1
|
Wagner EK, Qerqez AN, Stevens CA, Nguyen AW, Delidakis G, Maynard JA. Human cytomegalovirus-specific T-cell receptor engineered for high affinity and soluble expression using mammalian cell display. J Biol Chem 2019; 294:5790-5804. [PMID: 30796163 PMCID: PMC6463697 DOI: 10.1074/jbc.ra118.007187] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/07/2019] [Indexed: 01/01/2023] Open
Abstract
T-cell receptors (TCR) have considerable potential as therapeutics and antibody-like reagents to monitor disease progression and vaccine efficacy. Whereas antibodies recognize only secreted and surface-bound proteins, TCRs recognize otherwise inaccessible disease-associated intracellular proteins when they are presented as processed peptides bound to major histocompatibility complexes (pMHC). TCRs have been primarily explored for cancer therapy applications but could also target infectious diseases such as cytomegalovirus (CMV). However, TCRs are more difficult to express and engineer than antibodies, and advanced methods are needed to enable their widespread use. Here, we engineered the human CMV-specific TCR RA14 for high-affinity and robust soluble expression. To achieve this, we adapted our previously reported mammalian display system to present TCR extracellular domains and used this to screen CDR3 libraries for clones with increased pMHC affinity. After three rounds of selection, characterized clones retained peptide specificity and activation when expressed on the surface of human Jurkat T cells. We obtained high yields of soluble, monomeric protein by fusing the TCR extracellular domains to antibody hinge and Fc constant regions, adding a stabilizing disulfide bond between the constant domains and disrupting predicted glycosylation sites. One variant exhibited 50 nm affinity for its cognate pMHC, as measured by surface plasmon resonance, and specifically stained cells presenting this pMHC. Our work has identified a human TCR with high affinity for the immunodominant CMV peptide and offers a new strategy to rapidly engineer soluble TCRs for biomedical applications.
Collapse
Affiliation(s)
- Ellen K Wagner
- From the McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712
| | - Ahlam N Qerqez
- From the McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712
| | - Christopher A Stevens
- From the McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712
| | - Annalee W Nguyen
- From the McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712
| | - George Delidakis
- From the McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712
| | - Jennifer A Maynard
- From the McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712.
| |
Collapse
|
2
|
Watanabe K, Tsukahara T, Toji S, Saitoh S, Hirohashi Y, Nakatsugawa M, Kubo T, Kanaseki T, Kameshima H, Terui T, Sato N, Torigoe T. Development of a T-cell receptor multimer with high avidity for detecting a naturally presented tumor-associated antigen on osteosarcoma cells. Cancer Sci 2018; 110:40-51. [PMID: 30375705 PMCID: PMC6317924 DOI: 10.1111/cas.13854] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 11/28/2022] Open
Abstract
For efficacy of peptide vaccination immunotherapy for patients with cancer, endogenous expression of the target peptide/human leukocyte antigen (HLA) on cancer cells is required. However, it is difficult to evaluate the expression status of a peptide/HLA complex because of the lack of a soluble T‐cell receptor (TCR) that reacts with tumor‐associated antigen (TAA) with high avidity. In the present study, we developed two soluble TCR‐multimers that were each directed to TAA, survivin‐2B (SVN‐2B) and PBF in the context of HLA‐A24 (SVN‐2B TCR‐multimer and PBF TCR‐multimer, respectively), from CTL clones that were established from peptide‐vaccinated patients. Both TCR multimers could recognize cognate peptide‐pulsed antigen‐presenting cells, C1R‐A24 cells, in a CD8‐independent method. Moreover, the PBF TCR‐multimer successfully recognized a PBF peptide naturally presented on HLA‐A24+PBF+ osteosarcoma cells. Taken together, the results indicated that a TCR‐multimer might be useful for detection of a TAA‐derived peptide presented by HLA in patients receiving immunotherapy.
Collapse
Affiliation(s)
- Kazue Watanabe
- Department of Cancer Immunology, Medical and Biological Laboratories Co., Ltd, Ina, Japan.,Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomohide Tsukahara
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shingo Toji
- Department of Cancer Immunology, Medical and Biological Laboratories Co., Ltd, Ina, Japan
| | - Shogo Saitoh
- Department of Cancer Immunology, Medical and Biological Laboratories Co., Ltd, Ina, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Munehide Nakatsugawa
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Terufumi Kubo
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Kanaseki
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | - Noriyuki Sato
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Higashi-Sapporo Hospital, Sapporo, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
3
|
Anikeeva N, Sykulev Y, Delikatny EJ, Popov AV. Core-based lipid nanoparticles as a nanoplatform for delivery of near-infrared fluorescent imaging agents. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2014; 4:507-524. [PMID: 25250201 PMCID: PMC4171838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 08/06/2014] [Indexed: 06/03/2023]
Abstract
Pyropheophorbide a (Pyro) is a near-infrared (NIR) fluorescent dye and photosensitizer with high quantum yield that makes the dye suitable for tumor treatment both as an imaging and therapy agent. We have designed and synthesized a series of a Pyro-based NIR probes, based on the conjugation of Pyro with lipids. The nature of our probes requires the use of a lipophilic carrier to deliver the probes to cancer cell membranes. To address this, we have utilized lipid-based nanoparticles (LNPs) consisting of PEGylated lipids, which form the nanoparticle shell, and a lipid core. To endow the LNPs with targeting properties, nitrilotriacetic acid (NTA) lipids were included in the composition that enables the non-covalent attachment of His-tag targeting proteins preserving their functional activity. We found that the nature of the core molecules influence the nanoparticle size, shelf-life and stability at physiological temperature. Two different Pyro-lipid conjugates were loaded either into the core or shell of the LNPs. The conjugates revealed differential ability to be accumulated in the cell membrane of the target cells with time. Thus, the modular organization of the core-shell LNPs allows facile adjustment of their composition with goal to fine tuning the nanoparticle properties for in vivo application.
Collapse
Affiliation(s)
- Nadia Anikeeva
- Department of Microbiology and Immunology and Kimmel Cancer Center, Thomas Jefferson UniversityPhiladelphia, Pennsylvania, USA
| | - Yuri Sykulev
- Department of Microbiology and Immunology and Kimmel Cancer Center, Thomas Jefferson UniversityPhiladelphia, Pennsylvania, USA
| | - Edward J Delikatny
- Department of Radiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphia, Pennsylvania, USA
| | - Anatoliy V Popov
- Department of Radiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Low JL, Naidoo A, Yeo G, Gehring AJ, Ho ZZ, Yau YH, Shochat SG, Kranz DM, Bertoletti A, Grotenbreg GM. Binding of TCR multimers and a TCR-like antibody with distinct fine-specificities is dependent on the surface density of HLA complexes. PLoS One 2012; 7:e51397. [PMID: 23251518 PMCID: PMC3519586 DOI: 10.1371/journal.pone.0051397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 10/31/2012] [Indexed: 01/02/2023] Open
Abstract
Class I Major Histocompatibility Complex (MHC) molecules evolved to sample degraded protein fragments from the interior of the cell, and to display them at the surface for immune surveillance by CD8+ T cells. The ability of these lymphocytes to identify immunogenic peptide-MHC (pMHC) products on, for example, infected hepatocytes, and to subsequently eliminate those cells, is crucial for the control of hepatitis B virus (HBV). Various protein scaffolds have been designed to recapitulate the specific recognition of presented antigens with the aim to be exploited both diagnostically (e.g. to visualize cells exposed to infectious agents or cellular transformation) and therapeutically (e.g. for the delivery of drugs to compromised cells). In line with this, we report the construction of a soluble tetrameric form of an αβ T cell receptor (TCR) specific for the HBV epitope Env183–191 restricted by HLA-A*02:01, and compare its avidity and fine-specificity with a TCR-like monoclonal antibody generated against the same HLA target. A flow cytometry-based assay with streptavidin-coated beads loaded with Env183–191/HLA-A*02:01 complexes at high surface density, enabled us to probe the specific interaction of these molecules with their cognate pMHC. We demonstrate that the TCR tetramer has similar avidity for the pMHC as the antibody, but they differ in their fine-specificity, with only the TCR tetramer being capable of binding both natural variants of the Env183–191 epitope found in HBV genotypes A/C/D (187Arg) and genotype B (187Lys). Collectively, the results highlight the promiscuity of our soluble TCR, which could be an advantageous feature when targeting cells infected with a mutation-prone virus, but that binding of the soluble oligomeric TCR relies considerably on the surface density of the presented antigen.
Collapse
Affiliation(s)
- Jianrong L. Low
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, (A*STAR), Singapore, Singapore
- Department of Biochemistry, University of Illinois at Urbana-Champaign (UIUC), Urbana, Illinois, United States of America
| | - Anneta Naidoo
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, (A*STAR), Singapore, Singapore
| | - Gladys Yeo
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| | - Adam J. Gehring
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, (A*STAR), Singapore, Singapore
| | - Zi Zong Ho
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, (A*STAR), Singapore, Singapore
| | - Yin Hoe Yau
- Nanyang Technological University (NTU), School of Biological Sciences, Singapore, Singapore
| | - Susana G. Shochat
- Nanyang Technological University (NTU), School of Biological Sciences, Singapore, Singapore
| | - David M. Kranz
- Department of Biochemistry, University of Illinois at Urbana-Champaign (UIUC), Urbana, Illinois, United States of America
| | - Antonio Bertoletti
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, (A*STAR), Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| | - Gijsbert M. Grotenbreg
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- Department of Biological Sciences, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- * E-mail:
| |
Collapse
|
5
|
T cell receptor-like recognition of tumor in vivo by synthetic antibody fragment. PLoS One 2012; 7:e43746. [PMID: 22916301 PMCID: PMC3423377 DOI: 10.1371/journal.pone.0043746] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 07/23/2012] [Indexed: 11/20/2022] Open
Abstract
A major difficulty in treating cancer is the inability to differentiate between normal and tumor cells. The immune system differentiates tumor from normal cells by T cell receptor (TCR) binding of tumor-associated peptides bound to Major Histocompatibility Complex (pMHC) molecules. The peptides, derived from the tumor-specific proteins, are presented by MHC proteins, which then serve as cancer markers. The TCR is a difficult protein to use as a recombinant protein because of production issues and has poor affinity for pMHC; therefore, it is not a good choice for use as a tumor identifier outside of the immune system. We constructed a synthetic antibody-fragment (Fab) library in the phage-display format and isolated antibody-fragments that bind pMHC with high affinity and specificity. One Fab, fE75, recognizes our model cancer marker, the Human Epidermal growth factor Receptor 2 (HER2/neu) peptide, E75, bound to the MHC called Human Leukocyte Antigen-A2 (HLA-A2), with nanomolar affinity. The fE75 bound selectively to E75/HLA-A2 positive cancer cell lines in vitro. The fE75 Fab conjugated with 64Cu selectively accumulated in E75/HLA-A2 positive tumors and not in E75/HLA-A2 negative tumors in an HLA-A2 transgenic mouse as probed using positron emission tomography/computed tomography (PET/CT) imaging. Considering that hundreds to thousands of different peptides bound to HLA-A2 are present on the surface of each cell, the fact that fE75 arrives at the tumor at all shows extraordinary specificity. These antibody fragments have great potential for diagnosis and targeted drug delivery in cancer.
Collapse
|
6
|
Anikeeva N, Gakamsky D, Schøller J, Sykulev Y. Evidence that the density of self peptide-MHC ligands regulates T-cell receptor signaling. PLoS One 2012; 7:e41466. [PMID: 22870225 PMCID: PMC3411518 DOI: 10.1371/journal.pone.0041466] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 06/21/2012] [Indexed: 11/24/2022] Open
Abstract
Noncognate or self peptide-MHC (pMHC) ligands productively interact with T-cell receptor (TCR) and are always in a large access over the cognate pMHC on the surface of antigen presenting cells. We assembled soluble cognate and noncognate pMHC class I (pMHC-I) ligands at designated ratios on various scaffolds into oligomers that mimic pMHC clustering and examined how multivalency and density of the pMHCs in model clusters influences the binding to live CD8 T cells and the kinetics of TCR signaling. Our data demonstrate that the density of self pMHC-I proteins promotes their interaction with CD8 co-receptor, which plays a critical role in recognition of a small number of cognate pMHC-I ligands. This suggests that MHC clustering on live target cells could be utilized as a sensitive mechanism to regulate T cell responsiveness.
Collapse
Affiliation(s)
- Nadia Anikeeva
- Department of Microbiology and Immunology and Kimmel Cancer Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | | | | | - Yuri Sykulev
- Department of Microbiology and Immunology and Kimmel Cancer Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
7
|
Stone JD, Artyomov MN, Chervin AS, Chakraborty AK, Eisen HN, Kranz DM. Interaction of streptavidin-based peptide-MHC oligomers (tetramers) with cell-surface TCRs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:6281-90. [PMID: 22102724 PMCID: PMC3237744 DOI: 10.4049/jimmunol.1101734] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The binding of oligomeric peptide-MHC (pMHC) complexes to cell surface TCR can be considered to approximate TCR-pMHC interactions at cell-cell interfaces. In this study, we analyzed the equilibrium binding of streptavidin-based pMHC oligomers (tetramers) and their dissociation kinetics from CD8(pos) T cells from 2C-TCR transgenic mice and from T cell hybridomas that expressed the 2C TCR or a high-affinity mutant (m33) of this TCR. Our results show that the tetramers did not come close to saturating cell-surface TCR (binding only 10-30% of cell-surface receptors), as is generally assumed in deriving affinity values (K(D)), in part because of dissociative losses from tetramer-stained cells. Guided by a kinetic model, the oligomer dissociation rate and equilibrium constants were seen to depend not only on monovalent association and dissociation rates (k(off) and k(on)), but also on a multivalent association rate (μ) and TCR cell-surface density. Our results suggest that dissociation rates could account for the recently described surprisingly high frequency of tetramer-negative, functionally competent T cells in some T cell responses.
Collapse
MESH Headings
- Animals
- Hybridomas
- Major Histocompatibility Complex/genetics
- Major Histocompatibility Complex/immunology
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Models, Immunological
- Multiprotein Complexes/genetics
- Multiprotein Complexes/metabolism
- Peptides/metabolism
- Protein Binding/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Single-Chain Antibodies/metabolism
- Streptavidin/metabolism
Collapse
Affiliation(s)
- Jennifer D Stone
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA.
| | | | | | | | | | | |
Collapse
|
8
|
Mareeva T, Wanjalla C, Schnell MJ, Sykulev Y. A novel composite immunotoxin that suppresses rabies virus production by the infected cells. J Immunol Methods 2010; 353:78-86. [PMID: 19932697 PMCID: PMC2823984 DOI: 10.1016/j.jim.2009.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 11/01/2009] [Accepted: 11/10/2009] [Indexed: 10/20/2022]
Abstract
Using strepavidin as a scaffold, we have assembled a composite immunotoxin that consists of recombinant Pseudomonas exotoxin A subunit (PE38) and recombinant 25-D1.16 Fab fragment which recognizes the SIINFEKL (pOV8) peptide from ovalbumin in association with H-2K(b) MHC class I protein. The composite immunotoxin exercises cytotoxicity against H-2K(b+) cells sensitized with pOV8 peptide but not with irrelevant peptide. Specific binding of the immunotoxin to H-2K(b+) cells infected with recombinant rabies virus (RV) expressing pOV8 epitope (RV-pOV8) resulted in the suppression of the production of virus particles by the infected cells. This strategy allows readily produce different immunotoxins with desired specificity by combining various targeting and toxin molecules. The results provide a proof of concept that composite immunotoxins can be utilized as novel immunotherapeutics to stop virus spread in the acute phase of the infection allowing winning time for the development of protective immune response.
Collapse
Affiliation(s)
- Tatiana Mareeva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA19107
| | - Celestine Wanjalla
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA19107
| | - Matthias J. Schnell
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA19107
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA19107
- Jefferson Vaccine Center, Thomas Jefferson University, Philadelphia, PA19107
| | - Yuri Sykulev
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA19107
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA19107
- Jefferson Vaccine Center, Thomas Jefferson University, Philadelphia, PA19107
| |
Collapse
|
9
|
Beal AM, Anikeeva N, Varma R, Cameron TO, Vasiliver-Shamis G, Norris PJ, Dustin ML, Sykulev Y. Kinetics of early T cell receptor signaling regulate the pathway of lytic granule delivery to the secretory domain. Immunity 2009; 31:632-42. [PMID: 19833088 DOI: 10.1016/j.immuni.2009.09.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Revised: 09/02/2009] [Accepted: 09/22/2009] [Indexed: 01/03/2023]
Abstract
Cytolytic granules mediate killing of virus-infected cells by cytotoxic T lymphocytes. We show here that the granules can take long or short paths to the secretory domain. Both paths utilized the same intracellular molecular events, which have different spatial and temporal arrangements and are regulated by the kinetics of Ca(2+)-mediated signaling. Rapid signaling caused swift granule concentration near the microtubule-organizing center (MTOC) and subsequent delivery by the polarized MTOC directly to the secretory domain-the shortest path. Indolent signaling led to late recruitment of granules that moved along microtubules to the periphery of the synapse and then moved tangentially to fuse at the outer edge of the secretory domain-a longer path. The short pathway is associated with faster granule release and more efficient killing than the long pathway. Thus, the kinetics of early signaling regulates the quality of the T cell cytolytic response.
Collapse
Affiliation(s)
- Allison M Beal
- Department of Microbiology and Immunology and Kimmel Cancer Institute, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Pien GC, Basner-Tschakarjan E, Hui DJ, Mentlik AN, Finn JD, Hasbrouck NC, Zhou S, Murphy SL, Maus MV, Mingozzi F, Orange JS, High KA. Capsid antigen presentation flags human hepatocytes for destruction after transduction by adeno-associated viral vectors. J Clin Invest 2009; 119:1688-95. [PMID: 19436115 PMCID: PMC2689109 DOI: 10.1172/jci36891] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 03/23/2009] [Indexed: 01/25/2023] Open
Abstract
Adeno-associated virus (AAV) vectors are effective gene delivery vehicles mediating long-lasting transgene expression. Data from a clinical trial of AAV2-mediated hepatic transfer of the Factor IX gene (F9) into hemophilia B subjects suggests that CTL responses against AAV capsid can eliminate transduced hepatocytes and prevent long-term F9 expression. However, the capacity of hepatocytes to present AAV capsid-derived antigens has not been formally demonstrated, nor whether transduction by AAV sensitizes hepatocytes for CTL-mediated destruction. To investigate the fate of capsids after transduction, we engineered a soluble TCR for the detection of capsid-derived peptide:MHC I (pMHC) complexes. TCR multimers exhibited antigen and HLA specificity and possessed high binding affinity for cognate pMHC complexes. With this reagent, capsid pMHC complexes were detectable by confocal microscopy following AAV-mediated transduction of human hepatocytes. Although antigen presentation was modest, it was sufficient to flag transduced cells for CTL-mediated lysis in an in vitro killing assay. Destruction of hepatocytes was inhibited by soluble TCR, demonstrating a possible application for this reagent in blocking undesirable CTL responses. Together, these studies provide a mechanism for the loss of transgene expression and transient elevations in aminotransferases following AAV-mediated hepatic gene transfer in humans and a potential therapeutic intervention to abrogate these limitations imposed by the host T cell response.
Collapse
Affiliation(s)
- Gary C. Pien
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Etiena Basner-Tschakarjan
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel J. Hui
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ashley N. Mentlik
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan D. Finn
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicole C. Hasbrouck
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shangzhen Zhou
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Samuel L. Murphy
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marcela V. Maus
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Federico Mingozzi
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jordan S. Orange
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katherine A. High
- Division of Allergy and Immunology,
Division of Hematology, and
Howard Hughes Medical Institute, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|