatibility complex (pMHC) is one of the most-studied interactions in immunology, and yet the precise mechanism by which this system operates is still not fully understood. One key issue is whether TCR triggering minimally requires monomeric pMHC complexes or higher-order multimers (two or more pMHCs). Any model of TCR triggering must explain the high sensitivity, specificity, and dynamic range of ligand responsiveness that this receptor system exhibits. Most models of TCR triggering have not fully appreciated the dynamic aspects of TCR triggering. TCR triggering happens very quickly, and the properties of sensitivity and specificity can be explained by a model that accounts for the interaction dynamics of such a receptor system. In this paper, it is proposed that the important parameter in TCR triggering is the immobilization of the TCR-pMHC complex in the plasma membrane. Whether this involves monomeric or multimeric pMHCs may depend on the affinity of the TCR for the pMHC.
Collapse
Affiliation(s)
- Rajat Varma
- Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 4 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
19
|
Bakker AH, Hoppes R, Linnemann C, Toebes M, Rodenko B, Berkers CR, Hadrup SR, van Esch WJE, Heemskerk MHM, Ovaa H, Schumacher TNM. Conditional MHC class I ligands and peptide exchange technology for the human MHC gene products HLA-A1, -A3, -A11, and -B7. Proc Natl Acad Sci U S A 2008; 105:3825-30. [PMID: 18308940 PMCID: PMC2268811 DOI: 10.1073/pnas.0709717105] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Indexed: 01/28/2023] Open
Abstract
Major histocompatibility complex (MHC) class I multimer technology has become an indispensable immunological assay system to dissect antigen-specific cytotoxic CD8(+) T cell responses by flow cytometry. However, the development of high-throughput assay systems, in which T cell responses against a multitude of epitopes are analyzed, has been precluded by the fact that for each T cell epitope, a separate in vitro MHC refolding reaction is required. We have recently demonstrated that conditional ligands that disintegrate upon exposure to long-wavelength UV light can be designed for the human MHC molecule HLA-A2. To determine whether this peptide-exchange technology can be developed into a generally applicable approach for high throughput MHC based applications we set out to design conditional ligands for the human MHC gene products HLA-A1, -A3, -A11, and -B7. Here, we describe the development and characterization of conditional ligands for this set of human MHC molecules and apply the peptide-exchange technology to identify melanoma-associated peptides that bind to HLA-A3 with high affinity. The conditional ligand technology developed here will allow high-throughput MHC-based analysis of cytotoxic T cell immunity in the vast majority of Western European individuals.
Collapse
Affiliation(s)
| | - Rieuwert Hoppes
- Cellular Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | | | | | - Boris Rodenko
- Cellular Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Celia R. Berkers
- Cellular Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | | | | | - Mirjam H. M. Heemskerk
- Department of Hematology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Huib Ovaa
- Cellular Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | | |
Collapse
|
20
|
Risueño RM, Schamel WWA, Alarcón B. T cell receptor engagement triggers its CD3epsilon and CD3zeta subunits to adopt a compact, locked conformation. PLoS One 2008; 3:e1747. [PMID: 18320063 PMCID: PMC2254190 DOI: 10.1371/journal.pone.0001747] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Accepted: 10/08/2007] [Indexed: 11/24/2022] Open
Abstract
How the T cell antigen receptor (TCR) discriminates between molecularly related peptide/Major Histocompatibility Complex (pMHC) ligands and converts this information into different possible signaling outcomes is still not understood. One current model proposes that strong pMHC ligands, but not weak ones, induce a conformational change in the TCR. Evidence supporting this comes from a pull-down assay that detects ligand-induced binding of the TCR to the N-terminal SH3 domain of the adapter protein Nck, and also from studies with a neoepitope-specific antibody. Both methods rely on the exposure of a polyproline sequence in the CD3ε subunit of the TCR, and neither indicates whether the conformational change is transmitted to other CD3 subunits. Using a protease-sensitivity assay, we now show that the cytoplasmic tails of CD3ε and CD3ζ subunits become fully protected from degradation upon TCR triggering. These results suggest that the TCR conformational change is transmitted to the tails of CD3ε and CD3ζ, and perhaps all CD3 subunits. Furthermore, the resistance to protease digestion suggests that CD3 cytoplasmic tails adopt a compact structure in the triggered TCR. These results are consistent with a model in which transduction of the conformational change induced upon TCR triggering promotes condensation and shielding of the CD3 cytoplasmic tails.
Collapse
Affiliation(s)
- Ruth M. Risueño
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Balbino Alarcón
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
21
|
A permissive geometry model for TCR–CD3 activation. Trends Biochem Sci 2008; 33:51-7. [DOI: 10.1016/j.tibs.2007.10.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Revised: 10/09/2007] [Accepted: 10/19/2007] [Indexed: 01/23/2023]
|
22
|
Abstract
Through T cell receptors (TCRs), T cells can detect and respond to very small numbers of foreign peptides among a huge number of self-peptides presented by major histocompatibility complexes (pMHCs) on the surface of antigen-presenting cells (APCs). How T cells achieve such remarkable sensitivity and specificity through pMHC-TCR binding is an intensively pursued issue in immunology today; the key question is how pMHC-TCR binding initiates, or triggers, a signal from TCRs. Multiple competing models have been proposed, none of which fully explains the sensitivity and specificity of TCR triggering. What has been omitted from existing theories is that the pMHC-TCR interaction at the T cell/APC interface must be under constant mechanical stress, due to the dynamic nature of cell-cell interaction. Taking this condition into consideration, we propose the receptor deformation model of TCR triggering. In this model, TCR signaling is initiated by conformational changes of the TCR/CD3 complex, induced by a pulling force originating from the cytoskeleton and transmitted through pMHC-TCR binding interactions with enough strength to resist rupture. By introducing mechanical force into a model of T cell signal initiation, the receptor deformation model provides potential mechanistic solutions to the sensitivity and specificity of TCR triggering.
Collapse
Affiliation(s)
- Zhengyu Ma
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
| | | | | |
Collapse
|
23
|
Shimizu T, Osaka Y, Banri-Koike C, Yoshida M, Endo K, Furukawa K, Oda M, Murakami A, Ogawa S, Abe R, Azuma T. T cells specific to hapten carrier but not to carrier alone assist in the production of anti-hapten and anti-carrier antibodies. Int Immunol 2007; 19:1157-64. [PMID: 17881502 DOI: 10.1093/intimm/dxm080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We examined the immune response of Balb/c mice to antigens prepared by conjugating 2-phenyloxazolone (phOx) to a foreign protein, ovalbumin (OVA), or a self-protein, mouse serum albumin (MSA), in order to study how these chemical modifications would affect immune recognition. We found that anti-OVA antibodies and CD4(+) T cells produced by OVA immunization reacted with OVA as well as with phOx-OVA. Anti-phOx antibodies were produced by phOx-OVA immunization and, interestingly, T cells from these mice reacted only with phOx-OVA but not with the intact OVA. These results suggested that the classical model of hapten-carrier immunization, in which B cells specific to hapten are activated with assistance from T cells specific to a carrier protein, might not be a major route for production of anti-hapten antibodies in hapten-carrier immunization. Furthermore, phOx-MSA immunization induced production of anti-phOx antibodies, which could not be accounted for in terms of the assistance of carrier-specific T cells because of the absence of MSA-specific T cells. Therefore, we proposed a new model in which anti-hapten B cells are assisted by T cells specific to the haptenated carrier.
Collapse
Affiliation(s)
- Takeyuki Shimizu
- Division of Structural Immunology, Research Institute for Biological Sciences, Tokyo University of Science, Yamazaki 2669, Noda, Chiba 278-0022, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Minguet S, Swamy M, Alarcón B, Luescher IF, Schamel WWA. Full Activation of the T Cell Receptor Requires Both Clustering and Conformational Changes at CD3. Immunity 2007; 26:43-54. [PMID: 17188005 DOI: 10.1016/j.immuni.2006.10.019] [Citation(s) in RCA: 192] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 10/10/2006] [Accepted: 10/27/2006] [Indexed: 11/26/2022]
Abstract
T cell receptor (TCR-CD3) triggering involves both receptor clustering and conformational changes at the cytoplasmic tails of the CD3 subunits. The mechanism by which TCRalphabeta ligand binding confers conformational changes to CD3 is unknown. By using well-defined ligands, we showed that induction of the conformational change requires both multivalent engagement and the mobility restriction of the TCR-CD3 imposed by the plasma membrane. The conformational change is elicited by cooperative rearrangements of two TCR-CD3 complexes and does not require accompanying changes in the structure of the TCRalphabeta ectodomains. This conformational change at CD3 reverts upon ligand dissociation and is required for T cell activation. Thus, our permissive geometry model provides a molecular mechanism that rationalizes how the information of ligand binding to TCRalphabeta is transmitted to the CD3 subunits and to the intracellular signaling machinery.
Collapse
Affiliation(s)
- Susana Minguet
- Max Planck-Institut für Immunbiologie and Faculty of Biology, University of Freiburg, Stübeweg 51, 79108 Freiburg, Germany
| | | | | | | | | |
Collapse
|
25
|
Jones LL, Brophy SE, Bankovich AJ, Colf LA, Hanick NA, Garcia KC, Kranz DM. Engineering and characterization of a stabilized alpha1/alpha2 module of the class I major histocompatibility complex product Ld. J Biol Chem 2006; 281:25734-44. [PMID: 16815841 DOI: 10.1074/jbc.m604343200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The major histocompatibility complex (MHC) is the most polymorphic locus known, with thousands of allelic variants. There is considerable interest in understanding the diversity of structures and peptide-binding features represented by this class of proteins. Although many MHC proteins have been crystallized, others have not been amenable to structural or biochemical studies due to problems with expression or stability. In the present study, yeast display was used to engineer stabilizing mutations into the class I MHC molecule, Ld. The approach was based on previous studies that showed surface levels of yeast-displayed fusion proteins are directly correlated with protein stability. To engineer a more stable Ld, we selected Ld mutants with increased surface expression from randomly mutated yeast display libraries using anti-Ld antibodies or high affinity, soluble T-cell receptors (TCRs). The most stable Ld mutant, Ld-m31, consisted of a single-chain MHC module containing only the alpha1 and alpha2 domains. The enhanced stability was in part due to a single mutation (Trp-97 --> Arg), shown previously to be present in the allele Lq. Mutant Ld-m31 could bind to Ld peptides, and the specific peptide.Ld-m31 complex (QL9.Ld-m31) was recognized by alloreactive TCR 2C. A soluble form of the Ld-m31 protein was expressed in Escherichia coli and refolded from inclusion bodies at high yields. Surface plasmon resonance showed that TCRs bound to peptide.Ld-m31 complexes with affinities similar to those of native full-length Ld. The TCR and QL9.Ld-m31 formed complexes that could be resolved by native gel electrophoresis, suggesting that stabilized alpha1/alpha2 class I platforms may enable various structural studies.
Collapse
Affiliation(s)
- Lindsay L Jones
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | | | | | | | | | | | |
Collapse
|