1
|
Sakamoto-Rablah E, Bye J, Modak A, Hooker A, Uddin S, McManus JJ. Synthetic T-Cell Receptor-like Protein Behaves as a Janus Particle in Solution. J Am Chem Soc 2025; 147:247-256. [PMID: 39699993 PMCID: PMC11726545 DOI: 10.1021/jacs.4c08932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Protein engineering enables the creation of tailor-made proteins for a variety of applications. ImmTACs stand out as promising therapeutics for cancer and other treatments while also presenting unique challenges for stability, formulation, and delivery. We have shown that ImmTACs behave as Janus particles in solution, leading to self-association at low concentrations, even when the average protein-protein interactions suggest that the molecule should be stable. The formation of small but stable oligomers was confirmed by static and dynamic light scattering and analytical ultracentrifugation. Modeling of the structure using AlphaFold leads to a rational explanation for this behavior, consistent with the Janus particle assembly observed for inverse patchy particles.
Collapse
Affiliation(s)
- Emily Sakamoto-Rablah
- HH
Wills Physics Laboratory, University of
Bristol, Tyndall Avenue, Bristol BS8 1TL, U.K.
| | - Jordan Bye
- Immunocore
Limited, 92 Milton Park, Abingdon OX14 4RY, U.K.
| | - Arghya Modak
- Immunocore
Limited, 92 Milton Park, Abingdon OX14 4RY, U.K.
| | - Andrew Hooker
- Immunocore
Limited, 92 Milton Park, Abingdon OX14 4RY, U.K.
| | - Shahid Uddin
- Immunocore
Limited, 92 Milton Park, Abingdon OX14 4RY, U.K.
| | - Jennifer J. McManus
- HH
Wills Physics Laboratory, University of
Bristol, Tyndall Avenue, Bristol BS8 1TL, U.K.
- Bristol
Biodesign Institute, University of Bristol, Bristol BS8 1QU, U.K.
| |
Collapse
|
2
|
Zhang H, Grippin A, Sun M, Ma Y, Kim BYS, Teng L, Jiang W, Yang Z. New avenues for cancer immunotherapy: Cell-mediated drug delivery systems. J Control Release 2024; 375:712-732. [PMID: 39326499 DOI: 10.1016/j.jconrel.2024.09.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/15/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
Cancer research has become increasingly complex over the past few decades as knowledge of the heterogeneity of cancer cells, their proliferative ability, and their tumor microenvironments has become available. Although conventional therapies remain the most compelling option for cancer treatment to date, immunotherapy is a promising way to harness natural immune defenses to target and kill cancer cells. Cell-mediated drug delivery systems (CDDSs) have been an active line of research for enhancing the therapeutic efficacy and specificity of cancer immunotherapy. These systems can be tailored to different types of immune cells, allowing immune evasion and accumulation in the tumor microenvironment. By enabling the targeted delivery of therapeutic agents such as immune stimulants, cytokines, antibodies, and antigens, CDDSs have improved the survival of some patients with cancer. This review summarizes the research status of CDDSs, with a focus on their underlying mechanisms of action, biology, and clinical applications. We also discuss opportunities and challenges for implementation of CDDSs into mainstream cancer immunotherapy.
Collapse
Affiliation(s)
- Huan Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Adam Grippin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Man Sun
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yifan Ma
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
3
|
Robinson RA, McMurran C, McCully ML, Cole DK. Engineering soluble T-cell receptors for therapy. FEBS J 2021; 288:6159-6173. [PMID: 33624424 PMCID: PMC8596704 DOI: 10.1111/febs.15780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/11/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022]
Abstract
Immunotherapy approaches that target peptide-human leukocyte antigen (pHLA) complexes are becoming highly attractive because of their potential to access virtually all foreign and cellular proteins. For this reason, there has been considerable interest in the development of the natural ligand for pHLA, the T-cell receptor (TCR), as a soluble drug to target disease-associated pHLA presented at the cell surface. However, native TCR stability is suboptimal for soluble drug development, and natural TCRs generally have weak affinities for pHLAs, limiting their potential to reach efficacious receptor occupancy levels as soluble drugs. To overcome these limitations and make full use of the TCR as a soluble drug platform, several protein engineering solutions have been applied to TCRs to enhance both their stability and affinity, with a focus on retaining target specificity and selectivity. Here, we review these advances and look to the future for the next generation of soluble TCR-based therapies that can target monomorphic HLA-like proteins presenting both peptide and nonpeptide antigens.
Collapse
|
4
|
McDaid WJ, Lissin N, Pollheimer E, Greene M, Leach A, Smyth P, Bossi G, Longley D, Cole DK, Scott CJ. Enhanced target-specific delivery of docetaxel-loaded nanoparticles using engineered T cell receptors. NANOSCALE 2021; 13:15010-15020. [PMID: 34533174 PMCID: PMC8447836 DOI: 10.1039/d1nr04001d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/18/2021] [Indexed: 06/13/2023]
Abstract
For effective targeted therapy of cancer with chemotherapy-loaded nanoparticles (NPs), antigens that are selective for cancer cells should be targeted to minimise off-tumour toxicity. Human leukocyte antigens (HLAs) are attractive cancer targets as they can present peptides from tumour-selective proteins on the cell surface, which can be recognised by T cells via T cell receptors (TCRs). In this study, docetaxel-loaded polymeric NPs were conjugated to recombinant affinity-enhanced TCRs to target breast cancer cells presenting a tumour-selective peptide-HLA complex. The TCR-conjugated nanoparticles enabled enhanced delivery of docetaxel and induced cell death through tumour-specific peptide-HLA targeting. These in vitro data demonstrate the potential of targeting tumour-restricted peptide-HLA epitopes using high affinity TCR-conjugated nanoparticles, representing a novel treatment strategy to deliver therapeutic drugs specifically to cancer cells.
Collapse
Affiliation(s)
- William J McDaid
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK, BT9 7AE.
- Cancer Research UK Manchester Institute, Alderley Park, Congleton Rd, Alderley Edge, Macclesfield, UK, SK10 4TG
| | - Nikolai Lissin
- Immunocore Ltd, 101 Park Dr, Milton, Abingdon, United Kingdom OX14 4RY
| | - Ellen Pollheimer
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK, BT9 7AE.
| | - Michelle Greene
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK, BT9 7AE.
| | - Adam Leach
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK, BT9 7AE.
- Institute of Cancer Research, 15 Cotswold Rd, Sutton, London, SM2 5NG, UK
| | - Peter Smyth
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK, BT9 7AE.
| | - Giovanna Bossi
- Immunocore Ltd, 101 Park Dr, Milton, Abingdon, United Kingdom OX14 4RY
| | - Daniel Longley
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK, BT9 7AE.
| | - David K Cole
- Immunocore Ltd, 101 Park Dr, Milton, Abingdon, United Kingdom OX14 4RY
| | - Christopher J Scott
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK, BT9 7AE.
| |
Collapse
|
5
|
Myles IA, Zhao M, Nardone G, Olano LR, Reckhow JD, Saleem D, Break TJ, Lionakis MS, Myers TG, Gardina PJ, Kirkpatrick CH, Holland SM, Datta SK. CD8+ T cells produce a dialyzable antigen-specific activator of dendritic cells. J Leukoc Biol 2016; 101:307-320. [PMID: 27515950 DOI: 10.1189/jlb.3a0216-082r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/27/2016] [Accepted: 07/25/2016] [Indexed: 01/17/2023] Open
Abstract
Cellular lysates from PPD+ donors have been reported to transfer tuberculin reactivity to naïve recipients, but not diphtheria reactivity, and vice versa. A historically controversial topic, the terms "transfer factor" and "DLE" were used to characterize the reactivity-transferring properties of lysates. Intrigued by these reported phenomena, we found that the cellular extract derived from antigen-specific memory CD8+ T cells induces IL-6 from antigen-matched APCs. This ultimately elicits IL-17 from bystander memory CD8+ T cells. We have identified that dialyzable peptide sequences, S100a9, and the TCR β chain from CD8+ T cells contribute to the molecular nature of this activity. We further show that extracts from antigen-targeted T cells enhance immunity to Staphylococcus aureus and Candida albicans These effects are sensitive to immunization protocols and extraction methodology in ways that may explain past discrepancies in the reproducibility of passive cellular immunity.
Collapse
Affiliation(s)
- Ian A Myles
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA;
| | - Ming Zhao
- Research Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Glenn Nardone
- Research Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lisa R Olano
- Research Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jensen D Reckhow
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Danial Saleem
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Timothy J Break
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michail S Lionakis
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Timothy G Myers
- Genomic Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA; and
| | - Paul J Gardina
- Genomic Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA; and
| | | | - Steven M Holland
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sandip K Datta
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Abstract
Gene therapy has been proposed as a means to combat cancer. However, systemic toxicity observed in preclinical trials suggested the importance of selectively targeted delivery and inducible gene expression in tumor tissues. Discovery of radiation-inducible promoter sequences provides one way to minimize inadvertent toxicity from gene therapy in normal tissues. Radiation is administered to selectively induce cytotoxic gene expression in the targeted tumor tissues. With promising results from phase II clinical trials using TNF-expressing adenovirus, it is possible to have radiation-guided gene therapy regimes once the tumor-targeted delivery has been achieved. Tumor endothelium is an attractive biological target for gene therapy, because it has the advantage of stability, accessibility, and bioavailability for therapeutic agents. Technological development of DNA microarray, proteomic profiling, and phage-displayed libraries accelerates the identification of tumor-specific endothelial biomarkers and discovery of its relevant affinity reagents for targeted delivery. The application of radiation-guided gene delivery, its amplification, as well as expression of gene therapy presents great opportunities to be employed as an alternative cancer treatment.
Collapse
Affiliation(s)
- Zhaozhong Han
- Department of Radiation Oncology, School of Medicine, Vanderbilt University, 1161 21st Ave. South, Nashville, TN 37232, USA
| | | | | |
Collapse
|
7
|
Smethurst D. A pharmacologic perspective on newly emerging T-cell manipulation technologies. Br J Clin Pharmacol 2013; 76:173-87. [PMID: 23039307 DOI: 10.1111/j.1365-2125.2012.04475.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 09/19/2012] [Indexed: 12/18/2022] Open
Abstract
T cells are a multifaceted family pivotal in the operations of the immune system and many of its associated diseases. The pathway to understanding T cells has been marked by several pharmacological advances including the discoveries of ciclosporin, tacrolimus and the mTOR inhibitors which revolutionized transplant therapy along with providing relief for severe eczema, asthma and other immunological disorders towards the end of the last century. This article will revisit the current understanding and new developments in T cell pharmacology 10 years on from the TeGenero (TGN 1412) debacle and look at more recent successes with ex vivo antigen presenting cell incubation technologies; T cell receptor (TCR) engineering and adoptive T cell therapy both with chimaeric antibodies and also with modified T cell receptors themselves. Features of T cell biology will be explored and processes often highly unique to humans will be used to highlight what many are beginning to see as an exciting new monoclonal (T cell) frontier for drug development.
Collapse
Affiliation(s)
- Dominic Smethurst
- Adaptimmune Ltd & Immunocore Ltd, Abingdon Oxfordshire, OX14 4RX, UK.
| |
Collapse
|
8
|
Zheng J, Guo Y, Ji X, Cui L, He W. A novel antibody-like TCRγδ-Ig fusion protein exhibits antitumor activity against human ovarian carcinoma. Cancer Lett 2013; 341:150-8. [PMID: 23920126 DOI: 10.1016/j.canlet.2013.07.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 07/09/2013] [Accepted: 07/28/2013] [Indexed: 10/26/2022]
Abstract
TCRγ9δ2(OT3) is a tumor-specific TCR with an unique complementarity-determining region 3 (CDR3) sequence, referred to as OT3, in its δ2 chain. This region was identified in tumor-infiltrating lymphocytes (TILs) from human ovarian epithelial carcinoma. We demonstrated that TCRγ9δ2(OT3)-Fc, a fusion protein composed of the complete extracellular domains of the γ9 and δ2 chains linked to the Fc domains of human IgG1, exhibited successful binding to multiple human carcinoma cell lines. In vitro, TCRγ9δ2(OT3)-Fc mediated cell killing via antibody-dependent cellular cytotoxicity (ADCC) in a dose-dependent manner. In vivo, TCRγ9δ2(OT3)-Fc significantly inhibited tumor growth and enhanced survival in human ovarian carcinoma xenograft models. Our findings suggest that the TCRγ9δ2(OT3)-Fc fusion protein possesses both the antigen-recognition properties of TCR γδ and the Fc-mediated effector functions of the antibody.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, National Key Laboratory of Medical Molecular Biology, Beijing, China.
| | | | | | | | | |
Collapse
|
9
|
Huang C, Zhu Y, Jiang Y, Li Z, Yao J, Duan G, Li D, Wang Q. Anticancer Activities of Anti-Membrane Antibodies against Colon Carcinoma Cells Undergoing Chemotherapy. TUMORI JOURNAL 2013; 99:113-20. [DOI: 10.1177/030089161309900119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background Chemotherapy combined with target therapy using antibodies against tumor cell membrane antigens may greatly increase the survival of cancer patients. Similar to autoantigens in autoimmunity diseases, certain membrane components may be more heterogeneous and create new determinants of antigens or haptens after chemotherapy. The aim of the current study was to prepare anti-membrane antibodies against colon carcinoma cells undergoing chemotherapy and examine their anticancer activities in vitro. Methods After the colon carcinoma cells were treated by mimic chemotherapy, the synthesized poly-lysine was used as a carrier to link the membrane antigen or hapten with the covalent bond of carbodiimide bridging. It was affirmed by fluorescence-activated cell sorting under laser confocal microscopy that the vaccine with poly-lysine membrane-linked cells with a covalent bond was successfully engineered. Then, the cognate mice were vaccinated, and the anti-membrane polyclonal antibodies were prepared and validated for their activities. Results The anti-membrane polyclonal antibodies were effectively induced and prepared. Folliculus lymphaticus were found significantly increased in vaccinated mice, and B lymphocyte proliferation was also intensively stimulated by vaccine and generating antibodies. The polyclonal antibodies, exhibiting minimal endotoxicity, displayed intensive sensitivity, high affinity and strong specificity. They also elicited apoptosis and necrosis for wild type colon carcinoma cells and offered synergistic effect to repress the chemotherapy-resistant tumor cells. Conclusions The poly-lysine-linked membrane for colon carcinoma cells undergoing chemotherapy could produce the anti-membrane polyclonal antibodies (promising as novel antibody molecules for target therapy) and generate an effective immune attack on the surviving cancer cells.
Collapse
Affiliation(s)
- Changxin Huang
- Affiliated Hospital with Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Ying Zhu
- Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiqian Jiang
- Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhaoyang Li
- Affiliated Hospital with Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Jifen Yao
- Affiliated Hospital with Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Guangliang Duan
- Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Da Li
- Institute of Immunology, Zhejiang University, Hangzhou, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Ozawa T, Horii M, Kobayashi E, Jin A, Kishi H, Muraguchi A. The binding affinity of a soluble TCR-Fc fusion protein is significantly improved by crosslinkage with an anti-Cβ antibody. Biochem Biophys Res Commun 2012; 422:245-9. [PMID: 22575452 DOI: 10.1016/j.bbrc.2012.04.134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 04/24/2012] [Indexed: 11/24/2022]
Abstract
The identification and cloning of tumor antigen-specific T cell receptors (TCRs) and the production of the soluble form of the TCR (sTCR) contributed to the development of diagnostic and therapeutic tools for cancer. Recently, several groups have reported the development of technologies for the production of sTCRs. The native sTCR has a very low binding affinity for the antigenic peptide/MHC (p/MHC) complex. In this study, we established a technology to produce high affinity, functional sTCRs. We generated a novel sTCR-Fc fusion protein composed of the TCR V and C regions of the TCR linked to the immunoglobulin (Ig) Fc region. A Western blot analysis revealed that the molecular weight of the fusion protein was approximately 60 kDa under reducing conditions and approximately 100-200 kDa under non-reducing conditions. ELISAs using various antibodies showed that the structure of each domain of the TCR-Fc protein was intact. The TCR-Fc protein immobilized by an anti-Cβ antibody effectively bound to a p/MHC tetramer. An SPR analysis showed that the TCR-Fc protein had a low binding affinity (KD; 1.1 × 10(-5)M) to the p/MHC monomer. Interestingly, when the TCR-Fc protein was pre-incubated with an anti-Cβ antibody, its binding affinity for p/MHC increased by 5-fold (2.2 × 10(-6)M). We demonstrated a novel method for constructing a functional soluble TCR using the Ig Fc region and showed that the binding affinity of the functional sTCR-Fc was markedly increased by an anti-Cβ antibody, which is probably due to the stabilization of the Vα/Vβ region of the TCR. These findings provide new insights into the binding of sTCRs to p/MHCs and will hopefully be instrumental in establishing functional sTCR as a diagnostic and therapeutic tool for cancer.
Collapse
Affiliation(s)
- Tatsuhiko Ozawa
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | | | | | | | | | | |
Collapse
|
11
|
Dushek O, Aleksic M, Wheeler RJ, Zhang H, Cordoba SP, Peng YC, Chen JL, Cerundolo V, Dong T, Coombs D, van der Merwe PA. Antigen potency and maximal efficacy reveal a mechanism of efficient T cell activation. Sci Signal 2011; 4:ra39. [PMID: 21653229 PMCID: PMC4143974 DOI: 10.1126/scisignal.2001430] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
T cell activation, a critical event in adaptive immune responses, depends on productive interactions between T cell receptors (TCRs) and antigens presented as peptide-bound major histocompatibility complexes (pMHCs). Activated T cells lyse infected cells, secrete cytokines, and perform other effector functions with various efficiencies, which depend on the binding parameters of the TCR-pMHC complex. The mechanism through which binding parameters are translated to the efficiency of T cell activation, however, remains controversial. The "affinity model" suggests that the dissociation constant (KD) of the TCR-pMHC complex determines the response, whereas the "productive hit rate model" suggests that the off-rate (koff) is critical. Here, we used mathematical modeling to show that antigen potency, as determined by the EC50 (half-maximal effective concentration), which is used to support KD-based models, could not discriminate between the affinity and the productive hit rate models. Both models predicted a correlation between EC50 and KD, but only the productive hit rate model predicted a correlation between maximal efficacy (Emax), the maximal T cell response induced by pMHC, and koff. We confirmed the predictions made by the productive hit rate model in experiments with cytotoxic T cell clones and a panel of pMHC variants. Thus, we propose that the activity of an antigen is determined by both its potency (EC50) and maximal efficacy (Emax).
Collapse
Affiliation(s)
- Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, UK
- Centre for Mathematical Biology, University of Oxford, OX1 3LB, UK
| | - Milos Aleksic
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, UK
| | | | - Hao Zhang
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, UK
| | | | - Yan-Chun Peng
- Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS, UK
| | - Ji-Li Chen
- Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS, UK
| | - Vincenzo Cerundolo
- Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS, UK
| | - Tao Dong
- Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS, UK
| | - Daniel Coombs
- Department of Mathematics and Institute of Applied Mathematics, University of British Columbia, V6T 1Z2, Canada
| | | |
Collapse
|
12
|
Steele JC, Rao A, Marsden JR, Armstrong CJ, Berhane S, Billingham LJ, Graham N, Roberts C, Ryan G, Uppal H, Walker C, Young LS, Steven NM. Phase I/II trial of a dendritic cell vaccine transfected with DNA encoding melan A and gp100 for patients with metastatic melanoma. Gene Ther 2011; 18:584-93. [PMID: 21307889 DOI: 10.1038/gt.2011.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This trial tested a dendritic cell (DC) therapeutic cancer vaccine in which antigen is loaded using a novel non-viral transfection method enabling the uptake of plasmid DNA condensed with a cationic peptide. Proof of principle required the demonstration of diverse T lymphocyte responses following vaccination, including multiple reactivities restricted through both major histocompatibility complex (MHC) class I and II. Patients with advanced melanoma were offered four cycles of vaccination with autologous DC expressing melan A and gp100. Disease response was measured using Response Evaluation Criteria in Solid Tumours. Circulating MHC class I- and II-restricted responses were measured against peptide and whole antigen targets using interferon-γ ELIspot and enzyme-linked immunosorbent assay assays, respectively. Responses were analyzed across the trial population and presented descriptively for some individuals. Twenty-five patients received at least one cycle. Vaccination was well tolerated. Three patients had reduction in disease volume. Across the trial population, vaccination resulted in an expansion of effector responses to both antigens, to the human leukocyte antigen A2-restricted modified epitope, melan A ELAGIGILTV, and to a panel of MHC class I- and II-restricted epitopes. Vaccination with mature DC non-virally transfected with DNA encoding antigen had biological effect causing tumour regression and inducing diverse T lymphocyte responses.
Collapse
Affiliation(s)
- J C Steele
- Cancer Research UK Clinical Trials Unit, School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Udyavar A, Geiger TL. Rebalancing immune specificity and function in cancer by T-cell receptor gene therapy. Arch Immunol Ther Exp (Warsz) 2010; 58:335-46. [PMID: 20680493 PMCID: PMC2928402 DOI: 10.1007/s00005-010-0090-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 04/16/2010] [Indexed: 01/08/2023]
Abstract
Adoptive immunotherapy with tumor-specific T lymphocytes has demonstrated clinical benefit in some cancers, particularly melanoma. Yet isolating and expanding tumor-specific cells from patients is challenging and there is limited ability to control T-cell affinity and response characteristics. T-cell receptor (TCR) gene therapy, in which T lymphocytes for immunotherapy are redirected using an introduced rearranged TCR, has emerged as an important alternative. Successful TCR gene therapy requires consideration of a number of issues, including TCR specificity and affinity, optimal gene therapy constructs, types of T cells administered, and the survival and activity of the modified cells. In this review we highlight the rationale for and experience with TCR gene therapy as well as new approaches to enhancing it.
Collapse
Affiliation(s)
- Akshata Udyavar
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Terrence L. Geiger
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| |
Collapse
|
14
|
Pierce BG, Haidar JN, Yu Y, Weng Z. Combinations of affinity-enhancing mutations in a T cell receptor reveal highly nonadditive effects within and between complementarity determining regions and chains. Biochemistry 2010; 49:7050-9. [PMID: 20681514 DOI: 10.1021/bi901969a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Understanding the energetic and structural response to multiple mutations in a protein-protein interface is a key aspect of rational protein design. Here we investigate the cooperativity of combinations of point mutations of a T cell receptor (TCR) that binds in vivo to HLA-A2 MHC and a viral peptide. The mutations were obtained from two sources: a structure-based design study on the TCR alpha chain (nine mutations) and an in vitro selection study on the TCR beta chain (four mutations). In addition to combining the highest-affinity variants from each chain, we tested other combinations of mutations within and among the chains, for a total of 23 TCR mutants that we measured for binding kinetics to the peptide and major histocompatibility complex. A wide range of binding affinities was observed, from 2- to 1000-fold binding improvement versus that of the wild type, with significant nonadditive effects observed within and between TCR chains. This included an amino acid-dependent cooperative interaction between CDR1 and CDR3 residues that are separated by more than 9 A in the wild-type complex. When analyzing the kinetics of the mutations, we found that the association rates were primarily responsible for the cooperativity, while the dissociation rates were responsible for the anticooperativity (less-than-additive energetics). On the basis of structural modeling of anticooperative mutants, we determined that side chain clash between proximal mutants likely led to nonadditive binding energies. These results highlight the complex nature of TCR association and binding and will be informative in future design efforts that combine multiple mutant residues.
Collapse
Affiliation(s)
- Brian G Pierce
- Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
15
|
Lunde E, Løset GÅ, Bogen B, Sandlie I. Stabilizing mutations increase secretion of functional soluble TCR-Ig fusion proteins. BMC Biotechnol 2010; 10:61. [PMID: 20735812 PMCID: PMC2936418 DOI: 10.1186/1472-6750-10-61] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 08/24/2010] [Indexed: 12/02/2022] Open
Abstract
Background Whereas T cell receptors (TCRs) detect peptide/major histocompatibility complexes (pMHCs) with exquisite specificity, there are challenges regarding their expression and use as soluble detection molecules due to molecular instability. We have investigated strategies for the production of TCR-immunoglobulin (Ig) fusion proteins. Two different TCRs that are characteristic of a mouse model for idiotype (Id) dependent immune regulation were engineered. They are structurally unrelated with different variable (V), diversity (D) and joining (J) segments, but each share one V gene segment, either Vα or Vβ, with the well characterized murine TCR, 2C. Results Several TCR-Ig formats were assessed. In one, the TCR V domains were fused to Ig constant (C) regions. In others, the complete extracellular part of the TCR was fused either to a complete Ig or an Ig Fc region. All molecules were initially poorly secreted from eukaryotic cells, but replacement of unfavourable amino acids in the V regions improved secretion, as did the introduction of a disulfide bridge between the TCR C domains and the removal of an unpaired cysteine. A screening strategy for selection of mutations that stabilize the actual fusion molecules was developed and used successfully. Molecules that included the complete heterodimeric TCR, with a stabilizing disulfide bridge, were correctly folded as they bound TCR-specific antibodies (Abs) and detected pMHC on cells after specific peptide loading. Conclusions We show that fully functional TCR-Ig fusion proteins can be made in good yields following stabilizing engineering of TCR V and C region genes. This is important since TCR-Ig fusions will be important probes for the presence of specific pMHCs in vitro and in vivo. In the absence of further affinity maturation, the reagents will be very useful for the detection of kinetic stability of complexes of peptide and MHC.
Collapse
Affiliation(s)
- Elin Lunde
- Department of Molecular Biosciences, Centre for Immune Regulation, University of Oslo, Oslo 0316, Norway
| | | | | | | |
Collapse
|
16
|
Goff SL, Johnson LA, Black MA, Xu H, Zheng Z, Cohen CJ, Morgan RA, Rosenberg SA, Feldman SA. Enhanced receptor expression and in vitro effector function of a murine-human hybrid MART-1-reactive T cell receptor following a rapid expansion. Cancer Immunol Immunother 2010; 59:1551-60. [PMID: 20628878 DOI: 10.1007/s00262-010-0882-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 06/08/2010] [Indexed: 12/15/2022]
Abstract
Peripheral blood lymphocytes (PBL) genetically modified to express T cell receptors (TCR) specific to known melanoma antigens, such as melanoma antigen recognized by T cells-1 (MART-1), and gp100 can elicit objective tumor regression when administered to patients with metastatic melanoma. It has also been demonstrated that modifications within the constant regions of a fully human TCR can enhance surface expression and stability without altering antigen specificity. In this study, we evaluated the substitution of murine constant regions for their human counterpart within the DMF5 MART-1-specific TCR. Unlike previous studies, all modified TCRs were inserted into retroviral vectors and analyzed for expression and function following a clinical transduction protocol. PBL were transduced with retroviral supernatant generated from stable packaging lines encoding melanoma-specific TCRs. This protocol resulted in high levels of antigen-specific T cells without the need for additional peptide stimulation and selection. Both the human and murinized TCR efficiently transduced PBL; however, the murinized TCR exhibited significantly higher tetramer binding, mean fluorescence intensity, as well as, increased in vitro effector function following our clinical transduction and expansion protocol. Additional TCR modifications including insertion of a second disulfide bond or the linker modifications evaluated herein did not significantly enhance TCR expression or subsequent in vitro effector function. We conclude that the substitution of a human constant region with a murine constant region was sufficient to increase receptor expression and tetramer binding as well as antitumor activity of the DMF5 TCR and could be a tool to augment other antigen-specific TCR.
Collapse
Affiliation(s)
- Stephanie L Goff
- Department of Surgery, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Chervin AS, Aggen DH, Raseman JM, Kranz DM. Engineering higher affinity T cell receptors using a T cell display system. J Immunol Methods 2008; 339:175-84. [PMID: 18854190 DOI: 10.1016/j.jim.2008.09.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 09/16/2008] [Accepted: 09/18/2008] [Indexed: 02/02/2023]
Abstract
The T cell receptor (TCR) determines the cellular response to antigens, which are presented on the surface of target cells in the form of a peptide bound to a product of the major histocompatibility complex (pepMHC). The response of the T cell depends on the affinity of the TCR for the pepMHC, yet many TCRs have been shown to be of low affinity, and some naturally occurring T cell responses are poor due to low affinities. Accordingly, engineering the TCR for increased affinity for pepMHC, particularly tumor-associated antigens, has become an increasingly desirable goal, especially with the advent of adoptive T cell therapies. For largely technical reasons, to date there have been only a handful of TCRs engineered in vitro for higher affinity using well established methods of protein engineering. Here we report the use of a T cell display system, using a retroviral vector, for generating a high-affinity TCR from the mouse T cell clone 2C. The method relies on the display of the TCR, in its normal, signaling competent state, as a CD3 complex on the T cell surface. A library in the CDR3alpha of the 2C TCR was generated in the MSCV retroviral vector and transduced into a TCR-negative hybridoma. Selection of a high-affinity, CD8-independent TCR was accomplished after only two rounds of flow cytometric sorting using the pepMHC SIYRYYGL/Kb (SIY/Kb). The selected TCR contained a sequence motif in the CDR3alpha with characteristics of several other TCRs previously selected by yeast display. In addition, it was possible to directly use the selected T cell hybridoma in functional assays without the need for sub-cloning, revealing that the selected TCR was capable of mediating CD8-independent activity. The method may be useful in the direct isolation and characterization of TCRs that could be used in therapies with adoptive transferred T cells.
Collapse
Affiliation(s)
- Adam S Chervin
- Department of Biochemistry, University of Illinois, 600 S. Matthews Ave., Urbana, IL 61801, USA
| | | | | | | |
Collapse
|
18
|
Sergeeva A, Kolonin MG, Molldrem JJ, Pasqualini R, Arap W. Display technologies: application for the discovery of drug and gene delivery agents. Adv Drug Deliv Rev 2006; 58:1622-54. [PMID: 17123658 PMCID: PMC1847402 DOI: 10.1016/j.addr.2006.09.018] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Accepted: 09/29/2006] [Indexed: 01/17/2023]
Abstract
Recognition of molecular diversity of cell surface proteomes in disease is essential for the development of targeted therapies. Progress in targeted therapeutics requires establishing effective approaches for high-throughput identification of agents specific for clinically relevant cell surface markers. Over the past decade, a number of platform strategies have been developed to screen polypeptide libraries for ligands targeting receptors selectively expressed in the context of various cell surface proteomes. Streamlined procedures for identification of ligand-receptor pairs that could serve as targets in disease diagnosis, profiling, imaging and therapy have relied on the display technologies, in which polypeptides with desired binding profiles can be serially selected, in a process called biopanning, based on their physical linkage with the encoding nucleic acid. These technologies include virus/phage display, cell display, ribosomal display, mRNA display and covalent DNA display (CDT), with phage display being by far the most utilized. The scope of this review is the recent advancements in the display technologies with a particular emphasis on molecular mapping of cell surface proteomes with peptide phage display. Prospective applications of targeted compounds derived from display libraries in the discovery of targeted drugs and gene therapy vectors are discussed.
Collapse
Affiliation(s)
- Anna Sergeeva
- Department of Blood and Marrow Transplantation, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Mikhail G. Kolonin
- Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Jeffrey J. Molldrem
- Department of Blood and Marrow Transplantation, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Renata Pasqualini
- Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Wadih Arap
- Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| |
Collapse
|