1
|
Chen TT. Conditionally active T cell engagers for the treatment of solid tumors: rationale and clinical development. Expert Opin Biol Ther 2022; 22:955-963. [PMID: 35857922 DOI: 10.1080/14712598.2022.2098674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION T cell engagers are a class of bispecific molecules that induce highly potent T cell-dependent cytotoxicity by bringing T cell activating receptors into proximity with cancer-associated cell surface antigens. However, because of their high potency, there is a greater risk of on-target/off-tumor toxicity owing to normal tissues having tumor antigen expression even at low levels. To reduce these adverse events, the dysregulated activity of proteases within the tumor microenvironment has recently been explored to create inert prodrugs that become conditionally active engagers after their cleavage by these enzymes. AREAS COVERED T-cell engagers that have been introduced for clinical use, and their respective successes and failures are reviewed. The unique challenges of these bispecific molecules for treating solid tumors and prior technologies used to exploit the proteolytic tumor microenvironment to create better-tolerated prodrugs and how that experience has led to the current series of conditionally active T-cell engagers, are discussed. EXPERT OPINION Methods for modulating the serum half-life of both inert and activated T cell engagers could have important ramifications in how they infiltrate tumors and prevent toxicity. Alternative features of the tumor microenvironment can also be leveraged in the development of conditional T cell engagers.
Collapse
Affiliation(s)
- T Timothy Chen
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Development Center Americas, Inc
| |
Collapse
|
2
|
Lantz O, Teyton L. Identification of T cell antigens in the 21st century, as difficult as ever. Semin Immunol 2022; 60:101659. [PMID: 36183497 PMCID: PMC10332289 DOI: 10.1016/j.smim.2022.101659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Identifying antigens recognized by T cells is still challenging, particularly for innate like T cells that do not recognize peptides but small metabolites or lipids in the context of MHC-like molecules or see non-MHC restricted antigens. The fundamental reason for this situation is the low affinity of T cell receptors for their ligands coupled with a level of degeneracy that makes them bind to similar surfaces on antigen presenting cells. Herein we will describe non-exhaustively some of the methods that were used to identify peptide antigens and briefly mention the high throughput methods more recently proposed for that purpose. We will then present how the molecules recognized by innate like T cells (NKT, MAIT and γδ T cells) were discovered. We will show that serendipity was instrumental in many cases.
Collapse
Affiliation(s)
- Olivier Lantz
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France; Laboratoire d'Immunologie Clinique, Institut Curie, Paris 75005, France; Centre d'investigation Clinique en Biothérapie Gustave-Roussy Institut Curie (CIC-BT1428) Institut Curie, Paris 75005, France
| | - Luc Teyton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
3
|
Poussin M, Sereno A, Wu X, Huang F, Manro J, Cao S, Carpenito C, Glasebrook A, Powell Jr DJ, Demarest SJ. Dichotomous impact of affinity on the function of T cell engaging bispecific antibodies. J Immunother Cancer 2021; 9:e002444. [PMID: 34253637 PMCID: PMC8276301 DOI: 10.1136/jitc-2021-002444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Bispecific T cell engagers represent the majority of bispecific antibodies (BsAbs) entering the clinic to treat metastatic cancer. The ability to apply these agents safely and efficaciously in the clinic, particularly for solid tumors, has been challenging. Many preclinical studies have evaluated parameters related to the activity of T cell engaging BsAbs, but many questions remain. MAIN BODY This study investigates the impact of affinity of T cell engaging BsAbs with regards to potency, efficacy, and induction of immunomodulatory receptors/ligands using HER-2/CD3 BsAbs as a model system. We show that an IgG BsAb can be as efficacious as a smaller BsAb format both in vitro and in vivo. We uncover a dichotomous relationship between tumor-associated antigen (TAA) affinity and CD3 affinity requirements for cells that express high versus low levels of TAA. HER-2 affinity directly correlated with the CD3 engager lysis potency of HER-2/CD3 BsAbs when HER-2 receptor numbers are high (~200 K/cell), while the CD3 affinity did not impact potency until its binding affinity was extremely low (<600 nM). When HER-2 receptor numbers were lower (~20 K/cell), both HER-2 and CD3 affinity impacted potency. The high affinity anti-HER-2/low CD3 affinity BsAb also demonstrated lower cytokine induction levels in vivo and a dosing paradigm atypical of extremely high potency T cell engaging BsAbs reaching peak efficacy at doses >3 mg/kg. This data confirms that low CD3 affinity provides an opportunity for improved safety and dosing for T cell engaging BsAbs. T cell redirection also led to upregulation of Programmed cell death 1 (PD-1) and 4-1BB, but not CTLA-4 on T cells, and to Programmed death-ligand 1 (PD-L1) upregulation on HER-2HI SKOV3 tumor cells, but not on HER-2LO OVCAR3 tumor cells. Using this information, we combined anti-PD-1 or anti-4-1BB monoclonal antibodies with the HER-2/CD3 BsAb in vivo and demonstrated significantly increased efficacy against HER-2HI SKOV3 tumors via both combinations. CONCLUSIONS Overall, these studies provide an informational dive into the optimization process of CD3 engaging BsAbs for solid tumors indicating that a reduced affinity for CD3 may enable a better therapeutic index with a greater selectivity for the target tumor and a reduced cytokine release syndrome. These studies also provide an additional argument for combining T cell checkpoint inhibition and co-stimulation to achieve optimal efficacy. BACKGROUND
Collapse
Affiliation(s)
- Mathilde Poussin
- Pathology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Arlene Sereno
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
| | - Xiufeng Wu
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
| | - Flora Huang
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
| | - Jason Manro
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
| | - Shanshan Cao
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
| | - Carmine Carpenito
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
- Stelexis, New York, New York, USA
| | - Andrew Glasebrook
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
- Toralgen, San Diego, California, USA
| | - Daniel J Powell Jr
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephen J Demarest
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
- Tentarix, San Diego, California, USA
| |
Collapse
|
4
|
Yang B, Gao J, Pei Q, Xu H, Yu H. Engineering Prodrug Nanomedicine for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002365. [PMID: 33304763 PMCID: PMC7709995 DOI: 10.1002/advs.202002365] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/16/2020] [Indexed: 12/11/2022]
Abstract
Immunotherapy has shifted the clinical paradigm of cancer management. However, despite promising initial progress, immunotherapeutic approaches to cancer still suffer from relatively low response rates and the possibility of severe side effects, likely due to the low inherent immunogenicity of tumor cells, the immunosuppressive tumor microenvironment, and significant inter- and intratumoral heterogeneity. Recently, nanoformulations of prodrugs have been explored as a means to enhance cancer immunotherapy by simultaneously eliciting antitumor immune responses and reversing local immunosuppression. Prodrug nanomedicines, which integrate engineering advances in chemistry, oncoimmunology, and material science, are rationally designed through chemically modifying small molecule drugs, peptides, or antibodies to yield increased bioavailability and spatiotemporal control of drug release and activation at the target sites. Such strategies can help reduce adverse effects and enable codelivery of multiple immune modulators to yield synergistic cancer immunotherapy. In this review article, recent advances and translational challenges facing prodrug nanomedicines for cancer immunotherapy are overviewed. Last, key considerations are outlined for future efforts to advance prodrug nanomedicines aimed to improve antitumor immune responses and combat immune tolerogenic microenvironments.
Collapse
Affiliation(s)
- Bin Yang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Jing Gao
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Qing Pei
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Huixiong Xu
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| |
Collapse
|
5
|
Schreiber K, Karrison TG, Wolf SP, Kiyotani K, Steiner M, Littmann ER, Pamer EG, Kammertoens T, Schreiber H, Leisegang M. Impact of TCR Diversity on the Development of Transplanted or Chemically Induced Tumors. Cancer Immunol Res 2019; 8:192-202. [PMID: 31831634 DOI: 10.1158/2326-6066.cir-19-0567] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/24/2019] [Accepted: 12/06/2019] [Indexed: 12/21/2022]
Abstract
Burnet postulated that the diversity of T-cell receptors (TCR) allows T cells to protect against the development of cancers that display antigens with a similar, seemingly endless diversity. To test this hypothesis, we developed a strategy in which a single breeding pair of mice gives rise to four groups of sibling mice. Three of the four groups had a similar number of CD8+ T cells, but TCR diversity was either broad, significantly reduced, or absent when expressing only one type of TCR. The fourth group had no T cells. All mice shared the same housing, and, therefore, their microbial environment was similar. Only slight differences in the intestinal flora were observed under these conditions. An undisturbed broad TCR repertoire was required for the rejection of inoculated cancers displaying the natural antigenic heterogeneity of primary tumors, whereas even one type of TCR was sufficient to protect against artificial cancers stably expressing cognate antigens. The three groups of mice with limited or no TCR repertoire showed an increased risk of developing primary tumors after chemical induction. However, the risk of early death or morbidity in these cohorts of mice was significantly higher than in mice with a diverse TCR repertoire, and it remains unknown whether mice with reduced TCR diversity, who died early without cancer, would have developed tumors with higher, lower, or equal probability after induction. Together, TCR diversity seems crucial to overcome the natural genetic instability of cancers and their antigenic heterogeneity, which impacts the design of cellular therapies.
Collapse
Affiliation(s)
- Karin Schreiber
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Theodore G Karrison
- Department of Public Health Sciences, The University of Chicago, Chicago, Illinois
| | - Steven P Wolf
- Department of Pathology, The University of Chicago, Chicago, Illinois.,Institute of Immunology, Charité - Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Madeline Steiner
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Eric R Littmann
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric G Pamer
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas Kammertoens
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Hans Schreiber
- Department of Pathology, The University of Chicago, Chicago, Illinois.
| | - Matthias Leisegang
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| |
Collapse
|
6
|
Hwang I, Kim K, Choi S, Lomunova M. Potentiation of T Cell Stimulatory Activity by Chemical Fixation of a Weak Peptide-MHC Complex. Mol Cells 2017; 40:24-36. [PMID: 28152301 PMCID: PMC5303886 DOI: 10.14348/molcells.2017.2218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 12/12/2016] [Accepted: 12/27/2016] [Indexed: 12/13/2022] Open
Abstract
The stability of peptide-MHC complex (pMHC) is an important factor to shape the fate of peptide-specific T cell immune response, but how it influences on T cell activation process is poorly understood. To better understand that, we investigated various T cell activation events driven by Ld MHCI loaded with graded concentrations of P2Ca and QL9 peptides, respectively, with 2C TCR Tg T cells; the binding strength of P2Ca for Ld is measurably weaker than that of QL9, but either peptides in the context of Ld interact with 2C TCR with a similar strength. When their concentrations required for early T cell activation events, which occur within several minutes to an hour, were concerned, EC50s of QL9 were about 100 folds lower than those of P2Ca, which was expected from their association constants for Ld. When EC50s for late activation events, which takes over several hours to occur, were concerned, the differences grew even larger (> 300 folds), suggesting that, due to weak binding, Ld/P2Ca dissociate from each other more easily to lose its antigenicity in a short time. Accordingly, fixation of Ld/P2Ca with paraformaldehyde resulted in a significant improvement in its immunogenicity. These results imply that binding strength of a peptide for a MHC is a critical factor to determine the duration of pMHC-mediated T cell activation and thus the attainment of productive T cell activation. It is also suggested that paraformaldehyde fixation should be an effective tool to ameliorate the immunogenicity of pMHC with a poor stability.
Collapse
Affiliation(s)
- Inkyu Hwang
- Department of Chemistry and Chemical Biology, The Scripps Research Institute, La Jolla, California,
USA
- College of Pharmacy, Chungnam National University, Daejeon 34134,
Korea
| | - Kwangmi Kim
- Department of Chemistry and Chemical Biology, The Scripps Research Institute, La Jolla, California,
USA
- College of Pharmacy, Dankook University, Yongin 16890,
Korea
| | - Sojin Choi
- College of Pharmacy, Chungnam National University, Daejeon 34134,
Korea
| | - Maria Lomunova
- College of Pharmacy, Chungnam National University, Daejeon 34134,
Korea
| |
Collapse
|
7
|
Kaiser AD, Gadiot J, Guislain A, Blank CU. Mimicking homeostatic proliferation in vitro generates T cells with high anti-tumor function in non-lymphopenic hosts. Cancer Immunol Immunother 2013; 62:503-15. [PMID: 23001162 PMCID: PMC11029096 DOI: 10.1007/s00262-012-1350-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 09/02/2012] [Indexed: 10/27/2022]
Abstract
CD8(+) T cells undergoing homeostatic proliferation (HP) in a lymphopenic environment acquire a central memory-like phenotype (CD44(+) CD62L(+) Ly6c(+)). Such cells are readily functional in vitro, with a strong capacity to secrete IFNγ and IL-2 and to lyse target cells upon antigen recognition. In vivo, these memory-like T cells display potent anti-tumor reactivity. When addressing whether these remarkable properties were "acquired" or dependent on sustained HP, we observed, for the first time, that memory-like T cells retained full anti-tumor functions even when removed from their lymphopenic environment and retransferred into non-lymphopenic P14/Rag2(-/-) recipients (where HP is prevented). Moreover, memory-like T cells were superior to in vitro expanded effector T cells. We next sought to determine the conditions required to reproduce such a potent phenotype in vitro, in order to obtain optimal cells for adoptive cell transfer therapy. Assessing ex vivo lymph node cultures, dendritic cells, fibroblastic reticular cells, and HP-associated cytokines, we found that stimulation of naïve T cells with anti-CD3/CD28 beads and IL-15 (IL-7 was dispensable) led to the generation of memory-like T cell with a similar phenotype. Both in vitro and in vivo memory-like T cells retained the capacity to efficiently control tumor growth in non-lymphopenic hosts upon adoptive cell transfer. A similar phenotype could be imparted to human peripheral blood leukocytes with comparable culture conditions. Our data reinforce the idea that in vitro-generated memory-like T cells could benefit adoptive cell transfer therapies.
Collapse
Affiliation(s)
- Andrew D. Kaiser
- Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Jules Gadiot
- Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Aurelie Guislain
- Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Christian U. Blank
- Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
8
|
Kaiser AD, Schuster K, Gadiot J, Borkner L, Daebritz H, Schmitt C, Andreesen R, Blank C. Reduced tumor-antigen density leads to PD-1/PD-L1-mediated impairment of partially exhausted CD8⁺ T cells. Eur J Immunol 2012; 42:662-71. [PMID: 22144176 DOI: 10.1002/eji.201141931] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 11/07/2011] [Accepted: 11/29/2011] [Indexed: 11/10/2022]
Abstract
Clinical progression of cancer patients is often observed despite the presence of tumor-reactive T cells. Co-inhibitory ligands of the B7 superfamily have been postulated to play a part in this tumor-immune escape. One of these molecules, PD-L1 (B7-H1, CD274), is widely expressed on tumor cells and has been shown to mediate T-cell inhibition. However, attempts to correlate PD-L1 tumor expression with negative prognosis have been conflicting. To better understand when PD-1/PD-L1-mediated inhibition contributes to the functional impairment of tumor-specific CD8(+) T cells, we varied the levels of antigen density and/or PD-L1 expression at the surface of tumor cells and exposed them to CD8(+) T cells at different levels of functional exhaustion. We found that the gradual reduction of cognate antigen expression by PD-L1-expressing tumor cells increased the susceptibility of partially exhausted T cells to PD-1/PD-L1-mediated inhibition in vitro as well as in vivo. In conclusion, chronically stimulated CD8(+) T cells become sensitive to PD-1/PD-L1-mediated functional inhibition upon low antigen detection; a setting which is likely involved during tumor-immune escape.
Collapse
Affiliation(s)
- Andrew D Kaiser
- Division of Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy KM, Gajewski TF. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells. ACTA ACUST UNITED AC 2011; 208:2005-16. [PMID: 21930765 PMCID: PMC3182064 DOI: 10.1084/jem.20101159] [Citation(s) in RCA: 921] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The generation of antitumor CD8+ T cell responses requires type I interferon responsiveness in host antigen-presenting cells Despite lack of tumor control in many models, spontaneous T cell priming occurs frequently in response to a growing tumor. However, the innate immune mechanisms that promote natural antitumor T cell responses are undefined. In human metastatic melanoma, there was a correlation between a type I interferon (IFN) transcriptional profile and T cell markers in metastatic tumor tissue. In mice, IFN-β was produced by CD11c+ cells after tumor implantation, and tumor-induced T cell priming was defective in mice lacking IFN-α/βR or Stat1. IFN signaling was required in the hematopoietic compartment at the level of host antigen-presenting cells, and selectively for intratumoral accumulation of CD8α+ dendritic cells, which were demonstrated to be essential using Batf3−/− mice. Thus, host type I IFNs are critical for the innate immune recognition of a growing tumor through signaling on CD8α+ DCs.
Collapse
Affiliation(s)
- Mercedes B Fuertes
- Department of Pathology and Department of Medicine, Section of Hematology/Oncology, the University of Chicago, Chicago, IL, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Single-chain VαVβ T-cell receptors function without mispairing with endogenous TCR chains. Gene Ther 2011; 19:365-74. [PMID: 21753797 DOI: 10.1038/gt.2011.104] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Transduction of exogenous T-cell receptor (TCR) genes into patients' activated peripheral blood T cells is a potent strategy to generate large numbers of specific T cells for adoptive therapy of cancer and viral diseases. However, the remarkable clinical promise of this powerful approach is still being overshadowed by a serious potential consequence: mispairing of the exogenous TCR chains with endogenous TCR chains. These 'mixed' heterodimers can generate new specificities that result in graft-versus-host reactions. Engineering TCR constant regions of the exogenous chains with a cysteine promotes proper pairing and reduces the mispairing, but, as we show here, does not eliminate the formation of mixed heterodimers. By contrast, deletion of the constant regions, through use of a stabilized Vα/Vβ single-chain TCR (scTv), avoided mispairing completely. By linking a high-affinity scTv to intracellular signaling domains, such as Lck and CD28, the scTv was capable of activating functional T-cell responses in the absence of either the CD3 subunits or the co-receptors, and circumvented mispairing with endogenous TCRs. Such transduced T cells can respond to the targeted antigen independent of CD3 subunits via the introduced scTv, without the transduced T cells acquiring any new undefined and potentially dangerous specificities.
Collapse
|
11
|
Schrum AG, Gil D, Turka LA, Palmer E. Physical and functional bivalency observed among TCR/CD3 complexes isolated from primary T cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:870-8. [PMID: 21666056 DOI: 10.4049/jimmunol.1100538] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Unlike BCR and secreted Ig, TCR expression is not thought to occur in a bivalent form. The conventional monovalent model of TCR/CD3 is supported by published studies of complexes solubilized in the detergent digitonin, in which bivalency was not observed. We revisited the issue of TCR valency by examining complexes isolated from primary αβ T cells after solubilization in digitonin. Using immunoprecipitation followed by flow cytometry, we unexpectedly observed TCR/CD3 complexes that contained two TCRs per complex. Standard anti-TCR Abs, being bivalent themselves, tended to bind with double occupancy to bivalent TCRs; this property masked the presence of the second TCR per complex in certain Ab binding assays, which may partially explain why previous data did not reveal these bivalent complexes. We also found that the prevalence of bivalency among fully assembled, mature TCR/CD3 complexes was sufficient to impact the functional performance of immunoprecipitated TCRs in binding antigenic peptide/MHC-Ig fusion proteins. Both TCR positions per bivalent complex required an Ag-specific TCR to effect optimal binding to these soluble ligands. Therefore, we conclude that in primary T cells, TCR/CD3 complexes can be found that are physically and functionally bivalent. The expression of bivalent TCR/CD3 complexes has implications regarding potential mechanisms by which Ag may trigger signaling. It also suggests the possibility that the potential for bivalent expression could represent a general feature of Ag receptors.
Collapse
Affiliation(s)
- Adam G Schrum
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|
12
|
Wucherpfennig KW. The First Structures of T Cell Receptors Bound to Peptide–MHC. THE JOURNAL OF IMMUNOLOGY 2010; 185:6391-3. [DOI: 10.4049/jimmunol.1090110] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
13
|
Borkner L, Kaiser A, van de Kasteele W, Andreesen R, Mackensen A, Haanen JB, Schumacher TN, Blank C. RNA interference targeting programmed death receptor-1 improves immune functions of tumor-specific T cells. Cancer Immunol Immunother 2010; 59:1173-83. [PMID: 20349059 PMCID: PMC11030462 DOI: 10.1007/s00262-010-0842-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 02/23/2010] [Indexed: 12/21/2022]
Abstract
Adoptive cell transfer (ACT), either using rapidly expanded tumor infiltrating lymphocytes or T-cell receptor transduced peripheral blood lymphocytes, can be considered one of the most promising approaches in cancer immunotherapy. ACT results in the repopulation of the host with high frequencies of tumor-specific T cells; however, optimal function of these cells within the tumor micro-environment is required to reach long-term tumor clearance. We and others have shown that ongoing anti-tumor immune responses can be impaired by the expression of ligands, such as PD-L1 (B7-H1) on tumor cells. Such inhibitory molecules can affect T cells at the effector phase via their receptor PD-1. PD-L1/PD-1 interaction has indeed been shown crucial in inducing T-cell anergy and maintaining peripheral tolerance. In order to maximize anti-tumor responses, antibodies that target the PD-1/PD-L1 axis are currently in phase I/II trials. Alternatively, a more refined approach could be the selective targeting of PD-1 in tumor-specific T cells to obtain long-term resistance against PD-1-mediated inhibition. We addressed whether this goal could be achieved by means of retroviral siRNA delivery. Effective siRNA sequences resulting in the reduction of surface PD-1 expression led to improved murine as well as human T-cell immune functions in response to PD-L1 expressing melanoma cells. These data suggest that blockade of PD-1-mediated T-cell inhibition through siRNA forms a promising approach to achieve long-lasting enhancement of tumor-specific T-cell function in adoptive T-cell therapy protocols.
Collapse
MESH Headings
- Animals
- Antigens, Differentiation/genetics
- Antigens, Differentiation/metabolism
- Antigens, Neoplasm/immunology
- B7-1 Antigen/genetics
- B7-1 Antigen/immunology
- B7-1 Antigen/metabolism
- B7-H1 Antigen
- Cell Line, Tumor
- Genetic Vectors
- Humans
- Immunotherapy, Adoptive
- Melanoma/immunology
- Melanoma/therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Peptides/genetics
- Peptides/immunology
- Peptides/metabolism
- Programmed Cell Death 1 Receptor
- RNA Interference
- RNA, Small Interfering/genetics
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Retroviridae/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- Lisa Borkner
- Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, 1066 CX The Netherlands
- Department of Hematology and Oncology, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Andrew Kaiser
- Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, 1066 CX The Netherlands
| | - Willeke van de Kasteele
- Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, 1066 CX The Netherlands
| | - Reinhard Andreesen
- Department of Hematology and Oncology, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Andreas Mackensen
- Department of Hematology and Oncology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - John B. Haanen
- Department of Medical Oncology and Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, 1066 CX The Netherlands
| | - Ton N. Schumacher
- Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, 1066 CX The Netherlands
| | - Christian Blank
- Department of Medical Oncology and Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, 1066 CX The Netherlands
| |
Collapse
|
14
|
Hodes RJ. MHC Restricted Recognition by Cloned T Cells. Int Rev Immunol 2009. [DOI: 10.3109/08830188609056604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
15
|
Chakraverty R, Flutter B, Fallah-Arani F, Eom HS, Means T, Andreola G, Schwarte S, Buchli J, Cotter P, Zhao G, Sykes M. The host environment regulates the function of CD8+ graft-versus-host-reactive effector cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:6820-8. [PMID: 18981100 DOI: 10.4049/jimmunol.181.10.6820] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have examined how the host environment influences the graft-vs-leukemia (GVL) response following transfer of donor T cells to allogeneic chimeras. Donor T cells induce significant GVL when administered in large numbers to established mixed chimeras (MC). However, when using limiting numbers of T cells, we found that late transfer to MC induced less GVL than did early transfer to freshly irradiated allogeneic recipients. Late donor T cell transfer to MC was associated with marked accumulation of anti-host CD8 cells within the spleen, but delayed kinetics of differentiation, reduced expression of effector molecules including IFN-gamma, impaired cytotoxicity, and higher rates of sustained apoptosis. Furthermore, in contrast to the spleen, we observed a significant delay in donor CD8 cell recruitment to the bone marrow, a key location for hematopoietic tumors. Increasing the numbers of T cells transferred to MC led to the enhancement of CTL activity and detectable increases in absolute numbers of IFN-gamma(+) cells without inducing graft-vs-host disease (GVHD). TLR-induced systemic inflammation accelerated differentiation of functional CTL in MC but was associated with severe GVHD. In the absence of inflammation, both recipient T and non-T cell populations impeded the full development of GVHD-inducing effector function. We conclude that per-cell deficits in the function of donor CD8 cells activated in MC may be overcome by transferring larger numbers of T cells without inducing GVHD.
Collapse
Affiliation(s)
- Ronjon Chakraverty
- Transplantation Immunology Group, Department of Hematology, University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Fehr T, Wang S, Haspot F, Kurtz J, Blaha P, Hogan T, Chittenden M, Wekerle T, Sykes M. Rapid deletional peripheral CD8 T cell tolerance induced by allogeneic bone marrow: role of donor class II MHC and B cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:4371-80. [PMID: 18768896 DOI: 10.4049/jimmunol.181.6.4371] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mixed chimerism and donor-specific tolerance are achieved in mice receiving 3 Gy of total body irradiation and anti-CD154 mAb followed by allogeneic bone marrow (BM) transplantation. In this model, recipient CD4 cells are critically important for CD8 tolerance. To evaluate the role of CD4 cells recognizing donor MHC class II directly, we used class II-deficient donor marrow and were not able to achieve chimerism unless recipient CD8 cells were depleted, indicating that directly alloreactive CD4 cells were necessary for CD8 tolerance. To identify the MHC class II(+) donor cells promoting this tolerance, we used donor BM lacking certain cell populations or used positively selected cell populations. Neither donor CD11c(+) dendritic cells, B cells, T cells, nor donor-derived IL-10 were critical for chimerism induction. Purified donor B cells induced early chimerism and donor-specific cell-mediated lympholysis tolerance in both strain combinations tested. In contrast, positively selected CD11b(+) monocytes/myeloid cells did not induce early chimerism in either strain combination. Donor cell preparations containing B cells were able to induce early deletion of donor-reactive TCR-transgenic 2C CD8 T cells, whereas those devoid of B cells had reduced activity. Thus, induction of stable mixed chimerism depends on the expression of MHC class II on the donor marrow, but no requisite donor cell lineage was identified. Donor BM-derived B cells induced early chimerism, donor-specific cell-mediated lympholysis tolerance, and deletion of donor-reactive CD8 T cells, whereas CD11b(+) cells did not. Thus, BM-derived B cells are potent tolerogenic APCs for alloreactive CD8 cells.
Collapse
Affiliation(s)
- Thomas Fehr
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, 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: 2.9] [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
|
Peripheral deletional tolerance of alloreactive CD8 but not CD4 T cells is dependent on the PD-1/PD-L1 pathway. Blood 2008; 112:2149-55. [PMID: 18577709 DOI: 10.1182/blood-2007-12-127449] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Although interaction between programmed death-1 (PD-1) and the ligand PD-L1 has been shown to mediate CD8 cell exhaustion in the setting of chronic infection or the absence of CD4 help, a role for this pathway in attenuating early alloreactive CD8 cell responses has not been identified. We demonstrate that the PD-1/PD-L1 pathway is needed to rapidly tolerize alloreactive CD8 cells in a model that requires CD4 cells and culminates in CD8 cell deletion. This protocol involves allogeneic bone marrow transplantation (BMT) following conditioning with low-dose total body irradiation and anti-CD154 antibody. Tolerized donor-reactive T-cell receptor transgenic CD8 cells are shown to be in an abortive activation state prior to their deletion, showing early and prolonged expression of activation markers (compared with rejecting CD8 cells) while being functionally silenced by day 4 after transplantation. Although both tolerized and rejecting alloreactive CD8 cells up-regulate PD-1, CD8 cell tolerance is dependent on the PD-1/PD-L1 pathway. In contrast, CD4 cells are tolerized independently of this pathway following BMT with anti-CD154. These studies demonstrate a dichotomy between the requirements for CD4 and CD8 tolerance and identify a role for PD-1 in the rapid tolerization of an alloreactive T-cell population via a deletional mechanism.
Collapse
|
19
|
Kline J, Brown IE, Zha YY, Blank C, Strickler J, Wouters H, Zhang L, Gajewski TF. Homeostatic proliferation plus regulatory T-cell depletion promotes potent rejection of B16 melanoma. Clin Cancer Res 2008; 14:3156-67. [PMID: 18483384 DOI: 10.1158/1078-0432.ccr-07-4696] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate the antitumor efficacy of T-cell anergy reversal through homeostatic proliferation and regulatory T-cell (Treg) depletion in a clinically relevant murine adoptive immunotherapy model. EXPERIMENTAL DESIGN B16 melanoma cells were engineered to express the model SIYRYYGL (SIY) antigen to enable immune monitoring. Tumor-specific T cells expanded in tumor-challenged wild-type hosts but became hyporesponsive. To examine whether lymphopenia-induced homeostatic proliferation could reverse tumor-induced T-cell anergy, total splenic T cells were transferred into lymphopenic RAG2-/- mice or control P14/RAG2-/- mice. Tumor growth was measured, and SIY-specific immune responses were monitored using ELISPOT and SIY/K(b) tetramers. To determine whether Treg depletion could synergize with homeostatic proliferation, RAG2-/- mice received total or CD25-depleted T cells, followed or preceded by B16.SIY challenge. This approach was further investigated in wild-type mice lymphodepleted with sublethal total body irradiation. RESULTS Adoptive transfer of total splenic T cells into RAG2-/- mice moderately affected the growth rate of B16.SIY. As Treg expansion occurred in tumor-bearing mice, CD25+ T cells were depleted from total T cells before adoptive transfer. Interestingly, transfer of CD25-depleted T cells into RAG2-/- mice resulted in potent rejection of B16 melanoma in both prophylactic and short-term preimplanted tumor settings and was associated with maintained T-cell effector function. Using a clinically applicable approach, wild-type mice were lymphodepleted using sublethal total body irradiation, which similarly supported tumor rejection upon transfer of CD25-depleted T cells. CONCLUSIONS Our results indicate that combined CD25 depletion and homeostatic proliferation support a potent antitumor immune response--an approach with potential for clinical translation.
Collapse
Affiliation(s)
- Justin Kline
- Department of Medicine, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Brown IE, Blank C, Kline J, Kacha AK, Gajewski TF. Homeostatic Proliferation as an Isolated Variable Reverses CD8+T Cell Anergy and Promotes Tumor Rejection. THE JOURNAL OF IMMUNOLOGY 2006; 177:4521-9. [PMID: 16982889 DOI: 10.4049/jimmunol.177.7.4521] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although recent work has suggested that lymphopenia-induced homeostatic proliferation may improve T cell-mediated tumor rejection, there is little direct evidence isolating homeostatic proliferation as an experimental variable, and the mechanism by which improved antitumor immunity occurs via homeostatic proliferation is poorly understood. An adoptive transfer model was developed in which tumor-specific 2C/RAG2(-/-) TCR transgenic CD8+ T cells were introduced either into the lymphopenic environment of RAG2(-/-) mice or into P14/RAG2(-/-) mice containing an irrelevant CD8+ TCR transgenic population. RAG2(-/-), but not P14/RAG2(-/-) recipients supported homeostatic proliferation of transferred T cells as well as tumor rejection. Despite absence of tumor rejection in P14/RAG2(-/-) recipients, 2C cells did become activated, as reflected by CFSE dilution and CD44 up-regulation. However, these cells showed poor IFN-gamma and IL-2 production upon restimulation, consistent with T cell anergy and similar to the hyporesponsiveness induced by administration of soluble peptide Ag. To determine whether homeostatic proliferation could uncouple T cell anergy, anergic 2C cells were transferred into RAG(-/-) recipients, which resulted in vigorous homeostatic proliferation, recovery of IL-2 production, and acquisition of the ability to reject tumors. Taken together, our data suggest that a major mechanism by which homeostatic proliferation supports tumor rejection is by maintaining and/or re-establishing T cell responsiveness.
Collapse
Affiliation(s)
- Ian E Brown
- Department of Pathology and Department of Medicine, University of Chicago, 5841 South Maryland Avenue, Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
21
|
Chakraverty R, Eom HS, Sachs J, Buchli J, Cotter P, Hsu R, Zhao G, Sykes M. Host MHC class II+ antigen-presenting cells and CD4 cells are required for CD8-mediated graft-versus-leukemia responses following delayed donor leukocyte infusions. Blood 2006; 108:2106-13. [PMID: 16757687 PMCID: PMC1895539 DOI: 10.1182/blood-2006-03-007427] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Following bone marrow transplantation, delayed donor leukocyte infusions (DLIs) can induce graft-versus-leukemia (GVL) effects without graft-versus-host disease (GVHD). These antitumor responses are maximized by the presence of host hematopoietic antigen-presenting cells (APCs) at the time of DLI. Using a tumor-protection model, we demonstrate here that GVL activity following administration of DLIs to established mixed chimeras is dependent primarily on reactivity to allogeneic MHC antigens rather than minor histocompatibility or tumor-associated antigens. CD8(+) T-cell-dependent GVL responses against an MHC class II-negative tumor following delayed DLI require CD4(+) T-cell help and are reduced significantly when host APCs lack MHC class II expression. CD4(+) T cells primed by host APCs were required for maximal expansion of graft-versus-host reactive CD8(+) T cells but not their synthesis of IFN-gamma. In contrast, the GVL requirement for CD4(+) T-cell help was bypassed almost completely when DLI was administered to freshly irradiated recipients, indicating that the host environment is a major factor influencing the cellular mechanisms of GVL.
Collapse
Affiliation(s)
- Ronjon Chakraverty
- Transplantation Biology Research Center, Bone Marrow Transplantation Section, Massachusetts General Hospital/Harvard Medical School, MGH-East, 13th Street, Boston, MA 02129, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Fehr T, Takeuchi Y, Kurtz J, Wekerle T, Sykes M. Early regulation of CD8 T cell alloreactivity by CD4+CD25- T cells in recipients of anti-CD154 antibody and allogeneic BMT is followed by rapid peripheral deletion of donor-reactive CD8+ T cells, precluding a role for sustained regulation. Eur J Immunol 2005; 35:2679-90. [PMID: 16082727 DOI: 10.1002/eji.200526190] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
While acquisition of regulatory function by CD4+CD25- T cells has been reported following antigenic stimulation, "naturally occurring" regulatory CD4+ T cells (Treg) are believed to express CD25. We examined the mechanisms involved in peripheral CD8 T cell tolerance by induction of mixed chimerism using non-myeloablative conditioning with low-dose (3 Gy) total body irradiation and anti-CD154 antibody. Recipient CD4+ T cells were initially required for the induction of CD8 cell tolerance, but were not needed beyond 2 weeks. Depletion of CD25+ Treg prior to bone marrow transplantation and blockade of IL-2 with neutralizing antibody did not impede tolerance induction. Tolerance was dependent on CTLA4, but not on IFN-gamma. In C57BL/6 mice containing a fraction of 2C TCR transgenic CD8+ T cells, which recognize the MHC class I alloantigen Ld, induction of chimerism with L(d+), but not Ld-, bone marrow cells led to deletion of peripheral 2C+ CD8+ cells within 1 week in peripheral blood and spleen. Complete deletion required the presence of recipient CD4+ T cells. Thus, a novel, rapid form of regulation by CD4+CD25- T cells permits initial CD8 T cell tolerance in this model. Rapid peripheral deletion of donor-specific CD8 T cells precludes an ongoing requirement for CD4 T cell-mediated regulation.
Collapse
Affiliation(s)
- Thomas Fehr
- Bone Marrow Transplantation Section, Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
| | | | | | | | | |
Collapse
|
23
|
Ninova D, Dean PG, Deeds M, Stegall MD. A novel model of allograft rejection: immune reconstitution of Rag-1 recipients with 2C transgenic T-cell receptor lymphocytes. Transpl Int 2005; 18:101-10. [PMID: 15612991 DOI: 10.1111/j.1432-2277.2004.00012.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The complexity of allorejection (cell activation, homing, and effector function) makes detailed studies difficult. We have developed a model of allograft rejection using purified monoclonal alloreactive effector cells. Immunodeficient C57Bl/6-Rag-1 (H-2(b)) recipients of Balb/c (H-2(d)) islet or skin grafts were reconstituted via adoptive transfer of splenocytes from 2C transgenic mice containing CD8+ cytotoxic effector cells directed against L(d). Recipients were assessed for engraftment, activation and homing of effector cells, and ability to reject grafts. Both unpurified 2C splenocytes and purified 2C/CD8+ cells durably reconstitute immunodeficient mice. Naive 2C effector cells reject skin grafts, but not islet allografts. However, when effector cells are primed prior to reconstitution, islet allografts are rejected. Using this model, blockade of adhesion molecules LFA-1 and alpha4-integrin delayed infiltration of islet allografts and prolonged allograft survival. This model of allorejection may be useful to study the activation and homing of allospecific cells in vivo.
Collapse
Affiliation(s)
- Dora Ninova
- The Transplant Immunology Laboratory, Division of Transplant Surgery, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
24
|
Reed-Loisel LM, Sullivan BA, Laur O, Jensen PE. An MHC Class Ib-Restricted TCR That Cross-Reacts with an MHC Class Ia Molecule. THE JOURNAL OF IMMUNOLOGY 2005; 174:7746-52. [PMID: 15944277 DOI: 10.4049/jimmunol.174.12.7746] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TCR transgenic 6C5 T cells recognize an insulin B chain epitope presented by the nonclassical class I MHC molecule, Qa-1(b). Positive selection of these T cells was shown previously to require Qa-1(b). Despite dedicated specificity for Qa-1(b), evidence presented in the current study indicates that 6C5 T cells can cross-recognize a classical class I molecule. Clonal deletion was observed unexpectedly in 6C5.H-2(bxq) mice, which do not express I-E MHC class II molecules and thus should not be subject to superantigen-mediated negative selection. 6C5 T cells were observed to respond in vivo and in vitro to spleen cells from allogeneic H-2(q) mice, and specificity was mapped to D(q). Evidence was obtained for direct recognition of D(q), rather than indirect presentation of a D(q)-derived peptide presented by Qa-1(b). Polyclonal CD8(+) T cells from class Ia-deficient K(b)D(b-/-) mice reacted in vitro to allogeneic spleen cells with an apparent frequency comparable to conventional class Ia-restricted T cells. Our results provide a clear example of a Qa-1-specific TCR that can cross-react with a class Ia molecule and evidence supporting the idea that this may be a common property of T cells selected by class Ib molecules.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line, Transformed
- Cell Line, Tumor
- Cell Lineage/genetics
- Cell Lineage/immunology
- Clonal Deletion
- Crosses, Genetic
- H-2 Antigens/genetics
- H-2 Antigens/immunology
- H-2 Antigens/metabolism
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/metabolism
- Mammary Tumor Virus, Mouse/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Superantigens/genetics
- Superantigens/immunology
- Superantigens/metabolism
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Lisa M Reed-Loisel
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
25
|
Blank C, Brown I, Kacha AK, Markiewicz MA, Gajewski TF. ICAM-1 contributes to but is not essential for tumor antigen cross-priming and CD8+ T cell-mediated tumor rejection in vivo. THE JOURNAL OF IMMUNOLOGY 2005; 174:3416-20. [PMID: 15749875 DOI: 10.4049/jimmunol.174.6.3416] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
ICAM-1 has been described to provide both adhesion and costimulatory functions during T cell activation. In the setting of antitumor immunity, ICAM-1/LFA-1 interactions could be important at the level of T cell priming by APCs in draining lymph nodes as well as for transendothelial migration and tumor cell recognition at the tumor site. To determine the contribution of ICAM-1 to tumor rejection in vivo, we performed adoptive transfer of 2C TCR-transgenic/RAG2(-/-) T cells into TCRalpha(-/-) vs ICAM(-/-)/TCRalpha(-/-) recipient animals. ICAM-1-deficient mice successfully rejected HTR.C tumors expressing Ld recognized by the 2C TCR, albeit with a kinetic delay. Inasmuch as HTR.C tumor cells themselves express ICAM-1, a second model was pursued using B16-F10 melanoma cells that lack ICAM-1 expression. These cells were transduced to express the SIYRYYGL peptide recognized by the 2C TCR in the context of Kb, which is cross-presented by APCs in H-2b mice in vivo. These tumors also grew more slowly but were eventually rejected by the majority of ICAM-1(-/-)/TCRalpha(-/-) recipients. Delayed rejection in ICAM-1(-/-) mice was associated with diminished T cell priming as assessed by ELISPOT. In contrast, T cell penetration into the tumor was comparable in wild-type and ICAM-1(-/-) hosts, and adoptively transferred primed effector 2C cells rejected normally in ICAM-1(-/-) recipients. Our results suggest that ICAM-1 contributes to but is not absolutely required for CD8+ T cell-mediated tumor rejection in vivo and dominantly acts at the level of priming rather than the effector phase of the antitumor immune response.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigen Presentation
- Antigens, Neoplasm
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- In Vitro Techniques
- Intercellular Adhesion Molecule-1/genetics
- Intercellular Adhesion Molecule-1/immunology
- Mice
- Mice, Knockout
- Mice, Transgenic
- Neoplasm Transplantation
- Neoplasms, Experimental/immunology
- Receptors, Antigen, T-Cell, alpha-beta/deficiency
- Receptors, Antigen, T-Cell, alpha-beta/genetics
Collapse
Affiliation(s)
- Christian Blank
- Department of Pathology, Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
26
|
Zhang L, Lizzio EF, Chen T, Kozlowski S. Peptide immunization excludes antigen-specific T?cells from splenic lymphoid compartments. Eur J Immunol 2005; 35:776-85. [PMID: 15714585 DOI: 10.1002/eji.200425479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Using adoptive transfer of TCR-transgenic T cells, we examined the homing of transgenic T cells to splenic compartments in situ. After systemic immunization with peptide or protein antigen, the location of clonotypic T cells, cytokine production, cell surface markers, and apoptosis were assessed. There were distinct differences in the splenic homing of CD4(+) TCR-transgenic T cells in mice immunized with peptide as compared to mice immunized with whole-protein antigen. T cells in peptide-immunized mice were found almost exclusively in the splenic red pulp, but not in the T and B cell zones (white pulp), while the majority of T cells immunized with whole protein were found in the white pulp. Many more Fas ligand-expressing and apoptotic cells were present after peptide immunization than after whole-protein immunization. Localization of IL-4-, IL-2- and IFN-gamma-producing cells to the lymphocyte-containing splenic white pulp was only observed with whole-protein immunization. The unique homing and increased apoptosis of immune cells post peptide immunization may help explain the ineffectiveness of many peptide vaccines. Linkage of the same peptide epitope to a carrier protein increased white pulp T cell localization and decreased apoptosis, suggesting a strategy to enhance peptide vaccine responses.
Collapse
Affiliation(s)
- Lei Zhang
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Office of Pharmaceutical Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, USA
| | | | | | | |
Collapse
|
27
|
Palliser D, Guillen E, Ju M, Eisen HN. Multiple Intracellular Routes in the Cross-Presentation of a Soluble Protein by Murine Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:1879-87. [PMID: 15699114 DOI: 10.4049/jimmunol.174.4.1879] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Soluble heat shock fusion proteins (Hsfp) stimulate mice to produce CD8+ CTL, indicating that these proteins are cross-presented by dendritic cells (DC) to naive CD8 T cells. We report that cross-presentation of these proteins depends upon their binding to DC receptors, likely belonging to the scavenger receptor superfamily. Hsfp entered DC by receptor-mediated endocytosis that was either inhibitable by cytochalasin D or not inhibitable, depending upon aggregation state and time. Most endocytosed Hsfp was transported to lysosomes, but not the small cross-presented fraction that exited early from the endocytic pathway and required access to proteasomes and TAP. Naive CD8 T cell (2C and OT-I) responses to DC incubated with Hsfp at 1 microM were matched by incubating DC with cognate octapeptides at 1-10 pM, indicating that display of very few class I MHC-peptide complexes per DC can be sufficient for cross-presentation. With an Hsfp (heat shock protein-OVA) having peptide sequences for both CD4+ (OT-II) and CD8+ (OT-I) cells, the CD4 cells responded far more vigorously than the CD8 cells and many more class II MHC-peptide than class I MHC-peptide complexes were displayed.
Collapse
Affiliation(s)
- Deborah Palliser
- Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | |
Collapse
|
28
|
Blank C, Brown I, Peterson AC, Spiotto M, Iwai Y, Honjo T, Gajewski TF. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res 2004; 64:1140-5. [PMID: 14871849 DOI: 10.1158/0008-5472.can-03-3259] [Citation(s) in RCA: 593] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Although increased circulating tumor antigen-specific CD8(+) T cells can be achieved by vaccination or adoptive transfer, tumor progression nonetheless often occurs through resistance to effector function. To develop a model for identifying mechanisms of resistance to antigen-specific CTLs, poorly immunogenic B16-F10 melanoma was transduced to express the K(b)-binding peptide SIYRYYGL as a green fluorescent protein fusion protein that should be recognized by high-affinity 2C TCR transgenic T cells. Although B16.SIY cells expressed high levels of antigen and were induced to express K(b) in response to IFN-gamma, they were poorly recognized by primed 2C/RAG2(-/-) T cells. A screen for candidate inhibitory ligands revealed elevated PD-L1/B7H-1 on IFN-gamma-treated B16-F10 cells and also on eight additional mouse tumors and seven human melanoma cell lines. Primed 2C/RAG2(-/-)/PD-1(-/-) T cells showed augmented cytokine production, proliferation, and cytolytic activity against tumor cells compared with wild-type 2C cells. This effect was reproduced with anti-PD-L1 antibody present during the effector phase but not during the priming culture. Adoptive transfer of 2C/RAG2(-/-)/PD-1(-/-) T cells in vivo caused tumor rejection under conditions in which wild-type 2C cells or CTLA-4-deficient 2C cells did not reject. Our results support interfering with PD-L1/PD-1 interactions to augment the effector function of tumor antigen-specific CD8(+) T cells in the tumor microenvironment.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigens, CD
- Antigens, Differentiation/immunology
- B7-1 Antigen
- B7-H1 Antigen
- Blood Proteins/antagonists & inhibitors
- Blood Proteins/biosynthesis
- Blood Proteins/genetics
- Blood Proteins/physiology
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen
- Interferon-gamma/pharmacology
- Membrane Glycoproteins
- Mice
- Mice, Transgenic
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Peptides/antagonists & inhibitors
- Peptides/genetics
- Peptides/physiology
- Receptors, Antigen, T-Cell/antagonists & inhibitors
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Transduction, Genetic
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Christian Blank
- Department of Pathology, Section of Hematology/Oncology, The University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Palliser D, Ploegh H, Boes M. Myeloid Differentiation Factor 88 Is Required for Cross-Priming In Vivo. THE JOURNAL OF IMMUNOLOGY 2004; 172:3415-21. [PMID: 15004140 DOI: 10.4049/jimmunol.172.6.3415] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We describe a role for myeloid differentiation factor 88 (MyD88) in the induction of functional CTLs in vivo, in response to exogenously administered Ag, using a heat shock fusion protein, hsp65-P1, as a model Ag. CD8 T cells transferred into MyD88-deficient animals produce normal numbers of CD8 effector cells that have normal activation marker profiles after immunization with hsp65-P1. However, these CD8 T cells produced significantly less IFN-gamma and showed reduced killing activity. This reduction in activation of functional CTLs appears to be unrelated to Toll-like receptor 4 function, because in vitro hsp65-P1-experienced Toll-like receptor 4-deficient dendritic cells (DCs), but not MyD88-deficient DCs, activated CD8 T cells to a similar extent to wild-type DCs. We identify a cross-presentation defect in MyD88-deficient DCs that, when treated with hsp65-P1 fusion protein, results in surface display of fewer SIYRYYGL/class I MHC complexes. Thus, MyD88 plays a role in the developmental maturation of DCs that allows them to prime CD8 T cells through cross-presentation.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Antigen Presentation/genetics
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/metabolism
- Antigens, Differentiation/genetics
- Antigens, Differentiation/physiology
- Bacterial Proteins/administration & dosage
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- Bacterial Proteins/metabolism
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cells, Cultured
- Chaperonin 60
- Chaperonins/administration & dosage
- Chaperonins/genetics
- Chaperonins/immunology
- Chaperonins/metabolism
- Cytotoxicity, Immunologic/genetics
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Immunization/methods
- Injections, Subcutaneous
- Lymphocyte Activation/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Mycobacterium tuberculosis/immunology
- Myeloid Differentiation Factor 88
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/physiology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Deborah Palliser
- Center for Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | |
Collapse
|
30
|
Palliser D, Huang Q, Hacohen N, Lamontagne SP, Guillen E, Young RA, Eisen HN. A Role for Toll-Like Receptor 4 in Dendritic Cell Activation and Cytolytic CD8+ T Cell Differentiation in Response to a Recombinant Heat Shock Fusion Protein. THE JOURNAL OF IMMUNOLOGY 2004; 172:2885-93. [PMID: 14978090 DOI: 10.4049/jimmunol.172.5.2885] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Recombinant heat shock fusion proteins (Hsfp) injected into mice without added adjuvants can stimulate production of CD8 cytolytic T cells. Because initiation of productive immune responses generally requires dendritic cell (DC) activation, the question arises as to whether the Hsfp can activate DC independently of contaminating LPS. Using microarray analyses of DC from LPS-insensitive mice having a point mutation in Toll-like receptor 4 (Tlr4) (C3H/HeJ), or lacking Tlr4 (B10/ScNCr), we show here that unlike a LPS standard, Hsfp activated DC from HeJ mice almost as well as DC from wild-type mice. Consistent with the microarray analysis, the Hsfp's ability to activate DC was not eliminated by polymyxin B but was destroyed by proteinase K. The Hsfp did not, however, stimulate DC from mice lacking Tlr4. In vivo the CD8 T cell response to the Hsfp in mice lacking Tlr4 was impaired: the responding CD8 cells initially proliferated vigorously but their development into cytolytic effector cells was diminished. Overall, the results indicate that this Hsfp can activate DC independently of LPS but still requires Tlr4 for an optimal CD8 T cell response.
Collapse
MESH Headings
- Animals
- Bacterial Proteins/administration & dosage
- Bacterial Proteins/physiology
- Cell Differentiation/immunology
- Chaperonin 60
- Chaperonins/administration & dosage
- Chaperonins/physiology
- Cytotoxicity, Immunologic/genetics
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dose-Response Relationship, Immunologic
- Endopeptidase K/pharmacology
- Gene Expression Profiling
- Lipopolysaccharides/administration & dosage
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Oligonucleotide Array Sequence Analysis
- Polymyxin B/pharmacology
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/physiology
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Toll-Like Receptor 4
- Toll-Like Receptors
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Deborah Palliser
- Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology and the Whitehead Institute, Cambridge, MA 02139, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Blank C, Brown I, Marks R, Nishimura H, Honjo T, Gajewski TF. Absence of programmed death receptor 1 alters thymic development and enhances generation of CD4/CD8 double-negative TCR-transgenic T cells. THE JOURNAL OF IMMUNOLOGY 2004; 171:4574-81. [PMID: 14568931 DOI: 10.4049/jimmunol.171.9.4574] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Programmed death receptor 1 (PD-1) is expressed on thymocytes in addition to activated lymphocyte cells. Its ligation is thought to negatively regulate T cell activation, and PD-1(-/-) mice develop autoimmunity. To study the role of PD-1 on the development and function of a monoclonal CD8(+) T cell population, 2C TCR-transgenic/recombination-activating gene 2(-/-)/PD-1(-/-) mice were generated. Unexpectedly, approximately 30% of peripheral T cells in these mice were CD4/CD8 double negative (DN). Although the DN cells were not activated by Ag-expressing APCs, they functioned normally in response to anti-CD3/anti-CD28. These cells had a naive surface phenotype and lacked expression of NK1.1, B220, and gammadelta TCR; and the majority did not up-regulate CD8alphaalpha expression upon activation, arguing that they are not predominantly diverted gammadelta-lineage cells. The thymus was studied in detail to infer the mechanism of generation of DN peripheral T cells. Total thymus cellularity was reduced in 2C TCR-transgenic/recombination-activating gene 2(-/-)/PD-1(-/-) mice, and a relative increase in DN cells and decrease in double-positive (DP) cells were observed. Increased annexin V(+) cells among the DP population argued for augmented negative selection in PD-1(-/-) mice. In addition, an increased fraction of the DN thymocytes was HSA negative, suggesting that they had undergone positive selection. This possibility was supported by decreased emergence of DN PD-1(-/-) 2C cells in H-2(k) bone marrow chimera recipients. Our results are consistent with a model in which absence of PD-1 leads to greater negative selection of strongly interacting DP cells as well as increased emergence of DN alphabeta peripheral T cells.
Collapse
MESH Headings
- Animals
- Antigens/pharmacology
- Antigens, Surface/biosynthesis
- Antigens, Surface/genetics
- Antigens, Surface/physiology
- Apoptosis Regulatory Proteins
- CD28 Antigens/immunology
- CD3 Complex/immunology
- CD4 Antigens/biosynthesis
- CD4 Antigens/genetics
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Division/genetics
- Cell Division/immunology
- Cell Line, Tumor
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- Immune Sera/pharmacology
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Lymphocyte Count
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Programmed Cell Death 1 Receptor
- Receptors, Antigen, T-Cell, gamma-delta/biosynthesis
- Receptors, Antigen, T-Cell, gamma-delta/deficiency
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Christian Blank
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
32
|
Block MS, Hansen MJ, Van Keulen VP, Pease LR. MHC class I gene conversion mutations alter the CD8 T cell repertoire. THE JOURNAL OF IMMUNOLOGY 2004; 171:4006-10. [PMID: 14530320 DOI: 10.4049/jimmunol.171.8.4006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
MHC class I molecules are highly polymorphic within populations. This diversity is thought to be the result of selective maintenance of new class I alleles formed by gene conversion. It has been proposed that rare alleles are maintained by their ability to confer resistance to common pathogens. Investigation has focused on differences in the presentation of foreign Ags by class I alleles, but the majority of peptides presented by class I molecules are self peptides used in shaping the naive T cell repertoire. We propose that the key substrate for the natural selection of class I gene conversion variants is the diversity in immune potential formed by new alleles. We show that T cells compete with each other for niches in the thymus and spleen during development, and that competition between different clones is dramatically affected by class I mutations. We also show that peripheral naive T cells proliferate preferentially in the presence of the class I variant that directed T cell development. The data argue that class I gene conversion mutations dramatically affect both the development and the maintenance of the naive CD8 T cell repertoire.
Collapse
Affiliation(s)
- Matthew S Block
- Department of Immunology, Mayo Medical and Graduate Schools, Mayo Clinic Rochester, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
33
|
Richards DM, Dalheimer SL, Hertz MI, Mueller DL. Trachea Allograft Class I Molecules Directly Activate and Retain CD8+ T Cells That Cause Obliterative Airways Disease. THE JOURNAL OF IMMUNOLOGY 2003; 171:6919-28. [PMID: 14662899 DOI: 10.4049/jimmunol.171.12.6919] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human T cells responding against transplanted allogeneic lung tissue have been implicated in late graft failure secondary to obliterative bronchiolitis. This obliterative airways disease (OAD) also develops in heterotopic murine tracheal allografts in association with graft infiltration by both CD8(+) and CD4(+) T cells. To date, there has been little evidence to suggest that directly alloreactive CD8(+) T cells either promote chronic rejection or lead to the development of OAD following airway allotransplantation. Using L(d)-specific TCR-Tg 2C CD8(+) T cells adoptively transferred into wild-type B6 (H-2(b)) mice and the transplantation of BALB/c (H-2(d)) tracheal allografts, we now show that the direct recognition of donor-specific class I MHC molecules by host CD8(+) T cells leads to their activation, clonal expansion within the graft, and differentiation to an effector phenotype with the capacity to induce airway fibrosis. In addition, these experiments demonstrate that ongoing direct alloantigen recognition within the transplanted airway tissue is necessary for the recruitment and retention of these directly alloreactive CD8(+) T cells. Thus, these experiments are the first to definitively show a role for directly alloreactive CD8(+) T cells in the chronic rejection that leads to OAD.
Collapse
Affiliation(s)
- David M Richards
- Department of Medicine and. Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
34
|
Mapara MY, Kim YM, Marx J, Sykes M. Donor lymphocyte infusion-mediated graft-versus-leukemia effects in mixed chimeras established with a nonmyeloablative conditioning regimen: extinction of graft-versus-leukemia effects after conversion to full donor chimerism. Transplantation 2003; 76:297-305. [PMID: 12883182 DOI: 10.1097/01.tp.0000072014.83469.2d] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We investigated an approach to separating graft-versus-lymphoma (GVL) effects from graft-versus-host disease (GVHD) in mice receiving a nonmyeloablative conditioning regimen allowing establishment of mixed hematopoietic chimerism. METHODS We evaluated the ability of donor lymphocyte infusions (DLI) to mediate GVL effects without GVHD in mixed chimeras prepared with cyclophosphamide, anti-T-cell antibodies, and thymic irradiation. To examine the fate of GVH-reactive donor CD8+ T cells, we used the 2C T-cell receptor (TCR) transgenic mouse strain, which carries an Ld-specific transgenic TCR on the B6 background. RESULTS Administration of DLI on day 35 post-BMT led to conversion from mixed to full donor chimerism and mediated a powerful GVL effect with complete protection (100% survival) against mortality induced by a host-type lymphoma (EL4) administered 7 days later (100% mortality in non-DLI controls; P<0.001). No GVHD occurred in DLI recipients. Rechallenging the surviving DLI recipients, which had converted to full chimerism, with the same tumor dose 17 weeks later led to rapid tumor mortality. Long-term DLI recipients had anti-host proliferative responses, but not CTL responses in vitro. When given as DLI together with wild-type spleen cells, marked expansion of GVH-reactive 2C CD8+ T cells was observed on day 10, followed by a marked decline in their numbers by week 10 post-DLI. CONCLUSIONS Nonmyeloablative induction of mixed chimerism followed by administration of DLI can mediate powerful GVL effects. The late loss of DLI-mediated GVL effects may reflect the eventual loss of donor-derived GVH-reactive CTL, which occurs in association with conversion to full donor chimerism.
Collapse
Affiliation(s)
- Markus Y Mapara
- Transplantation Biology Research Center, Bone Marrow Transplantation Section, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
35
|
Young KJ, DuTemple B, Phillips MJ, Zhang L. Inhibition of graft-versus-host disease by double-negative regulatory T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:134-41. [PMID: 12816991 DOI: 10.4049/jimmunol.171.1.134] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pretransplant infusion of lymphocytes that express a single allogeneic MHC class I Ag has been shown to induce tolerance to skin and heart allografts that express the same alloantigens. In this study, we demonstrate that reconstitution of immunoincompetent mice with spleen cells from MHC class I L(d)-mismatched donors does not cause graft-vs-host disease (GVHD). Recipient mice become tolerant to skin allografts of lymphocyte donor origin while retaining immunity to third-party alloantigens. The mechanism involves donor-derived CD3(+)CD4(-)CD8(-) double-negative T regulatory (DN Treg) cells, which greatly increase and form the majority of T lymphocytes in the spleen of recipient mice. DN Treg cells isolated from tolerant recipient mice can suppress the proliferation of syngeneic antihost CD8(+) T cells in vitro. Furthermore, we demonstrate that DN Treg cells can be generated in vitro by stimulating them with MHC class I L(d)-mismatched lymphocytes. These in vitro generated L(d)-specific DN Treg cells are able to down-regulate the activity of antihost CD8(+) T cells in vitro by directly killing activated CD8(+) T cells. Moreover, infusing in vitro generated L(d)-mismatched DN Treg cells prevented the development of GVHD caused by allogeneic CD8(+) T cells. Together these data demonstrate that infusion of single MHC class I locus-mismatched lymphocytes may induce donor-specific transplantation tolerance through activation of DN Treg cells, which can suppress antihost CD8(+) T cells and prevent the development of GVHD. This finding indicates that using single class I locus-mismatched grafts may be a viable alternative to using fully matched grafts in bone marrow transplantation.
Collapse
MESH Headings
- Animals
- CD3 Complex/biosynthesis
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/transplantation
- Cell Division/genetics
- Cell Division/immunology
- Clone Cells
- Coculture Techniques
- Cytotoxicity, Immunologic/genetics
- Graft vs Host Disease/genetics
- Graft vs Host Disease/immunology
- Graft vs Host Disease/pathology
- Graft vs Host Disease/prevention & control
- H-2 Antigens/administration & dosage
- H-2 Antigens/genetics
- H-2 Antigens/immunology
- Histocompatibility Antigen H-2D
- Histocompatibility Testing
- Lymphocyte Transfusion
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, SCID
- Mice, Transgenic
- Skin Transplantation/adverse effects
- Skin Transplantation/immunology
- Spleen/cytology
- Spleen/immunology
- Spleen/transplantation
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Transplantation Tolerance/genetics
- Transplantation, Isogeneic/immunology
- Transplantation, Isogeneic/pathology
Collapse
Affiliation(s)
- Kevin J Young
- Department of Laboratory Medicine and Pathobiology, Multi Organ Transplantation Program, Toronto General Research Institute, University Health Network, and Department of Immunology, University of Toronto, Toronto, Canada
| | | | | | | |
Collapse
|
36
|
Hornell TMC, Myers N, Hansen TH, Connolly JM. Homology between an alloantigen and a self MHC allele calibrates the avidity of the alloreactive T cell repertoire independent of TCR affinity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4506-14. [PMID: 12707327 DOI: 10.4049/jimmunol.170.9.4506] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The self-restricted T cell repertoire exhibits a high frequency of alloreactivity. Because these alloreactive T cells are derived from the pool of cells selected on several different self MHC alleles, it is unknown how development of the alloantigenic repertoire is influenced by homology between a self MHC allele and an alloantigen. To address this, we used the 2C transgenic TCR that is selected by K(b), is alloreactive for L(d), and cross-reacts with L(q). L(q) is highly homologous to L(d) and binds several of the same peptide ligands, including p2Ca, the peptide recognized by 2C. We find that L(d)/p2Ca is a high avidity agonist ligand, whereas L(q)/p2Ca is a low avidity agonist ligand for 2C T cells. When mice transgenic for the 2C TCR are bred to L(q)-expressing mice, 2C(+) T cells develop; however, they express lower levels of either the 2C TCR or CD8 and require a higher L(d)/p2Ca ligand density to be activated than 2C(+) T cells selected by K(b). Furthermore, the 2C T cells selected in the presence of L(q) fail to detect L(q)/p2Ca complexes even at high ligand density. Thus, despite possessing the identical TCR, there is a functional avidity difference between 2C(+) T cells selected in the presence of L(q) vs K(b). These data provide evidence that homology between the selecting ligand and an alloantigen can influence the avidity of the T cell repertoire for the alloantigen, and suggest that thymic selection can fine tune T cell avidity independent of intrinsic TCR affinity.
Collapse
MESH Headings
- Alleles
- Amino Acid Sequence
- Animals
- CD8 Antigens/biosynthesis
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line
- Cytotoxicity Tests, Immunologic
- Cytotoxicity, Immunologic
- H-2 Antigens/biosynthesis
- H-2 Antigens/chemistry
- H-2 Antigens/genetics
- H-2 Antigens/metabolism
- Histocompatibility Antigen H-2D
- Isoantigens/metabolism
- Ligands
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Transgenic
- Molecular Sequence Data
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Protein Binding/genetics
- Protein Binding/immunology
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell/metabolism
- Sequence Homology, Amino Acid
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Tara M C Hornell
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
37
|
Abstract
When T-cell clones were first grown in long-term cell culture, each clone was considered to be capable of displaying a limited range of functional activities, constrained by the clones' coreceptor, CD4 or CD8, and the specificity of its antigen-specific receptor (TCR) for one or a few peptides in association with a class I or class II MHC molecule. Subsequent studies, especially with transgenic mice, have shown, however, that T cells expressing the same receptor can be obtained in a variety of differentiated states, including naïve cells, activated effector cells, memory cells, and anergic or tolerant cells, as well as cells with or without a coreceptor. In each of these states T cells can display distinctly different responses to the peptide-MHC (pepMHC) complexes the TCR recognizes. Recently, memory T cells have received considerable attention, in part because of the likelihood that they confer long-term protective immunity against diverse infectious agents and possibly against some forms of cancer. Here we review some recent studies that our colleagues and we have carried out on memory CD8(+) T cells. These studies have made extensive use of cells that express the TCR called 2C. The diverse set of cells expressing the 2C TCR, in mice and in cultured clones and cell lines, are referred to as the 2C system. Before reviewing the studies of memory cells, we summarize the 2C system's main features, including evidence that a large and diverse array of pepMHC complexes, involving at least four class I MHC proteins, can stimulate TCR-mediated responses of 2C cells.
Collapse
Affiliation(s)
- Jianzhu Chen
- Center for Cancer Research and Department of Biology, E17-128, 40 Ames Street, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | | | |
Collapse
|
38
|
Lee PUY, Kranz DM. Allogeneic and syngeneic class I MHC complexes drive the association of CD8 and TCR on 2C T cells. Mol Immunol 2003; 39:687-95. [PMID: 12531280 DOI: 10.1016/s0161-5890(02)00259-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In most cases, cytotoxic T cell activation is dependent on the interaction of the T cell receptor (TCR) and CD8 with MHC class I molecules. In the CD8(+) T cell system based on the mouse cytotoxic T cell clone 2C, recognition of the allogeneic MHC L(d) exhibits a less significant role for CD8 than recognition of the syngeneic MHC K(b). Here, we examined whether this difference is related to the relative abilities of the two pepMHC complexes to drive the association of CD8 and TCR on the T cell surface. We show that both the syngeneic and allogeneic pepMHC induced association of CD8 and TCR, as revealed by fluorescence resonance energy transfer (FRET). Thus, the orientation of the syngeneic and allogeneic ligands when bound to the same TCR both allow CD8 to be recruited to the TCR complex. The conserved diagonal orientation of TCRs on different pepMHC ligands may facilitate such associations. The FRET results are consistent with the known binding properties and the CD8 involvement of the two different TCR:pepMHC interactions.
Collapse
Affiliation(s)
- Peter U Y Lee
- Department of Biochemistry, University of Illinois, 600 S. Mathews Ave., Urbana, IL 61801, USA
| | | |
Collapse
|
39
|
Zhang L, Lizzio EF, Gubina E, Chen T, Mostowski H, Kozlowski S. Organ-specific cytokine polarization induced by adoptive transfer of transgenic T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5514-21. [PMID: 12421927 DOI: 10.4049/jimmunol.169.10.5514] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There are two distinct phenotypes of T cell cytokine responses that lead to different effector functions and different outcomes in disease processes. Although evidence suggests a possible role of the local microenvironment in the differentiation or localization of T cells with these phenotypes, there are no examples of divergent T cell cytokine phenotypes with the same Ag specificity concurrently existing in different tissue compartments. Using a CD8(+) T cell adoptive transfer model for graft-vs-host disease, we demonstrate that a potent type 2 cytokine response develops in the spleen while a potent type 1 cytokine response simultaneously develops in the testis. These experiments demonstrate for the first time that cytokine production can be oppositely polarized in different organs of the same individual. This may have important implications for organ-specific pathology in infection or autoimmunity: infections or autoimmune diseases that affect multiple organs may have heterogeneity in tissue cytokine responses that is not revealed in systemic lymphocyte cytokine responses. Therefore, attempts to modulate the immune response phenotype may ameliorate pathology in one organ while exacerbating pathology in another.
Collapse
Affiliation(s)
- Lei Zhang
- Divisions of Monoclonal Antibodies, Center for Biologics Evaluation and Research, Food and Drug Administration, Building 29B, 29 Lincoln Drive, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
40
|
Pepperl-Klindworth S, Frankenberg N, Plachter B. Development of novel vaccine strategies against human cytomegalovirus infection based on subviral particles. J Clin Virol 2002; 25 Suppl 2:S75-85. [PMID: 12361759 DOI: 10.1016/s1386-6532(02)00099-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Pre- and perinatal human cytomegalovirus (HCMV) infection remains one of the major causes of mental defects and sensineural hearing loss in children. In addition, it is a prominent infectious complication in immunosuppressed individuals such as AIDS patients or transplant recipients. Therefore, the development of an HCMV vaccine has been given top priority by health care institutions. STUDY DESIGN Defective subviral particles of HCMV, termed Dense Bodies (DB) contain the dominant target antigens for humoral and cellular immune responses elicited during natural infection. These enveloped particles are released from infected culture cells and can be purified by gradient centrifugation. DB were analyzed for their ability to induce virus neutralizing antibodies and cytotoxic T cells (CTL) after immunization of mice. RESULTS Purified DB entered human and murine hematopoetic and fibroblast cells very efficiently, thereby delivering their protein content into the cytoplasm. The cellular uptake was abrogated after sonication and freeze-thawing of the particles, indicating that the integrity of the viral envelope was important for this process. DB immunization of HLA-A2.K(b) transgenic mice induced significant CTL responses in the absence of viral gene expression and without the use of adjuvant. Induction of cytolytic cells by DB was sensitive to sonication and freeze-thawing as determined by CD3epsilon -redirected lysis analysis. In accordance with that, induction of virus neutralizing antibodies was much more effective when untreated DB were used as immunogen. CONCLUSIONS DB provide a promising basis for the development of a subunit vaccine against HCMV infection. The ability to genetically engineer HCMV provides a rationale to optimize such a vaccine and to develop concepts for future multicomponent vaccines.
Collapse
Affiliation(s)
- Sandra Pepperl-Klindworth
- Institut für Virologie, Johannes Gutenberg-Universität Mainz, Obere Zahlbacher Str. 67, 55101 Mainz, Germany
| | | | | |
Collapse
|
41
|
Halamay KE, Kirkman RL, Sun L, Yamada A, Fragoso RC, Shimizu K, Mitchell RN, McKay DB. CD8 T cells are sufficient to mediate allorecognition and allograft rejection. Cell Immunol 2002; 216:6-14. [PMID: 12381345 DOI: 10.1016/s0008-8749(02)00530-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Different T cell subsets may play different roles in allorecognition and allograft rejection. It has been suggested that CD8 T cells can only initiate rejection with help from CD4 T cells. Since CD8 T cells may have different requirements for allorecognition and for costimulation, it is important to clarify the role of CD8 cells in rejection. We examined the role of CD8 cells in allorecognition using a TCR transgenic mouse transplantation model. In our study, CD8 cells were able to recognize alloantigens and reject allografts in the absence of help from CD4 T cells. Furthermore our study provides a model to study the mechanisms of CD8-mediated allograft rejection. It may be important in the future, to consider that CD8 T cells may need to be targeted independently of CD4 T cells in strategies used to prevent rejection and induce tolerance.
Collapse
Affiliation(s)
- Kate E Halamay
- Division of Pediatrics, Department of Pediatric Oncology, Dana Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Borenstein SH, Tao KS, West LJ, Chamberlain JW. Extrathymic deletion of CD8+ alloreactive T cells in a transgenic T cell receptor model of neonatal tolerance. Transplantation 2001; 72:1807-16. [PMID: 11740393 DOI: 10.1097/00007890-200112150-00017] [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: 11/26/2022]
Abstract
BACKGROUND Immunological tolerance to foreign antigen is most easily achieved during the neonatal period. Although deletion of T cells has been demonstrated in neonatal tolerance models in which donor and recipient express different MHC class II molecules, the requirement for deletion in MHC class I-disparate models is less clear. To address this issue, we used as recipient the T cell receptor (TCR) transgenic mouse (TgM) strain 2C in which the majority of CD8+ T cells express a single alpha/beta TCR alloreactive to H-2Ld, thus facilitating direct monitoring of the class I alloreactive population. METHODS Newborn (less than 24 hr of age) 2C TgM received injections i.v.with syngeneic C57BL/6J (H-2b) (B6) or semiallogeneic (B6xDBA)F1 (H-2bd; H-2Ld+) splenocytes. Adults were subsequently analyzed in terms of tolerance, deletion of 2C+ T cells, and chimerism. RESULTS The results showed that semiallogeneic-, but not syngeneic-, injected neonates were unresponsive as adults to H-2Ld-expressing target cells in vitro and the majority of these mice accepted H-2Ld+ skin grafts. Delaying the injection to 72 hr after birth or reducing the number of cells injected essentially abolished in vivo unresponsiveness in 2C recipients. Thus, the 2C TCR Tg model demonstrates the characteristics typical of neonatal tolerance. Injection of 2C neonates within 24 hr of birth with semiallogeneic versus syngeneic cells led to more than a 12-fold reduction of CD8+ 2C+ T cells in adult spleen and LNCs. In contrast, deletion of CD8+ 2C+ cells in adult thymus was not consistently observed. Based on MHC class II expression to distinguish donor (I-E+) and recipient (I-E-) cells, semiallogeneic-injected mice were chimeric in spleens and lymph nodes (LNs). CONCLUSIONS These results demonstrate that neonatal MHC class I tolerance in the adult is associated with low level hematopoietic chimerism and extrathymic deletion of alloreactive CD8+ T cells.
Collapse
Affiliation(s)
- S H Borenstein
- Research Institute, Program in Infection, Immunity, Injury and Repair, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
43
|
Block MS, Johnson AJ, Mendez-Fernandez Y, Pease LR. Monomeric class I molecules mediate TCR/CD3 epsilon/CD8 interaction on the surface of T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:821-6. [PMID: 11441088 DOI: 10.4049/jimmunol.167.2.821] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Both CD8 and the TCR bind to MHC class I molecules during physiologic T cell activation. It has been shown that for optimal T cell activation to occur, CD8 must be able to bind the same class I molecule that is bound by the TCR. However, no direct evidence for the class I-dependent association of CD8 and the TCR has been demonstrated. Using fluorescence resonance energy transfer, we show directly that a single class I molecule causes TCR/CD8 interaction by serving as a docking molecule for both CD8 and the TCR. Furthermore, we show that CD3epsilon is brought into close proximity with CD8 upon TCR/CD8 association. These interactions are not dependent on the phosphorylation events characteristic of T cell activation. Thus, MHC class I molecules, by binding to both CD8 and the TCR, mediate the reorganization of T cell membrane components to promote cellular activation.
Collapse
Affiliation(s)
- M S Block
- Department of Immunology, Mayo Graduate and Medical Schools, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
44
|
Holtappels R, Podlech J, Grzimek NK, Thomas D, Pahl-Seibert MF, Reddehase MJ. Experimental preemptive immunotherapy of murine cytomegalovirus disease with CD8 T-cell lines specific for ppM83 and pM84, the two homologs of human cytomegalovirus tegument protein ppUL83 (pp65). J Virol 2001; 75:6584-600. [PMID: 11413326 PMCID: PMC114382 DOI: 10.1128/jvi.75.14.6584-6600.2001] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD8 T cells are the principal antiviral effectors controlling cytomegalovirus (CMV) infection. For human CMV, the virion tegument protein ppUL83 (pp65) has been identified as a source of immunodominant peptides and is regarded as a candidate for cytoimmunotherapy and vaccination. Two sequence homologs of ppUL83 are known for murine CMV, namely the virion protein ppM83 (pp105) expressed late in the viral replication cycle and the nonstructural protein pM84 (p65) expressed in the early phase. Here we show that ppM83, unlike ppUL83, is not delivered into the antigen presentation pathway after virus penetration before or in absence of viral gene expression, while other virion proteins of murine CMV are processed along this route. In cytokine secretion-based assays, ppM83 and pM84 appeared to barely contribute to the acute immune response and to immunological memory. Specifically, the frequencies of M83 and M84 peptide-specific CD8 T cells were low and undetectable, respectively. Nonetheless, in a murine model of cytoimmunotherapy of lethal CMV disease, M83 and M84 peptide-specific cytolytic T-cell lines proved to be highly efficient in resolving productive infection in multiple organs of cell transfer recipients. These findings demonstrate that proteins which fail to prime a quantitatively dominant immune response can nevertheless represent relevant antigens in the effector phase. We conclude that quantitative and qualitative immunodominance are not necessarily correlated. As a consequence of these findings, there is no longer a rationale for considering T-cell abundance as the key criterion for choosing specificities to be included in immunotherapy and immunoprophylaxis of CMV disease and of viral infections in general.
Collapse
Affiliation(s)
- R Holtappels
- Institute for Virology, Johannes Gutenberg University, Hochhaus am Augustusplatz, 55101 Mainz, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Maurice MM, Gould DS, Carroll J, Vugmeyster Y, Ploegh HL. Positive selection of an MHC class-I restricted TCR in the absence of classical MHC class I molecules. Proc Natl Acad Sci U S A 2001; 98:7437-42. [PMID: 11404484 PMCID: PMC34687 DOI: 10.1073/pnas.141143298] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2001] [Indexed: 11/18/2022] Open
Abstract
The H-2Ld alloreactive 2C T cell receptor (TCR) is commonly considered as being positively selected on the H-2Kb molecule. Surprisingly, 2C TCR+ CD8+ single-positive T cells emerge in massive numbers in fetal thymic organ culture originating from 2C transgenic, H-2KbD(b-/-) (2C+KbD(b-/-)) but not in fetal thymic organ culture from beta2-microglobulin(-/-) 2C transgenic animals. Mature CD8+ T cells are observed in newborn but not in adult 2C+KbD(b-/-) mice. These CD8+ T cells express the alpha4beta7 integrin, which allows them to populate the intestine, a pattern of migration visualized by intrathymic injection of FITC and subsequent accrual of FITC-labeled lymphocytes in the gut. We conclude that the 2C TCR is reactive not only with H-2Ld and H-2Kb, but also with nonclassical MHC class I products to enable positive selection of 2C+ T cells in the fetal and newborn thymus and to support their maintenance in the intestine.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- CD8-Positive T-Lymphocytes/immunology
- Crosses, Genetic
- Genes, MHC Class I
- H-2 Antigens/genetics
- H-2 Antigens/immunology
- Histocompatibility Antigen H-2D
- Major Histocompatibility Complex
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Thymus Gland/immunology
- beta 2-Microglobulin/deficiency
- beta 2-Microglobulin/genetics
- beta 2-Microglobulin/physiology
Collapse
Affiliation(s)
- M M Maurice
- Department of Pathology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
46
|
Holtappels R, Pahl-Seibert MF, Thomas D, Reddehase MJ. Enrichment of immediate-early 1 (m123/pp89) peptide-specific CD8 T cells in a pulmonary CD62L(lo) memory-effector cell pool during latent murine cytomegalovirus infection of the lungs. J Virol 2000; 74:11495-503. [PMID: 11090146 PMCID: PMC112429 DOI: 10.1128/jvi.74.24.11495-11503.2000] [Citation(s) in RCA: 185] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2000] [Accepted: 09/19/2000] [Indexed: 11/20/2022] Open
Abstract
Interstitial cytomegalovirus (CMV) pneumonia is a clinically relevant complication in recipients of bone marrow transplantation (BMT). Recent data for a model of experimental syngeneic BMT and concomitant infection of BALB/c mice with murine CMV (mCMV) have documented the persistence of tissue-resident CD8 T cells after clearance of productive infection of the lungs (J. Podlech, R. Holtappels, M.-F. Pahl-Seibert, H.-P. Steffens, and M. J. Reddehase, J. Virol. 74:7496-7507, 2000). It was proposed that these cells represent antiviral "standby" memory cells whose functional role might be to help prevent reactivation of latent virus. The pool of pulmonary CD8 T cells was composed of two subsets defined by the T-cell activation marker L-selectin (CD62L): a CD62L(hi) subset of quiescent memory cells, and a CD62L(lo) subset of recently resensitized memory-effector cells. In this study, we have continued this line of investigation by quantitating CD8 T cells specific for the three currently published antigenic peptides of mCMV: peptide YPHFMPTNL processed from the immediate-early protein IE1 (pp89), and peptides YGPSLYRRF and AYAGLFTPL, derived from the early proteins m04 (gp34) and M84 (p65), respectively. IE1-specific CD8 T cells dominated in acute-phase pulmonary infiltrates and were selectively enriched in latently infected lungs. Notably, most IE1-specific CD8 T cells were found to belong to the CD62L(lo) subset representing memory-effector cells. This finding is in accordance with the interpretation that IE1-specific CD8 T cells are frequently resensitized during latent infection of the lungs and may thus be involved in the maintenance of mCMV latency.
Collapse
Affiliation(s)
- R Holtappels
- Institute for Virology, Johannes Gutenberg University, 55101 Mainz, Germany
| | | | | | | |
Collapse
|
47
|
Shusta EV, Holler PD, Kieke MC, Kranz DM, Wittrup KD. Directed evolution of a stable scaffold for T-cell receptor engineering. Nat Biotechnol 2000; 18:754-9. [PMID: 10888844 DOI: 10.1038/77325] [Citation(s) in RCA: 180] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Here we have constructed a single-chain T-cell receptor (scTCR) scaffold with high stability and soluble expression efficiency by directed evolution and yeast surface display. We evolved scTCRs in parallel for either enhanced resistance to thermal denaturation at 46 degrees C, or improved intracellular processing at 37 degrees C, with essentially equivalent results. This indicates that the efficiency of the consecutive kinetic processes of membrane translocation, protein folding, quality control, and vesicular transport can be well predicted by the single thermodynamic parameter of thermal stability. Selected mutations were recombined to create an scTCR scaffold that was stable for over an hour at 65 degrees C, had solubility of over 4 mg ml(-1), and shake-flask expression levels of 7.5 mg l(-1), while retaining specific ligand binding to peptide-major histocompatibility complexes (pMHCs) and bacterial superantigen. These properties are comparable to those for stable single-chain antibodies, but are markedly improved over existing scTCR constructs. Availability of this scaffold allows engineering of high-affinity soluble scTCRs as antigen-specific antagonists of cell-mediated immunity. Moreover, yeast displaying the scTCR formed specific conjugates with antigen-presenting cells (APCs), which could allow development of novel cell-to-cell selection strategies for evolving scTCRs with improved binding to various pMHC ligands in situ.
Collapse
Affiliation(s)
- E V Shusta
- Department of Chemical Engineering, University of Illinois, Urbana, IL 61801, USA
| | | | | | | | | |
Collapse
|
48
|
Pepperl S, Münster J, Mach M, Harris JR, Plachter B. Dense bodies of human cytomegalovirus induce both humoral and cellular immune responses in the absence of viral gene expression. J Virol 2000; 74:6132-46. [PMID: 10846097 PMCID: PMC112112 DOI: 10.1128/jvi.74.13.6132-6146.2000] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2000] [Accepted: 04/10/2000] [Indexed: 11/20/2022] Open
Abstract
Infection of fibroblast cell cultures with human cytomegalovirus (HCMV) leads to the production of significant amounts of defective enveloped particles, termed dense bodies (DB). These noninfectious structures contain major antigenic determinants which are responsible for induction of both the humoral and the cellular immune response against HCMV. We tested the hypothesis that, by virtue of their unique antigenic and structural properties, DB could induce a significant immune response in the absence of infectious virus. Mice were immunized with gradient-purified DB, which were either left untreated or subjected to sequential rounds of sonication and freeze-thawing to prevent cellular entry. Titers of neutralizing antibodies induced by DB were in a range comparable to levels present in convalescent human sera. The virus-neutralizing antibody response was surprisingly durable, with neutralizing antibodies detected 12 months following primary immunization. The HCMV-specific major histocompatibility complex class I-restricted cytolytic T-cell (CTL) response was assayed using mice transgenic for the human HLA-A2 molecule. Immunization with DB led to high levels of HCMV-specific CTL in the absence of de novo viral protein synthesis. Maximal total cytolytic activity in mice immunized with DB was nearly as efficient as the cytolytic activity induced by a standard immunization with murine cytomegalovirus. Furthermore, DB induced a typical T-helper 1 (Th1)-dominated immune response in mice, as determined by cytokine and immunoglobulin G isotype analysis. Induction of humoral and cellular immune responses was achieved without the concomitant use of adjuvant. We thus propose that DB can serve as a basis for the future development of a recombinant nonreplicating vaccine against HCMV. Finally, such particles could be engineered for efficient delivery of antigens from other pathogens to the immune system.
Collapse
Affiliation(s)
- S Pepperl
- Institute for Virology, University of Mainz, Germany
| | | | | | | | | |
Collapse
|
49
|
Lee PU, Churchill HR, Daniels M, Jameson SC, Kranz DM. Role of 2CT cell receptor residues in the binding of self- and allo-major histocompatibility complexes. J Exp Med 2000; 191:1355-64. [PMID: 10770802 PMCID: PMC2193126 DOI: 10.1084/jem.191.8.1355] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/1999] [Accepted: 02/10/2000] [Indexed: 11/13/2022] Open
Abstract
T cell clone 2C recognizes the alloantigen L(d) and the positive selecting major histocompatibility complex (MHC), K(b). To explore the molecular basis of T cell antigen receptor (TCR) binding to different peptide/MHC (pMHC) complexes, we performed alanine scanning mutagenesis of the 2C TCR. The TCR energy maps for QL9/L(d) and SIYR/K(b) were remarkably similar, in that 16 of 41 Valpha and Vbeta alanine mutants showed reduced binding to both ligands. Several TCR residues varied in the magnitude of energy contributed to binding the two ligands, indicating that there are also unique interactions. Residues in complementarity determining region 3alpha showed the most notable differences in binding energetics among the ligands QL9/L(d), SIYR/K(b), and the clonotypic antibody 1B2. Various lines of evidence suggest that these differences relate to the mobility of this loop and point to the key role of conformational dynamics in pMHC recognition.
Collapse
Affiliation(s)
- Peter U.Y. Lee
- Department of Biochemistry, University of Illinois, Urbana, Illinois 61801
| | | | - Mark Daniels
- Department of Laboratory Medicine and Pathology and the Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Stephen C. Jameson
- Department of Laboratory Medicine and Pathology and the Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - David M. Kranz
- Department of Biochemistry, University of Illinois, Urbana, Illinois 61801
| |
Collapse
|
50
|
McKay DB, Irie HY, Hollander G, Ferrara JLMF, Strom TB, Li Y, Burakoff SJ. Antigen-Induced Unresponsiveness Results in Altered T Cell Signaling. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.12.6455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Pretransplant exposure to allogeneic lymphocytes can result in donor-specific unresponsiveness and prolonged allograft survival. Intracellular signaling events have been described in anergic T cell clones, but the biochemical events underlying in vivo induced unresponsiveness have not been studied in detail. We employed a TCR transgenic mouse, bearing the 2C TCR, providing adequate numbers of homogenous peripheral T cells to study biochemical aspects of T cell unresponsiveness in vivo. 2C mice exposed to semiallogeneic lymphocytes (H-2b × H-2d) experienced prolonged H-2d cardiac allograft survival, and cells from these mice did not proliferate or make IL-2 in response to alloantigen (H-2d). Importantly, there were marked differences in TCR-associated tyrosine phosphorylation activation patterns. The targets for the unresponsive state appear to be diminished Lck activation and absent ZAP-70 and LAT (linker for activation of T cells) phosphorylation. Our study demonstrates that Ag-induced tolerance in vivo is accompanied by altered early TCR-mediated signaling events.
Collapse
Affiliation(s)
- Dianne B. McKay
- *Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115
- †Renal Division and Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Hanna Y. Irie
- *Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115
| | - Georg Hollander
- ‡Department of Pediatric Research, Kantonsspital, Basel, Switzerland
| | | | - Terry B. Strom
- ¶Renal Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115
| | - YongSheng Li
- ¶Renal Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115
| | - Steven J. Burakoff
- *Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115
| |
Collapse
|