51
|
Dharmasiri U, Isenberg SL, Glish GL, Armistead PM. Differential ion mobility spectrometry coupled to tandem mass spectrometry enables targeted leukemia antigen detection. J Proteome Res 2014; 13:4356-62. [PMID: 25184817 PMCID: PMC4184456 DOI: 10.1021/pr500527c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Differential ion mobility spectrometry (DIMS) can be used as a filter to remove undesired background ions from reaching the mass spectrometer. The ability to use DIMS as a filter for known analytes makes DIMS coupled to tandem mass spectrometry (DIMS-MS/MS) a promising technique for the detection of cancer antigens that can be predicted by computational algorithms. In experiments using DIMS-MS/MS that were performed without the use of high-performance liquid chromatography (HPLC), a predicted model antigen, GLR (FLSSANEHL), was detected at a concentration of 10 pM (20 amol) in a mixture containing 94 competing model peptide antigens, each at a concentration of 1 μM. Without DIMS filtering, the GLR peptide was undetectable in the mixture even at 100 nM. Again, without using HPLC, DIMS-MS/MS was used to detect 2 of 3 previously characterized antigens produced by the leukemia cell line U937.A2. Because of its sensitivity, a targeted DIMS-MS/MS methodology can likely be used to probe for predicted cancer antigens from cancer cell lines as well as human tumor samples.
Collapse
Affiliation(s)
- Udara Dharmasiri
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill , 450 West Drive, 21-244, Chapel Hill, North Carolina 27599, United States
| | | | | | | |
Collapse
|
52
|
Hosen N, Maeda T, Hashii Y, Tsuboi A, Nishida S, Nakata J, Nakae Y, Takashima S, Oji Y, Oka Y, Kumanogoh A, Sugiyama H. Vaccination strategies to improve outcome of hematopoietic stem cell transplant in leukemia patients: early evidence and future prospects. Expert Rev Hematol 2014; 7:671-81. [DOI: 10.1586/17474086.2014.953925] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
53
|
Garber HR, Mirza A, Mittendorf EA, Alatrash G. Adoptive T-cell therapy for Leukemia. MOLECULAR AND CELLULAR THERAPIES 2014; 2:25. [PMID: 26056592 PMCID: PMC4452065 DOI: 10.1186/2052-8426-2-25] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/02/2014] [Indexed: 01/15/2023]
Abstract
Allogeneic stem cell transplantation (alloSCT) is the most robust form of adoptive cellular therapy (ACT) and has been tremendously effective in the treatment of leukemia. It is one of the original forms of cancer immunotherapy and illustrates that lymphocytes can specifically recognize and eliminate aberrant, malignant cells. However, because of the high morbidity and mortality that is associated with alloSCT including graft-versus-host disease (GvHD), refining the anti-leukemia immunity of alloSCT to target distinct antigens that mediate the graft-versus-leukemia (GvL) effect could transform our approach to treating leukemia, and possibly other hematologic malignancies. Over the past few decades, many leukemia antigens have been discovered that can separate malignant cells from normal host cells and render them vulnerable targets. In concert, the field of T-cell engineering has matured to enable transfer of ectopic high-affinity antigen receptors into host or donor cells with greater efficiency and potency. Many preclinical studies have demonstrated that engineered and conventional T-cells can mediate lysis and eradication of leukemia via one or more leukemia antigen targets. This evidence now serves as a foundation for clinical trials that aim to cure leukemia using T-cells. The recent clinical success of anti-CD19 chimeric antigen receptor (CAR) cells for treating patients with acute lymphoblastic leukemia and chronic lymphocytic leukemia displays the potential of this new therapeutic modality. In this review, we discuss some of the most promising leukemia antigens and the novel strategies that have been implemented for adoptive cellular immunotherapy of lymphoid and myeloid leukemias. It is important to summarize the data for ACT of leukemia for physicians in-training and in practice and for investigators who work in this and related fields as there are recent discoveries already being translated to the patient setting and numerous accruing clinical trials. We primarily focus on ACT that has been used in the clinical setting or that is currently undergoing preclinical testing with a foreseeable clinical endpoint.
Collapse
Affiliation(s)
- Haven R Garber
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center Houston, Houston, 77030 Texas
| | - Asma Mirza
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center Houston, Houston, 77030 Texas
| | - Elizabeth A Mittendorf
- Department Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Gheath Alatrash
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center Houston, Houston, 77030 Texas
| |
Collapse
|
54
|
Cobbold M, De La Peña H, Norris A, Polefrone JM, Qian J, English AM, Cummings KL, Penny S, Turner JE, Cottine J, Abelin JG, Malaker SA, Zarling AL, Huang HW, Goodyear O, Freeman SD, Shabanowitz J, Pratt G, Craddock C, Williams ME, Hunt DF, Engelhard VH. MHC class I-associated phosphopeptides are the targets of memory-like immunity in leukemia. Sci Transl Med 2014; 5:203ra125. [PMID: 24048523 DOI: 10.1126/scitranslmed.3006061] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Deregulation of signaling pathways is a hallmark of malignant transformation. Signaling-associated phosphoproteins can be degraded to generate cancer-specific phosphopeptides that are presented by major histocompatibility complex (MHC) class I and II molecules and recognized by T cells; however, the contribution of these phosphoprotein-specific T cells to immune surveillance is unclear. We identified 95 phosphopeptides presented on the surface of primary hematological tumors and normal tissues, including 61 that were tumor-specific. Phosphopeptides were more prevalent on more aggressive and malignant samples. CD8(+) T cell lines specific for these phosphopeptides recognized and killed both leukemia cell lines and human leukocyte antigen-matched primary leukemia cells ex vivo. Notably, healthy individuals showed robust CD8(+) T cell responses against many of these phosphopeptides within the circulating memory compartment. This immunity was significantly reduced or absent in some leukemia patients. This reduction correlated with clinical outcome; however, immunity was restored after allogeneic stem cell transplantation. These results suggest that phosphopeptides may be targets of cancer immune surveillance in humans, and point to their importance for development of vaccine-based and T cell adoptive transfer immunotherapies.
Collapse
Affiliation(s)
- Mark Cobbold
- Carter Immunology Center and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Impact of genomic polymorphisms on the repertoire of human MHC class I-associated peptides. Nat Commun 2014; 5:3600. [PMID: 24714562 PMCID: PMC3996541 DOI: 10.1038/ncomms4600] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 03/10/2014] [Indexed: 12/23/2022] Open
Abstract
For decades, the global impact of genomic polymorphisms on the repertoire of peptides presented by major histocompatibility complex (MHC) has remained a matter of speculation. Here we present a novel approach that enables high-throughput discovery of polymorphic MHC class I-associated peptides (MIPs), which play a major role in allorecognition. On the basis of comprehensive analyses of the genomic landscape of MIPs eluted from B lymphoblasts of two MHC-identical siblings, we show that 0.5% of non-synonymous single nucleotide variations are represented in the MIP repertoire. The 34 polymorphic MIPs found in our subjects are encoded by bi-allelic loci with dominant and recessive alleles. Our analyses show that, at the population level, 12% of the MIP-coding exome is polymorphic. Our method provides fundamental insights into the relationship between the genomic self and the immune self and accelerates the discovery of polymorphic MIPs (also known as minor histocompatibility antigens). Mass spectrometry (MS) has furthered our understanding of MHC class I-associated peptides (MIPs), but the technique is inadequate for studying MIP-associated polymorphisms. Here, the authors combine high-throughput MS with exome and transcriptome sequencing to identify polymorphic MIPs from two female siblings.
Collapse
|
56
|
Construction and molecular characterization of a T-cell receptor-like antibody and CAR-T cells specific for minor histocompatibility antigen HA-1H. Gene Ther 2014; 21:575-84. [PMID: 24694533 DOI: 10.1038/gt.2014.30] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 12/03/2013] [Accepted: 01/06/2014] [Indexed: 01/19/2023]
Abstract
The genetic transfer of T-cell receptors (TCRs) directed toward target antigens into T lymphocytes has been used to generate antitumor T cells efficiently without the need for the in vitro induction and expansion of T cells with cognate specificity. Alternatively, T cells have been gene-modified with a TCR-like antibody or chimeric antigen receptor (CAR). We show that immunization of HLA-A2 transgenic mice with tetramerized recombinant HLA-A2 incorporating HA-1 H minor histocompatibility antigen (mHag) peptides and β2-microglobulin (HA-1 H/HLA-A2) generate highly specific antibodies. One single-chain variable region moiety (scFv) antibody, #131, demonstrated high affinity (KD=14.9 nM) for the HA-1 H/HLA-A2 complex. Primary human T cells transduced with #131 scFV coupled to CD28 transmembrane and CD3ζ domains were stained with HA-1 H/HLA-A2 tetramers slightly more intensely than a cytotoxic T lymphocyte (CTL) clone specific for endogenously HLA-A2- and HA-1 H-positive cells. Although #131 scFv CAR-T cells required >100-fold higher antigen density to exert cytotoxicity compared with the cognate CTL clone, they could produce inflammatory cytokines against cells expressing HLA-A2 and HA-1 H transgenes. These data implicate that T cells with high-affinity antigen receptors reduce the ability to lyse targets with low-density peptide/MHC complexes (~100 per cell), while they could respond at cytokine production level.
Collapse
|
57
|
Fritsch EF, Rajasagi M, Ott PA, Brusic V, Hacohen N, Wu CJ. HLA-binding properties of tumor neoepitopes in humans. Cancer Immunol Res 2014; 2:522-9. [PMID: 24894089 DOI: 10.1158/2326-6066.cir-13-0227] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer genome sequencing has enabled the rapid identification of the complete repertoire of coding sequence mutations within a patient's tumor and facilitated their use as personalized immunogens. Although a variety of techniques are available to assist in the selection of mutation-defined epitopes to be included within the tumor vaccine, the ability of the peptide to bind to patient MHC is a key gateway to peptide presentation. With advances in the accuracy of predictive algorithms for MHC class I binding, choosing epitopes on the basis of predicted affinity provides a rapid and unbiased approach to epitope prioritization. We show herein the retrospective application of a prediction algorithm to a large set of bona fide T cell-defined mutated human tumor antigens that induced immune responses, most of which were associated with tumor regression or long-term disease stability. The results support the application of this approach for epitope selection and reveal informative features of these naturally occurring epitopes to aid in epitope prioritization for use in tumor vaccines.
Collapse
Affiliation(s)
- Edward F Fritsch
- Authors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, MassachusettsAuthors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, MassachusettsAuthors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Mohini Rajasagi
- Authors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, MassachusettsAuthors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Patrick A Ott
- Authors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, MassachusettsAuthors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Vladimir Brusic
- Authors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, MassachusettsAuthors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Nir Hacohen
- Authors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, MassachusettsAuthors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Catherine J Wu
- Authors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, MassachusettsAuthors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, MassachusettsAuthors' Affiliations: Cancer Vaccine Center; Department of Medical Oncology, Dana-Farber Cancer Institute; The Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Massachusetts General Hospital; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; and Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
58
|
Morris GP, Uy GL, Donermeyer D, Dipersio JF, Allen PM. Dual receptor T cells mediate pathologic alloreactivity in patients with acute graft-versus-host disease. Sci Transl Med 2014; 5:188ra74. [PMID: 23740900 DOI: 10.1126/scitranslmed.3005452] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Acute graft-versus-host disease (aGVHD) results from a robust response of donor T cells transferred during hematopoietic stem cell transplantation (HSCT) to allogeneic peptide-major histocompatibility complex antigens. Previous investigations have not identified T cell subsets that selectively mediate either protective immunity or pathogenic alloreactivity. We demonstrate that the small subset of peripheral T cells that naturally express two T cell receptors (TCRs) on the cell surface contributes disproportionately to aGVHD in patients after allogeneic HSCT. Dual TCR T cells from patients with aGVHD demonstrate an activated phenotype and produce pathogenic cytokines ex vivo. Dual receptor clones from a patient with symptomatic aGVHD responded specifically to mismatched recipient human leukocyte antigens (HLAs), demonstrating pathologic alloreactivity. Human dual TCR T cells are strongly activated and expanded by allogeneic stimulation in vitro, and disproportionately contribute to the repertoire of T cells recognizing both major (HLA) and minor histocompatibility antigens, providing a mechanism for their observed activity in vivo in patients with aGVHD. These results identify dual TCR T cells as a target for focused analysis of a T cell subset mediating GVHD and as a potential prognostic indicator.
Collapse
Affiliation(s)
- Gerald P Morris
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
59
|
Abstract
Minor histocompatibility (H) antigen mismatching leads to clinically relevant alloimmune reactivity. Depending on the tissue expression pattern of the involved minor H antigens, the immune response may either cause graft-versus-host disease and a graft-versus-tumor effect or lead to only a graft-versus-leukemia effect. Thus, identification of recipient-donor pairs with minor H antigen mismatches has clinical importance. This chapter describes molecular typing methods for molecular typing of minor H antigens.
Collapse
Affiliation(s)
- Eric Spierings
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
60
|
van Loenen MM, de Boer R, van Liempt E, Meij P, Jedema I, Falkenburg JHF, Heemskerk MHM. A Good Manufacturing Practice procedure to engineer donor virus-specific T cells into potent anti-leukemic effector cells. Haematologica 2013; 99:759-68. [PMID: 24334296 DOI: 10.3324/haematol.2013.093690] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A sequential, two-step procedure in which T-cell-depleted allogeneic stem cell transplantation is followed by treatment with donor lymphocyte infusion at 6 months can significantly reduce the risk and severity of graft-versus-host disease, with postponed induction of the beneficial graft-versus-leukemia effect. However, patients with high-risk leukemia have a substantial risk of relapse early after transplantation, at a time when administration of donor lymphocytes has a high likelihood of resulting in graft-versus-host disease, disturbing a favorable balance between the graft-versus-leukemia effect and graft-versus-host disease. New therapeutic modalities are, therefore, required to allow early administration of T cells capable of exerting a graft-versus-leukemia effect without causing graft-versus-host disease. Here we describe the isolation of virus-specific T cells using Streptamer-based isolation technology and subsequent transfer of the minor histocompatibility antigen HA-1-specific T-cell receptor using retroviral vectors. Isolation of virus-specific T cells and subsequent transduction with HA-1-T-cell receptor resulted in rapid in vitro generation of highly pure, dual-specific T cells with potent anti-leukemic reactivity. Due to the short production procedure of only 10-14 days and the defined specificity of the T cells, administration of virus-specific T cells transduced with the HA-1-T-cell receptor as early as 8 weeks after allogeneic stem cell transplantation is feasible. (This clinical trial is registered at www.clinicaltrialsregister.eu as EudraCT number 2010-024625-20).
Collapse
|
61
|
Roeven MWH, Hobo W, Schaap N, Dolstra H. Immunotherapeutic approaches to treat multiple myeloma. Hum Vaccin Immunother 2013; 10:896-910. [PMID: 24335570 PMCID: PMC4896532 DOI: 10.4161/hv.27380] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 11/21/2013] [Accepted: 11/29/2013] [Indexed: 11/19/2022] Open
Abstract
Cellular immunotherapy can be an effective adjuvant treatment for multiple myeloma (MM), as demonstrated by induction of durable remissions after allogeneic stem cell transplantation. However, anti-myeloma immunity is often hampered by suppressive mechanisms in the tumor micro-environment resulting in relapse or disease progression. To overcome this immunosuppression, new cellular immunotherapies have been developed, based on the important effector cells in anti-myeloma immunity, namely T cells and natural killer cells. These effectors can be modulated to improve their functionality, activated by dendritic cell vaccines, or combined with immune stimulating antibodies or immunomodulatory drugs to enhance their efficacy. In this review, we discuss promising pre-clinical and clinical data in the field of cellular immunotherapy in MM. In addition, we address the potential of combining these strategies with other therapies to maximize clinical effects without increasing toxicity. The reviewed therapies might pave the way to effective personalized treatments for MM patients.
Collapse
Affiliation(s)
- Mieke WH Roeven
- Department of Hematology; Radboud University Medical Center; Nijmegen, The Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine—Laboratory of Hematology Radboud; University Medical Center; Nijmegen, The Netherlands
| | - Nicolaas Schaap
- Department of Hematology; Radboud University Medical Center; Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine—Laboratory of Hematology Radboud; University Medical Center; Nijmegen, The Netherlands
| |
Collapse
|
62
|
Dierselhuis MP, Spierings E, Drabbels J, Hendriks M, Alaez C, Alberú J, Alvarez MB, Burlingham W, Campos E, Christiaans M, Claas F, Fasano ME, Gerbase-DeLima M, Gervais T, Gorodezky C, Larriba J, Lardy NM, Latinne D, Morales-Buenrostro LE, Moreno MJ, Oguz F, Opelz G, Sergeant R, Tambutti M, Teper S, Tilanus M, Turkmen A, Warrens AN, Weimar W, Goulmy E. Minor H antigen matches and mismatches are equally distributed among recipients with or without complications after HLA identical sibling renal transplantation. ACTA ACUST UNITED AC 2013; 82:312-6. [DOI: 10.1111/tan.12209] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/31/2013] [Accepted: 09/06/2013] [Indexed: 11/28/2022]
Affiliation(s)
- M. P. Dierselhuis
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden; The Netherlands
| | - E. Spierings
- Department of Immunology; University Medical Center Utrecht; Utrecht; The Netherlands
| | - J. Drabbels
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden; The Netherlands
| | - M. Hendriks
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden; The Netherlands
| | - C. Alaez
- Department of Immunology & Immunogenetics; Instituto de Diagnostico y Referencia Epidemiologicos; Mexico City; Mexico
| | - J. Alberú
- Department of Transplantation; Instituto Nacional de Ciencias, Médicas y de la Nutrición; Mexico City; Mexico
| | - M. B. Alvarez
- Centro de Inmunología y Genética molecular; Buenos Aires; Argentina
| | - W. Burlingham
- Department of Surgery; University of Wisconsin; Madison; WI; USA
| | - E. Campos
- Universidade Federal de São Paulo and Associação Fundo de Incentivo à Pesquisa; São Paulo; Brazil
| | - M. Christiaans
- Department of Nephrology; Maastricht University Medical Center; Maastricht; The Netherlands
| | - F. Claas
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden; The Netherlands
| | - M. E. Fasano
- Immunologia Trapianti c/o Genetica; Turin; Italy
| | - M. Gerbase-DeLima
- Universidade Federal de São Paulo and Associação Fundo de Incentivo à Pesquisa; São Paulo; Brazil
| | - T. Gervais
- Immunohaematology, Cliniques St. Luc; Université Catholique de Louvain; Brussels; Belgium
| | - C. Gorodezky
- Department of Immunology & Immunogenetics; Instituto de Diagnostico y Referencia Epidemiologicos; Mexico City; Mexico
| | - J. Larriba
- Histocompatibility and Immunogenetics-ICBME; Hospital Italiano de Buenos Aires; Buenos Aires; Argentina
| | - N. M. Lardy
- Sanquin-Diagnostic Services; Amsterdam; The Netherlands
| | - D. Latinne
- Immunohaematology, Cliniques St. Luc; Université Catholique de Louvain; Brussels; Belgium
| | - L.-E. Morales-Buenrostro
- Department of Nephrology and Mineral Metabolism; Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran; México City; Mexico
| | - M. J. Moreno
- Histocompatibility Laboratory; CEMIC-Centro de Educacion Medica e Investigaciones Clinicas Norberto Quirno; Buenos Aires; Argentina
| | - F. Oguz
- Department of Medical Biology, Medical Faculty of Istanbul; Istanbul University; Istanbul; Turkey
| | - G. Opelz
- Institute of Immunology; University of Heidelberg; Heidelberg; Germany
| | - R. Sergeant
- Immunology; Imperial College Healthcare NHS Trust; London; UK
| | - M. Tambutti
- Histocompatibility and Immunogenetics-ICBME; Hospital Italiano de Buenos Aires; Buenos Aires; Argentina
| | - S. Teper
- Histocompatibility Laboratory; CEMIC-Centro de Educacion Medica e Investigaciones Clinicas Norberto Quirno; Buenos Aires; Argentina
| | - M. Tilanus
- Transplantation Immunology, Tissue Typing Laboratory; Maastricht University Medical Center; Maastricht; The Netherlands
| | - A. Turkmen
- Department of Medical Biology, Medical Faculty of Istanbul; Istanbul University; Istanbul; Turkey
| | - A. N. Warrens
- Immunology; Imperial College Healthcare NHS Trust; London; UK
| | - W. Weimar
- Internal medicine; Erasmus Medical Center; Rotterdam; The Netherlands
| | - E. Goulmy
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden; The Netherlands
| |
Collapse
|
63
|
de Haart SJ, van de Donk NW, Minnema MC, Huang JH, Aarts-Riemens T, Bovenschen N, Yuan H, Groen RW, McMillin DW, Jakubikova J, Lokhorst HM, Martens AC, Mitsiades CS, Mutis T. Accessory Cells of the Microenvironment Protect Multiple Myeloma from T-Cell Cytotoxicity through Cell Adhesion-Mediated Immune Resistance. Clin Cancer Res 2013; 19:5591-601. [DOI: 10.1158/1078-0432.ccr-12-3676] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
64
|
Warren EH, Deeg HJ. Dissecting graft-versus-leukemia from graft-versus-host-disease using novel strategies. ACTA ACUST UNITED AC 2013; 81:183-93. [PMID: 23510414 DOI: 10.1111/tan.12090] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The intrinsic anti-leukemic effect of allogeneic hematopoietic cell transplantation (HCT) is dependent on genetic disparity between donor and recipient, intimately associated with graft-versus-host disease (GVHD), and mediated by lymphocytes contained in or derived from the donor hematopoietic cell graft. Three decades of intense effort have not identified clinical strategies that can reliably separate the graft-versus-leukemia (GVL) effect from the alloimmune reaction that drives clinical GVHD. For patients who require HCT and for whom two or more human leukocyte antigen (HLA)-A, -B, -C, and -DRB1-matched donor candidates can be identified, consideration of donor and recipient genotype at additional genetic loci both within and outside the major histocompatibility complex may offer the possibility of selecting the donor [candidate(s)] that poses the lowest probability of GVHD and the highest probability of a potent GVL effect. Strategies for engineering conventional donor lymphocyte infusion also hold promise for prevention or improved treatment of post-transplant relapse. The brightest prospects for selectively enhancing the anti-leukemic efficacy of allogeneic HCT, however, are likely to be interventions that are designed to enhance specific antitumor immunity via vaccination or adoptive cell transfer, rather than those that attempt to exploit donor alloreactivity against the host. Adoptive transfer of donor-derived T cells genetically modified for tumor-specific reactivity, in particular, has the potential to transform the practice of allogeneic HCT by selectively enhancing antitumor immunity without causing GVHD.
Collapse
Affiliation(s)
- E H Warren
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA.
| | | |
Collapse
|
65
|
Linscheid C, Petroff MG. Minor histocompatibility antigens and the maternal immune response to the fetus during pregnancy. Am J Reprod Immunol 2013; 69:304-14. [PMID: 23398025 PMCID: PMC4048750 DOI: 10.1111/aji.12075] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 12/20/2012] [Indexed: 12/21/2022] Open
Abstract
The tolerance of the semiallogeneic fetus by the maternal immune system is an important area of research for understanding how the maternal and fetal systems interact during pregnancy to ensure a successful outcome. Several lines of research reveal that the maternal immune system can recognize and respond to fetal minor histocompatibility antigens during pregnancy. Reactions to these antigens arise because of allelic differences between the mother and fetus and have been shown more broadly to play an important role in mediating transplantation outcomes. This review outlines the discovery of minor histocompatibility antigens and their importance in solid organ and hematopoietic stem cell transplantations, maternal T-cell responses to minor histocompatibility antigens during pregnancy, expression of minor histocompatibility antigens in the human placenta, and the potential involvement of minor histocompatibility antigens in the development and manifestation of pregnancy complications.
Collapse
Affiliation(s)
- Caitlin Linscheid
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS
| | - Margaret G. Petroff
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
66
|
Hassan C, Kester MGD, de Ru AH, Hombrink P, Drijfhout JW, Nijveen H, Leunissen JAM, Heemskerk MHM, Falkenburg JHF, van Veelen PA. The human leukocyte antigen-presented ligandome of B lymphocytes. Mol Cell Proteomics 2013; 12:1829-43. [PMID: 23481700 DOI: 10.1074/mcp.m112.024810] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Peptides presented by human leukocyte antigen (HLA) molecules on the cell surface play a crucial role in adaptive immunology, mediating the communication between T cells and antigen presenting cells. Knowledge of these peptides is of pivotal importance in fundamental studies of T cell action and in cellular immunotherapy and transplantation. In this paper we present the in-depth identification and relative quantification of 14,500 peptide ligands constituting the HLA ligandome of B cells. This large number of identified ligands provides general insight into the presented peptide repertoire and antigen presentation. Our uniquely large set of HLA ligands allowed us to characterize in detail the peptides constituting the ligandome in terms of relative abundance, peptide length distribution, physicochemical properties, binding affinity to the HLA molecule, and presence of post-translational modifications. The presented B-lymphocyte ligandome is shown to be a rich source of information by the presence of minor histocompatibility antigens, virus-derived epitopes, and post-translationally modified HLA ligands, and it can be a good starting point for solving a wealth of specific immunological questions. These HLA ligands can form the basis for reversed immunology approaches to identify T cell epitopes based not on in silico predictions but on the bona fide eluted HLA ligandome.
Collapse
Affiliation(s)
- Chopie Hassan
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Schmitt M, Freund M. Blood. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
68
|
Occurrence and Impact of Minor Histocompatibility Antigens' Disparities on Outcomes of Hematopoietic Stem Cell Transplantation from HLA-Matched Sibling Donors. BONE MARROW RESEARCH 2012. [PMID: 23193478 PMCID: PMC3502767 DOI: 10.1155/2012/257086] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We have examined the alleles of eleven minor histocompatibility antigens (MiHAs) and investigated the occurrence of immunogenic MiHA disparities in 62 recipients of allogeneic hematopoietic cell transplantation (allo-HCT) with myeloablative conditioning performed between 2000 and 2008 and in their HLA-matched sibling donors. Immunogenic MiHA mismatches were detected in 42 donor-recipient pairs: in 29% MiHA was mismatched in HVG direction, in another 29% in GVH direction; bidirectional MiHA disparity was detected in 10% and no MiHA mismatches in 32%. Patients with GVH-directed HY mismatches had lower both overall survival and disease-free survival at 3 years than patients with compatible HY; also higher incidence of both severe acute GvHD and extensive chronic GVHD was observed in patients with GVH-directed HY mismatch. On contrary, GVH-directed mismatches of autosomally encoded MiHAs had no negative effect on overall survival. Results of our study help to understand why posttransplant courses of allo-HCT from siblings may vary despite the complete high-resolution HLA matching of a donor and a recipient.
Collapse
|
69
|
Kotsiou E, Davies JK. New ways to separate graft-versus-host disease and graft-versus-tumour effects after allogeneic haematopoietic stem cell transplantation. Br J Haematol 2012; 160:133-45. [PMID: 23121307 DOI: 10.1111/bjh.12115] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 09/13/2012] [Indexed: 12/25/2022]
Abstract
A major challenge to transplant immunologists and physicians remains the separation of harmful graft-versus-host disease (GvHD) and beneficial graft-versus-tumour (GvT) effects after allogeneic haematopoietic stem cell transplantation. Recent advances in our understanding of the allogeneic immune response provide potential new opportunities to achieve this goal. Three potential new approaches that capitalize on this new knowledge are considered in depth; the manipulation of organ-specific cytokines and other pro-inflammatory signals, the selective manipulation of donor effector T cell migration, and the development of cell-mediated immunosuppressive strategies using donor-derived regulatory T cells. These new approaches could provide strategies for local control of allogeneic immune responses, a new paradigm to separate GvHD and GvT effects. Although these strategies are currently in their infancy and have challenges to successful translation to clinical practice, all have exciting potential for the future.
Collapse
Affiliation(s)
- Eleni Kotsiou
- Centre for Haemato-Oncology, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, London, UK
| | | |
Collapse
|
70
|
Spierings E, Goulmy E. Minor histocompatibility antigen typing by DNA sequencing for clinical practice in hematopoietic stem-cell transplantation. Methods Mol Biol 2012; 882:509-30. [PMID: 22665253 DOI: 10.1007/978-1-61779-842-9_29] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
In HLA-matched stem-cell transplantation (SCT), minor H antigens are key molecules driving allo-immune responses in both graft-versus-host disease (GvHD) and in graft-versus-leukemia (GvL) reactivity. Dissection of the dual function of minor H antigens became evident through their different modes of tissue and cell expression, i.e., hematopoietic system restricted or broad. Broadly expressed minor H antigens are the targets of immune responses in both arms of graft-versus-host (GvH) responses, i.e., both GvHD and GvL, whereas the immune responses against the hematopoietic system-specific minor H antigens are restricted to the GvL arm of SCT. Evidently, it is this latter group of minor H antigens that can function as curative tools for stem-cell (SC)-based immunotherapy of hematological malignancies and disorders. The HLA-matched patient/donor combinations, incompatible for one of the hematopoietic-specific minor H antigens, are suitable for minor H antigen immunotherapy (Goulmy, Immunol Rev 157:125-140, 1997). Information on the minor H antigen phenotype is therefore needed. Hereto, genomic typing for minor H antigens has been implemented in many HLA laboratories. Here, we firstly summarize the relevance of minor H antigens particularly in hematopoietic SCT. Secondly, we describe a method for typing the various polymorphic minor H antigens molecularly identified to date by DNA sequencing.
Collapse
Affiliation(s)
- Eric Spierings
- Department of Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
71
|
Mortensen BK, Rasmussen AH, Larsen ME, Larsen MV, Lund O, Braendstrup P, Harndahl M, Rasmussen M, Buus S, Stryhn A, Vindeløv L. Identification of a novel UTY-encoded minor histocompatibility antigen. Scand J Immunol 2012; 76:141-50. [PMID: 22536994 DOI: 10.1111/j.1365-3083.2012.02708.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Minor histocompatibility antigens (mHags) encoded by the Y-chromosome (H-Y-mHags) are known to play a pivotal role in allogeneic haematopoietic cell transplantation (HCT) involving female donors and male recipients. We present a new H-Y-mHag, YYNAFHWAI (UTY(139-147)), encoded by the UTY gene and presented by HLA-A*24:02. Briefly, short peptide stretches encompassing multiple putative H-Y-mHags were designed using a bioinformatics predictor of peptide-HLA binding, NetMHCpan. These peptides were used to screen for peptide-specific HLA-restricted T cell responses in peripheral blood mononuclear cells obtained post-HCT from male recipients of female donor grafts. In one of these recipients, a CD8+ T cell response was observed against a peptide stretch encoded by the UTY gene. Another bioinformatics tool, HLArestrictor, was used to identify the optimal peptide and HLA-restriction element. Using peptide/HLA tetramers, the specificity of the CD8+ T cell response was successfully validated as being HLA-A*24:02-restricted and directed against the male UTY(139-147) peptide. Functional analysis of these T cells demonstrated male UTY(139-147) peptide-specific cytokine secretion (IFNγ, TNFα and MIP-1β) and cytotoxic degranulation (CD107a). In contrast, no responses were seen when the T cells were stimulated with patient tumour cells alone. CD8+ T cells specific for this new H-Y-mHag were found in three of five HLA-A*24:02-positive male recipients of female donor HCT grafts available for this study.
Collapse
Affiliation(s)
- B K Mortensen
- Allogeneic Hematopoietic Cell Transplantation Laboratory, Department of Hematology, Rigshospitalet, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Towards effective and safe immunotherapy after allogeneic stem cell transplantation: identification of hematopoietic-specific minor histocompatibility antigen UTA2-1. Leukemia 2012; 27:642-9. [PMID: 23079962 PMCID: PMC3593180 DOI: 10.1038/leu.2012.277] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Donor T cells directed at hematopoietic system-specific minor histocompatibility antigens (mHags) are considered important cellular tools to induce therapeutic graft-versus-tumor (GvT) effects with low risk of graft-versus-host disease after allogeneic stem cell transplantation. To enable the clinical evaluation of the concept of mHag-based immunotherapy and subsequent broad implementation, the identification of more hematopoietic mHags with broad applicability is imperative. Here we describe novel mHag UTA2-1 with ideal characteristics for this purpose. We identified this antigen using genome-wide zygosity-genotype correlation analysis of a mHag-specific CD8+ cytotoxic T lymphocyte (CTL) clone derived from a multiple myeloma patient who achieved a long-lasting complete remission after donor lymphocyte infusion from an human leukocyte antigen (HLA)-matched sibling. UTA2-1 is a polymorphic peptide presented by the common HLA molecule HLA-A*02:01, which is encoded by the bi-allelic hematopoietic-specific gene C12orf35. Tetramer analyses demonstrated an expansion of UTA2-1-directed T cells in patient blood samples after several donor T-cell infusions that mediated clinical GvT responses. More importantly, UTA2-1-specific CTL effectively lysed mHag+ hematopoietic cells, including patient myeloma cells, without affecting non-hematopoietic cells. Thus, with the capacity to induce relevant immunotherapeutic CTLs, it's HLA-A*02 restriction and equally balanced phenotype frequency, UTA2-1 is a highly valuable mHag to facilitate clinical application of mHag-based immunotherapy.
Collapse
|
73
|
Hobo W, Broen K, van der Velden WJFM, Greupink-Draaisma A, Adisty N, Wouters Y, Kester M, Fredrix H, Jansen JH, van der Reijden B, Falkenburg JHF, de Witte T, Preijers F, Schattenberg T, Feuth T, Blijlevens NM, Schaap N, Dolstra H. Association of disparities in known minor histocompatibility antigens with relapse-free survival and graft-versus-host disease after allogeneic stem cell transplantation. Biol Blood Marrow Transplant 2012; 19:274-82. [PMID: 23022467 DOI: 10.1016/j.bbmt.2012.09.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 09/12/2012] [Indexed: 11/15/2022]
Abstract
Allogeneic stem cell transplantation (allo-SCT) can induce remission in patients with hematologic malignancies due to graft-versus-tumor (GVT) responses. This immune-mediated antitumor effect is often accompanied by detrimental graft-versus-host disease (GVHD), however. Both GVT and GVHD are mediated by minor histocompatibility antigen (MiHA)-specific T cells recognizing peptide products from polymorphic genes that differ between recipient and donor. In this study, we evaluated whether mismatches in a panel of 17 MiHAs are associated with clinical outcome after partially T cell-depleted allo-SCT. Comprehensive statistical analysis revealed that DNA mismatches for one or more autosomal-encoded MiHAs was associated with increased relapse-free survival in recipients of sibling transplants (P = .04), particularly in those with multiple myeloma (P = .02). Moreover, mismatches for the ubiquitous Y chromosome-derived MiHAs resulted in a higher incidence of acute GVHD grade III-IV (P = .004), whereas autosomal MiHA mismatches, ubiquitous or restricted to hematopoietic cells, were not associated with severe GVHD. Finally, we found considerable differences among MiHAs in their capability of inducing in vivo T cell responses using dual-color tetramer analysis of peripheral blood samples collected after allo-SCT. Importantly, detection of MiHA-specific T cell responses was associated with improved relapse-free survival in recipients of sibling transplants (P = .01). Our findings provide a rationale for further boosting GVT immunity toward autosomal MiHAs with a hematopoietic restriction to improve outcomes after HLA-matched allo-SCT.
Collapse
Affiliation(s)
- Willemijn Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Effect of MHC and non-MHC donor/recipient genetic disparity on the outcome of allogeneic HCT. Blood 2012; 120:2796-806. [PMID: 22859606 DOI: 10.1182/blood-2012-04-347286] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The outcome of allogeneic hematopoietic cell transplantation is influenced by donor/recipient genetic disparity at loci both inside and outside the MHC on chromosome 6p. Although disparity at loci within the MHC is the most important risk factor for the development of severe GVHD, disparity at loci outside the MHC that encode minor histocompatibility (H) antigens can elicit GVHD and GVL activity in donor/recipient pairs who are otherwise genetically identical across the MHC. Minor H antigens are created by sequence and structural variations within the genome. The enormous variation that characterizes the human genome suggests that the total number of minor H loci is probably large and ensures that all donor/recipient pairs, despite selection for identity at the MHC, will be mismatched for many minor H antigens. In addition to mismatch at minor H loci, unrelated donor/recipient pairs exhibit genetic disparity at numerous loci within the MHC, particularly HLA-DP, despite selection for identity at HLA-A, -B, -C, and -DRB1. Disparity at HLA-DP exists in 80% of unrelated pairs and clearly influences the outcome of unrelated hematopoietic cell transplantation; the magnitude of this effect probably exceeds that associated with disparity at any locus outside the MHC.
Collapse
|
75
|
Bleakley M, Turtle CJ, Riddell SR. Augmentation of anti-tumor immunity by adoptive T-cell transfer after allogeneic hematopoietic stem cell transplantation. Expert Rev Hematol 2012; 5:409-25. [PMID: 22992235 PMCID: PMC3590108 DOI: 10.1586/ehm.12.28] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (HCT) is currently the standard of care for most patients with high-risk acute leukemias and some other hematologic malignancies. Although HCT can be curative, many patients who undergo allogeneic HCT will later relapse. There is, therefore, a critical need for the development of novel post-HCT therapies for patients who are at high risk for disease recurrence following HCT. One potentially efficacious approach is adoptive T-cell immunotherapy, which is currently undergoing a renaissance that has been inspired by scientific insight into the key issues that impeded its previous clinical application. Translation of the next generation of adoptive T-cell therapies to the allogeneic HCT setting, using donor T cells of defined specificity and function, presents a unique set of challenges and opportunities. The challenges, progress and future of adoptive T-cell therapy following allogeneic HCT are discussed in this review.
Collapse
Affiliation(s)
- Marie Bleakley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | | | | |
Collapse
|
76
|
Yamamura T, Hikita J, Bleakley M, Hirosawa T, Sato-Otsubo A, Torikai H, Hamajima T, Nannya Y, Demachi-Okamura A, Maruya E, Saji H, Yamamoto Y, Takahashi T, Emi N, Morishima Y, Kodera Y, Kuzushima K, Riddell SR, Ogawa S, Akatsuka Y. HapMap SNP Scanner: an online program to mine SNPs responsible for cell phenotype. ACTA ACUST UNITED AC 2012; 80:119-25. [PMID: 22568758 DOI: 10.1111/j.1399-0039.2012.01883.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Minor histocompatibility (H) antigens are targets of graft-vs-host disease and graft-vs-tumor responses after human leukocyte antigen matched allogeneic hematopoietic stem cell transplantation. Recently, we reported a strategy for genetic mapping of linkage disequilibrium blocks that encoded novel minor H antigens using the large dataset from the International HapMap Project combined with conventional immunologic assays to assess recognition of HapMap B-lymphoid cell line by minor H antigen-specific T cells. In this study, we have constructed and provide an online interactive program and demonstrate its utility for searching for single-nucleotide polymorphisms (SNPs) responsible for minor H antigen generation. The website is available as 'HapMap SNP Scanner', and can incorporate T-cell recognition and other data with genotyping datasets from CEU, JPT, CHB, and YRI to provide a list of candidate SNPs that correlate with observed phenotypes. This method should substantially facilitate discovery of novel SNPs responsible for minor H antigens and be applicable for assaying of other specific cell phenotypes (e.g. drug sensitivity) to identify individuals who may benefit from SNP-based customized therapies.
Collapse
Affiliation(s)
- T Yamamura
- Division of Immunology, Aichi Cancer Center Research Center, Nagoya, Aichi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Broen K, Greupink-Draaisma A, Fredrix H, Schaap N, Dolstra H. Induction of multiple myeloma-reactive T cells during post-transplantation immunotherapy with donor lymphocytes and recipient DCs. Bone Marrow Transplant 2012; 47:1229-34. [PMID: 22246088 DOI: 10.1038/bmt.2011.258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Recently, we demonstrated that reduced intensity conditioning (RIC) followed by partial T-cell-depleted SCT creates a platform for inducing the graft-versus-myeloma effect by adjuvant immunotherapy. Here, we evaluated mHA-specific T-cell responses in a multiple myeloma (MM) patient who was treated with RIC-SCT followed by donor lymphocyte infusion (DLI) and subsequent recipient DC vaccination. We isolated a mHA-specific CTL clone with the capacity to target MM tumor cells from this patient experiencing long-term CR. This CTL clone recognizes an HLA-A3-restricted mHA and mediates killing of both primary MM cells and the MM-cell line U266, while BM-derived fibroblasts are not recognized. CTL-specific T-cell receptor (TCR) transcripts could be detected by quantitative PCR analysis in both peripheral blood and BM during tumor remission. Interestingly, a strong increase of CTL-specific TCR transcripts at the BM tumor site was observed following DLI and recipient DC vaccination, while the TCR signal in peripheral blood decreased. These findings illustrate that the approach of partial T-cell-depleted RIC-SCT followed by post-transplantation immunotherapy induces mHA-specific T-cell responses targeting MM tumor cells.
Collapse
Affiliation(s)
- K Broen
- Department of Laboratory Medicine-Laboratory of Hematology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
78
|
Holland OJ, Linscheid C, Hodes HC, Nauser TL, Gilliam M, Stone P, Chamley LW, Petroff MG. Minor histocompatibility antigens are expressed in syncytiotrophoblast and trophoblast debris: implications for maternal alloreactivity to the fetus. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:256-66. [PMID: 22079431 PMCID: PMC3338347 DOI: 10.1016/j.ajpath.2011.09.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 08/09/2011] [Accepted: 09/20/2011] [Indexed: 01/29/2023]
Abstract
The fetal semi-allograft can induce expansion and tolerance of antigen-specific maternal T and B cells through paternally inherited major histocompatibility complex and minor histocompatibility antigens (mHAgs). The effects of these antigens have important consequences on the maternal immune system both during and long after pregnancy. Herein, we investigate the possibility that the placental syncytiotrophoblast and deported trophoblastic debris serve as sources of fetal mHAgs. We mapped the expression of four mHAgs (human mHAg 1, pumilio domain-containing protein KIAA0020, B-cell lymphoma 2-related protein A1, and ribosomal protein S4, Y linked) in the placenta. Each of these proteins was expressed in several placental cell types, including the syncytiotrophoblast. These antigens and two additional Y chromosome-encoded antigens [DEAD box polypeptide 3, Y linked (DDX3Y), and lysine demethylase5D] were also identified by RT-PCR in the placenta, purified trophoblast cells, and cord blood cells. Finally, we used a proteomic approach to investigate the presence of mHAgs in the syncytiotrophoblast and trophoblast debris shed from first-trimester placenta. By this method, four antigens (DDX3Y; ribosomal protein S4, Y linked; solute carrier 1A5; and signal sequence receptor 1) were found in the syncytiotrophoblast, and one antigen (DDX3Y) was found in shed trophoblast debris. The finding of mHAgs in the placenta and in trophoblast debris provides the first direct evidence that fetal antigens are present in debris shed from the human placenta. The data, thus, suggest a mechanism by which the maternal immune system is exposed to fetal alloantigens, possibly explaining the relationship between parity and graft-versus-host disease.
Collapse
Affiliation(s)
- Olivia J. Holland
- Department of Obstetrics and Gynecology, University of Auckland, Auckland, New Zealand
| | - Caitlin Linscheid
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | | | | | - Melissa Gilliam
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, Illinois
| | - Peter Stone
- Department of Obstetrics and Gynecology, University of Auckland, Auckland, New Zealand
| | - Larry W. Chamley
- Department of Obstetrics and Gynecology, University of Auckland, Auckland, New Zealand
| | - Margaret G. Petroff
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
79
|
Simpson AA, Mohammed F, Salim M, Tranter A, Rickinson AB, Stauss HJ, Moss PAH, Steven NM, Willcox BE. Structural and energetic evidence for highly peptide-specific tumor antigen targeting via allo-MHC restriction. Proc Natl Acad Sci U S A 2011; 108:21176-81. [PMID: 22160697 PMCID: PMC3248497 DOI: 10.1073/pnas.1108422109] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Immunotherapies targeting peptides presented by allogeneic MHC molecules offer the prospect of circumventing tolerance to key tumor-associated self-antigens. However, the degree of antigen specificity mediated by alloreactive T cells, and their ability to discriminate normal tissues from transformed cells presenting elevated antigen levels, is poorly understood. We examined allorecognition of an HLA-A2-restricted Hodgkin's lymphoma-associated antigen and were able to isolate functionally antigen-specific allo-HLA-A2-restricted T cells from multiple donors. Binding and structural studies, focused on a prototypic allo-HLA-A2-restricted T-cell receptor (TCR) termed NB20 derived from an HLA-A3 homozygote, suggested highly peptide-specific allorecognition that was energetically focused on antigen, involving direct recognition of a distinct allopeptide presented within a conserved MHC recognition surface. Although NB20/HLA-A2 affinity was unremarkable, TCR/MHC complexes were very short-lived, consistent with suboptimal TCR triggering and tolerance to low antigen levels. These data provide strong molecular evidence that within the functionally heterogeneous alloreactive repertoire, there is the potential for highly antigen-specific "allo-MHC-restricted" recognition and suggest a kinetic mechanism whereby allo-MHC-restricted T cells may discriminate normal from transformed tissue, thereby outlining a suitable basis for broad-based therapeutic targeting of tolerizing tumor antigens.
Collapse
Affiliation(s)
- Amy A. Simpson
- Birmingham Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Fiyaz Mohammed
- Birmingham Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Mahboob Salim
- Birmingham Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Amy Tranter
- Birmingham Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Alan B. Rickinson
- Birmingham Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Hans J. Stauss
- Division of Infection and Immunity, Department of Immunology, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom
| | - Paul A. H. Moss
- Birmingham Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Neil M. Steven
- Birmingham Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Benjamin E. Willcox
- Birmingham Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| |
Collapse
|
80
|
Pabón M, Navarro C, Martin R, Rodríguez M, Martin I, Gaitán L, Gómez A, Lozano E. Minor Histocompatibility Antigens as Risk Factor for Poor Prognosis in Kidney Transplantation. Transplant Proc 2011; 43:3319-23. [DOI: 10.1016/j.transproceed.2011.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
81
|
Armistead PM, Liang S, Li H, Lu S, Van Bergen CAM, Alatrash G, St John L, Hunsucker SA, Sarantopoulos S, Falkenburg JHF, Molldrem JJ. Common minor histocompatibility antigen discovery based upon patient clinical outcomes and genomic data. PLoS One 2011; 6:e23217. [PMID: 21858034 PMCID: PMC3153501 DOI: 10.1371/journal.pone.0023217] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 07/14/2011] [Indexed: 11/19/2022] Open
Abstract
Background Minor histocompatibility antigens (mHA) mediate much of the graft vs. leukemia (GvL) effect and graft vs. host disease (GvHD) in patients who undergo allogeneic stem cell transplantation (SCT) [1], [2], [3], [4]. Therapeutic decision making and treatments [5] based upon mHAs will require the evaluation of multiple candidate mHAs and the selection of those with the potential to have the greatest impact on clinical outcomes. We hypothesized that common, immunodominant mHAs, which are presented by HLA-A, B, and C molecules, can mediate clinically significant GvL and/or GvHD, and that these mHAs can be identified through association of genomic data with clinical outcomes. Methodology/Principal Findings Because most mHAs result from donor/recipient cSNP disparities, we genotyped 57 myeloid leukemia patients and their donors at 13,917 cSNPs [6]. We correlated the frequency of genetically predicted mHA disparities with clinical evidence of an immune response and then computationally screened all peptides mapping to the highly associated cSNPs for their ability to bind to HLA molecules. As proof-of-concept, we analyzed one predicted antigen, T4A, whose mHA mismatch trended towards improved overall and disease free survival in our cohort. T4A mHA mismatches occurred at the maximum theoretical frequency for any given SCT. T4A-specific CD8+ T lymphocytes (CTLs) were detected in 3 of 4 evaluable post-transplant patients predicted to have a T4A mismatch. Conclusions/Significance Our method is the first to combine clinical outcomes data with genomics and bioinformatics methods to predict and confirm a mHA. Refinement of this method should enable the discovery of clinically relevant mHAs in the majority of transplant patients and possibly lead to novel immunotherapeutics [5].
Collapse
Affiliation(s)
- Paul M Armistead
- Section of Transplantation Immunology, Department of Stem Cell Transplant and Cellular Therapy, M.D. Anderson Cancer Center, Houston, Texas, United States of America.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Kotsiou E, Brzostek J, Lenart I, Antoniou AN, Dyson J, Gould KG. Dimerization of soluble disulfide trap single-chain major histocompatibility complex class I molecules dependent on peptide binding affinity. Antioxid Redox Signal 2011; 15:635-44. [PMID: 21050141 DOI: 10.1089/ars.2010.3691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Stable presentation of peptide epitope by major histocompatibility complex (MHC) class I molecules is a prerequisite for the efficient expansion of CD8(+) T cells. The construction of single-chain MHC class I molecules in which the peptide, β(2)-microglobulin, and MHC heavy chain are all joined together via flexible linkers increases peptide-MHC stability. We have expressed two T cell epitopes that may be useful in leukemia treatment as single-chain MHC class I molecules, aiming to develop a system for the expansion of antigen-specific CD8(+) T cells in vitro. Disulfide trap versions of these single-chain MHC molecules were also created to improve anchoring of the peptides in the MHC molecule. Unexpectedly, we observed that soluble disulfide trap single-chain molecules expressed in eukaryotic cells were prone to homodimerization, depending on the binding affinity of the peptide epitope. The dimers were remarkably stable and efficiently recognized by conformation-specific antibodies, suggesting that they consisted of largely correctly folded molecules. However, dimerization was not observed when the disulfide trap molecules were expressed as full-length, transmembrane-anchored molecules. Our results further emphasize the importance of peptide binding affinity for the efficient folding of MHC class I molecules.
Collapse
Affiliation(s)
- Eleni Kotsiou
- Department of Immunology, Wright-Fleming Institute, Imperial College London, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
83
|
Amir AL, van der Steen DM, van Loenen MM, Hagedoorn RS, de Boer R, Kester MDG, de Ru AH, Lugthart GJ, van Kooten C, Hiemstra PS, Jedema I, Griffioen M, van Veelen PA, Falkenburg JHF, Heemskerk MHM. PRAME-specific Allo-HLA-restricted T cells with potent antitumor reactivity useful for therapeutic T-cell receptor gene transfer. Clin Cancer Res 2011; 17:5615-25. [PMID: 21771875 DOI: 10.1158/1078-0432.ccr-11-1066] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE In human leukocyte antigen (HLA)-matched stem cell transplantation (SCT), it has been shown that beneficial immune response mediating graft-versus-tumor (GVT) responses can be separated from graft-versus-host disease (GVHD) immune responses. In this study, we investigated whether it would be possible to dissect the beneficial immune response of allo-HLA-reactive T cells with potent antitumor reactivity from GVHD-inducing T cells present in the detrimental immune response after HLA-mismatched SCT. EXPERIMENTAL DESIGN The presence of specific tumor-reactive T cells in the allo-HLA repertoire was analyzed at the time of severe GVHD after HLA-mismatched SCT, using tetramers composed of different tumor-associated antigens (TAA). RESULTS High-avidity allo-HLA-restricted T cells specific for the TAA preferentially expressed antigen on melanomas (PRAME) were identified that exerted highly single-peptide-specific reactivity. The T cells recognized multiple different tumor cell lines and leukemic cells, whereas no reactivity against a large panel of nonmalignant cells was observed. These T cells, however, also exerted low reactivity against mature dendritic cells (DC) and kidney epithelial cells, which was shown to be because of low PRAME expression. CONCLUSIONS On the basis of potential beneficial specificity and high reactivity, the T-cell receptors of these PRAME-specific T cells may be effective tools for adoptive T-cell therapy. Clinical studies have to determine the significance of the reactivity observed against mature DCs and kidney epithelial cells.
Collapse
Affiliation(s)
- Avital L Amir
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
A polymorphism in the splice donor site of ZNF419 results in the novel renal cell carcinoma-associated minor histocompatibility antigen ZAPHIR. PLoS One 2011; 6:e21699. [PMID: 21738768 PMCID: PMC3125305 DOI: 10.1371/journal.pone.0021699] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/05/2011] [Indexed: 01/12/2023] Open
Abstract
Nonmyeloablative allogeneic stem cell transplantation (SCT) can induce remission in patients with renal cell carcinoma (RCC), but this graft-versus-tumor (GVT) effect is often accompanied by graft-versus-host disease (GVHD). Here, we evaluated minor histocompatibility antigen (MiHA)-specific T cell responses in two patients with metastatic RCC who were treated with reduced-intensity conditioning SCT followed by donor lymphocyte infusion (DLI). One patient had stable disease and emergence of SMCY.A2-specific CD8+ T cells was observed after DLI with the potential of targeting SMCY-expressing RCC tumor cells. The second patient experienced partial regression of lung metastases from whom we isolated a MiHA-specific CTL clone with the capability of targeting RCC cell lines. Whole genome association scanning revealed that this CTL recognizes a novel HLA-B7-restricted MiHA, designated ZAPHIR, resulting from a polymorphism in the splice donor site of the ZNF419 gene. Tetramer analysis showed that emergence of ZAPHIR-specific CD8+ T cells in peripheral blood occurred in the absence of GVHD. Furthermore, the expression of ZAPHIR in solid tumor cell lines indicates the involvement of ZAPHIR-specific CD8+ T cell responses in selective GVT immunity. These findings illustrate that the ZNF419-encoded MiHA ZAPHIR is an attractive target for specific immunotherapy after allogeneic SCT.
Collapse
|
85
|
Broen K, Greupink-Draaisma A, Woestenenk R, Schaap N, Brickner AG, Dolstra H. Concurrent detection of circulating minor histocompatibility antigen-specific CD8+ T cells in SCT recipients by combinatorial encoding MHC multimers. PLoS One 2011; 6:e21266. [PMID: 21731686 PMCID: PMC3123304 DOI: 10.1371/journal.pone.0021266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 05/26/2011] [Indexed: 12/21/2022] Open
Abstract
Allogeneic stem cell transplantation (SCT) is a potentially curative treatment for patients with hematologic malignancies. Its therapeutic effect is largely dependent on recognition of minor histocompatibility antigens (MiHA) by donor-derived CD8+ T cells. Therefore, monitoring of multiple MiHA-specific CD8+ T cell responses may prove to be valuable for evaluating the efficacy of allogeneic SCT. In this study, we investigated the use of the combinatorial encoding MHC multimer technique to simultaneously detect MiHA-specific CD8+ T cells in peripheral blood of SCT recipients. Feasibility of this approach was demonstrated by applying dual-color encoding MHC multimers for a set of 10 known MiHA. Interestingly, single staining using a fluorochrome- and Qdot-based five-color combination showed comparable results to dual-color staining for most MiHA-specific CD8+ T cell responses. In addition, we determined the potential value of combinatorial encoding MHC multimers in MiHA identification. Therefore, a set of 75 candidate MiHA peptides was predicted from polymorphic genes with a hematopoietic expression profile and further selected for high and intermediate binding affinity for HLA-A2. Screening of a large cohort of SCT recipients resulted in the detection of dual-color encoded CD8+ T cells following MHC multimer-based T cell enrichment and short ex vivo expansion. Interestingly, candidate MiHA-specific CD8+ T cell responses for LAG3 and TLR10 derived polymorphic peptides could be confirmed by genotyping of the respective SNPs. These findings demonstrate the potency of the combinatorial MHC multimer approach in the monitoring of CD8+ T cell responses to known and potential MiHA in limited amounts of peripheral blood from allogeneic SCT recipients.
Collapse
Affiliation(s)
- Kelly Broen
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, The Netherlands
| | - Annelies Greupink-Draaisma
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, The Netherlands
| | - Rob Woestenenk
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, The Netherlands
| | - Nicolaas Schaap
- Department of Hematology, Radboud University Nijmegen Medical Centre, The Netherlands
| | - Anthony G. Brickner
- Departments of Medicine and Immunology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Harry Dolstra
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, The Netherlands
- * E-mail:
| |
Collapse
|
86
|
Falkenburg JHF, Warren EH. Graft versus leukemia reactivity after allogeneic stem cell transplantation. Biol Blood Marrow Transplant 2011; 17:S33-8. [PMID: 21195308 DOI: 10.1016/j.bbmt.2010.11.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
87
|
Hudecek M, Anderson LD, Nishida T, Riddell SR. Adoptive T-cell therapy for B-cell malignancies. Expert Rev Hematol 2011; 2:517-32. [PMID: 21083018 DOI: 10.1586/ehm.09.47] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The success of allogeneic hematopoietic cell transplantation (HCT) for B-cell malignancies is evidence that these tumors can be eliminated by T lymphocytes. This has encouraged the development of specific adoptive T-cell therapy, both for augmenting the anti-tumor effect of HCT and for patients not undergoing HCT. T cells that are capable of recognizing antigens expressed on malignant B cells may be recruited from the endogenous repertoire or engineered to express tumor-targeting receptors. Critical insights into the qualities of T cells that enable their persistence and function in vivo have been derived, and obstacles to effective T-cell-mediated tumor eradication are being elucidated. These advances provide the tools to translate adoptive T-cell transfer into reliable clinical therapies.
Collapse
Affiliation(s)
- Michael Hudecek
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | | | | | | |
Collapse
|
88
|
Bleakley M, Riddell SR. Exploiting T cells specific for human minor histocompatibility antigens for therapy of leukemia. Immunol Cell Biol 2011; 89:396-407. [PMID: 21301477 PMCID: PMC3061548 DOI: 10.1038/icb.2010.124] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Minor histocompatibility (H) antigens are major targets of a graft-versus-leukemia (GVL) effect mediated by donor CD8(+) and CD4(+) T cells following allogeneic hematopoietic cell transplantation (HCT) between human leukocyte antigen identical individuals. In the 15 years since the first molecular characterization of human minor H antigens, significant strides in minor H antigen discovery have been made as a consequence of advances in cellular, genetic and molecular techniques. Much has been learned about the mechanisms of minor H antigen immunogenicity, their expression on normal and malignant cells, and their role in GVL responses. T cells specific for minor H antigens expressed on leukemic cells, including leukemic stem cells, can be isolated and expanded in vitro and infused into allogeneic HCT recipients to augment the GVL effect to prevent and treat relapse. The first report of the adoptive transfer of minor H antigen-specific T-cell clones to patients with leukemic relapse in 2010 illustrates the potential for the manipulation of alloreactivity for therapeutic benefit. This review describes the recent developments in T-cell recognition of human minor H antigens, and efforts to translate these discoveries to reduce leukemia relapse after allogeneic HCT.
Collapse
Affiliation(s)
- Marie Bleakley
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-981024, USA.
| | | |
Collapse
|
89
|
Bleakley M, Riddell SR. Exploiting T cells specific for human minor histocompatibility antigens for therapy of leukemia. Immunol Cell Biol 2011. [PMID: 21301477 DOI: 10.1038/icb.2010.124.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Minor histocompatibility (H) antigens are major targets of a graft-versus-leukemia (GVL) effect mediated by donor CD8(+) and CD4(+) T cells following allogeneic hematopoietic cell transplantation (HCT) between human leukocyte antigen identical individuals. In the 15 years since the first molecular characterization of human minor H antigens, significant strides in minor H antigen discovery have been made as a consequence of advances in cellular, genetic and molecular techniques. Much has been learned about the mechanisms of minor H antigen immunogenicity, their expression on normal and malignant cells, and their role in GVL responses. T cells specific for minor H antigens expressed on leukemic cells, including leukemic stem cells, can be isolated and expanded in vitro and infused into allogeneic HCT recipients to augment the GVL effect to prevent and treat relapse. The first report of the adoptive transfer of minor H antigen-specific T-cell clones to patients with leukemic relapse in 2010 illustrates the potential for the manipulation of alloreactivity for therapeutic benefit. This review describes the recent developments in T-cell recognition of human minor H antigens, and efforts to translate these discoveries to reduce leukemia relapse after allogeneic HCT.
Collapse
Affiliation(s)
- Marie Bleakley
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-981024, USA.
| | | |
Collapse
|
90
|
Harnessing dendritic cells to improve allogeneic hematopoietic cell transplantation outcome. Semin Immunol 2011; 23:50-7. [DOI: 10.1016/j.smim.2011.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 01/05/2011] [Indexed: 11/20/2022]
|
91
|
Abstract
For the last two decades the immunotherapy of patients with solid and hematopoietic tumors has met with variable success. We have reviewed the field of tumor vaccines to examine what has worked and what has not, why this has been the case, how the anti-tumor responses were examined, and how we can make tumor immunity successful for the majority of individuals rather than for the exceptional patients who currently show successful immune responses against their tumors.
Collapse
Affiliation(s)
- Jan Joseph Melenhorst
- Stem Cell Allogeneic Transplant Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
92
|
Blood. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
93
|
Wright SE, Rewers-Felkins KA, Quinlin IS, Phillips CA, Townsend M, Philip R, Zorsky P, Klug P, Dai L, Hussain M, Thomas AA, Sundaramurthy C. Number of treatment cycles influences development of cytotoxic T cells in metastatic breast cancer patients - a phase I/II study. Immunol Invest 2010; 39:570-86. [PMID: 20653425 DOI: 10.3109/08820131003713798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The influence of the number of apheresis-stimulation-infusion(s) cycles, and the time in culture before the infusion (one vs. two weeks), on the generation of tumor antigen-specific cytotoxic T-lymphocytes (CTL) was investigated in a phase I/II clinical adoptive immunotherapy trial. Two previously treated metastatic breast cancer patients with no evidence of disease, in complete remission (CR), were enrolled. Each apheretic peripheral blood mononuclear cell (PBMC) sample was stimulated twice with MUC-1 before infusion back into the patients. Killer T-cells responses against MUC-1-expressing MCF-7 (CTL), nonspecific natural killer (NK) and lymphokine-activated killer (LAK) target cell lines, as well as, cytokine production were measured before each infusion. Patients received 2 infusions per month for 4 months. There were no tumor recurrences or toxicity. CTL, NK and LAK cells, type 1 cytokine, gamma-interferon (G-INF), and CD4(+) and CD8(+) memory T-lymphocytes were initially generated, produced or induced, respectively, and then declined. The CTL, NK and LAK cells were only induced at the first infusion of the first month. Thus, maintaining PBMC in culture longer than the first infusion was of no benefit with regards to retaining functional killer T-cells. In conclusion, this study implies that one treatment is optimal.
Collapse
Affiliation(s)
- Stephen E Wright
- Medical Service, Amarillo Veterans Affairs Health Care System, Amarillo, TX 79106, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Ma Q, Wang C, Jones D, Quintanilla KE, Li D, Wang Y, Wieder ED, Clise-Dwyer K, Alatrash G, Mj Y, Munsell MF, Lu S, Qazilbash MH, Molldrem JJ. Adoptive transfer of PR1 cytotoxic T lymphocytes associated with reduced leukemia burden in a mouse acute myeloid leukemia xenograft model. Cytotherapy 2010; 12:1056-62. [PMID: 20735170 PMCID: PMC3365857 DOI: 10.3109/14653249.2010.506506] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Tumor antigen-specific cytotoxic T lymphocytes (CTL) have been used in the treatment of human cancer, including leukemia. Several studies have established PR1 peptide, an HLA-A2.1-restricted peptide derived from proteinase 3 (P3), as a human leukemia-associated antigen. PR1-specific CTL elicited in vitro from healthy donors have been shown to lyse P3-expressing AML cells from patients. We investigated whether PR1-CTL can be adoptively transferred into NOD/SCID mice to eliminate human leukemia cells. METHODS PR1-CTL were generated in bulk culture from peripheral blood mononuclear cells (PBMC) stimulated with autologous dendritic cells. Human acute myeloid leukemia (AML) patient samples were injected and engrafted in murine bone marrow at 2 weeks post-transfer. RESULTS Following adoptive transfer, bone marrow aspirate from mice that received AML alone had 72-88% blasts in a hypercellular marrow, whereas mice that received AML plus PR1-CTL co-infusion had normal hematopoietic elements and only 3-18% blasts in a hypocellular marrow. The PR1-CTL persisted in the bone marrow and liver and maintained a CD45RA⁻CD28+ effector phenotype. CONCLUSIONS We found that adoptive transfer of PR1-CTL generated in vitro is associated with reduced AML cells in NOD/SCID mice. PR1-CTL can migrate to the sites of disease and maintain their capacity to kill the AML cells. The surface phenotype of PR1-CTL was consistent with their trafficking pattern in both vascular and end-organ tissues.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Bone Marrow/growth & development
- Bone Marrow/pathology
- Cell Movement
- Cells, Cultured
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/pathology
- Disease Models, Animal
- Humans
- Immunophenotyping
- Immunotherapy, Adoptive
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Lymphocyte Activation
- Mice
- Mice, SCID
- Myeloblastin/immunology
- Myeloblastin/metabolism
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/pathology
- T-Lymphocytes, Cytotoxic/transplantation
- Transplantation, Heterologous
- Tumor Burden
Collapse
Affiliation(s)
- Qing Ma
- Section of Transplantation Immunology, Department of Stem Cell Transplantation and Cellular Therapy, University of Texas, MD Anderson Cancer Center, Unit 900, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Van Bergen CA, Rutten CE, Van Der Meijden ED, Van Luxemburg-Heijs SA, Lurvink EG, Houwing-Duistermaat JJ, Kester MG, Mulder A, Willemze R, Falkenburg JF, Griffioen M. High-Throughput Characterization of 10 New Minor Histocompatibility Antigens by Whole Genome Association Scanning. Cancer Res 2010; 70:9073-83. [DOI: 10.1158/0008-5472.can-10-1832] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
96
|
Lampen MH, Verweij MC, Querido B, van der Burg SH, Wiertz EJHJ, van Hall T. CD8+ T cell responses against TAP-inhibited cells are readily detected in the human population. THE JOURNAL OF IMMUNOLOGY 2010; 185:6508-17. [PMID: 20980626 DOI: 10.4049/jimmunol.1001774] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Target cell recognition by CTLs depends on the presentation of peptides by HLA class I molecules. Tumors and herpes viruses have adopted strategies to greatly hamper this peptide presentation at the important bottleneck, the peptide transporter TAP. Previously, we described the existence of a CD8(+) CTL subpopulation that selectively recognizes such TAP-deficient cells in mouse models. In this study, we show that the human counterpart of this CTL subset is readily detectable in healthy subjects. Autologous PBMC cultures were initiated with dendritic cells rendered TAP-impaired by gene transfer of the viral evasion molecule UL49.5. Strikingly, specific reactivity to B-LCLs expressing one of the other viral TAP-inhibitors (US6, ICP47, or BNLF2a) was already observed after three rounds of stimulation. These short-term T cell cultures and isolated CD8(+) CTL clones derived thereof did not recognize the normal B-LCL, indicating that the cognate peptide-epitopes emerge at the cell surface upon an inhibition in the MHC class I processing pathway. A diverse set of TCRs was used by the clones, and the cellular reactivity was TCR-dependent and HLA class I-restricted, implying the involvement of a broad antigenic peptide repertoire. Our data indicate that the human CD8(+) T cell pool comprises a diverse reactivity to target cells with impairments in the intracellular processing pathway, and these might be exploited for cancers that are associated with such defects and for infections with immune-evading herpes viruses.
Collapse
Affiliation(s)
- Margit H Lampen
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
97
|
Minor histocompatibility antigens: presentation principles, recognition logic and the potential for a healing hand. Curr Opin Organ Transplant 2010; 15:512-25. [PMID: 20616723 DOI: 10.1097/mot.0b013e32833c1552] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW There is ample evidence indicating a pathologic role for minor histocompatibility antigens in inciting graft-versus-host disease in major histocompatibility complex (MHC)-matched bone marrow transplantation and rejection of solid organ allografts. Here we review the current knowledge of the genetic and biochemical bases for the cause of minor histoincompatibility and the structural basis for the recognition of the resulting alloantigens by the T-cell receptor. RECENT FINDINGS Recent evidence indicates that we as independently conceived individuals are genetically unique, thus, offering a mechanism for minor histoincompatibility between MHC-identical donor-recipient pairs. Furthermore, advances in delineating the mechanisms underlying antigen cross-presentation by MHC class I molecules and a critical role for autophagy in presenting cytoplasmic antigens by MHC class II molecules have been made. These new insights coupled with the X-ray crystallographic solution of several peptide/MHC-T-cell receptor structures have revealed mechanisms of histoincompatibility. SUMMARY On the basis of these new insights, ways to test for allograft compatibility and concoction of immunotherapies are discussed.
Collapse
|
98
|
Hambach L, Aghai Z, Pool J, Kröger N, Goulmy E. Peptide length extension skews the minor HA-1 antigen presentation toward activated dendritic cells but reduces its presentation efficiency. THE JOURNAL OF IMMUNOLOGY 2010; 185:4582-9. [PMID: 20855877 DOI: 10.4049/jimmunol.1000213] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Minor histocompatibility Ags (mHags) are important targets of the graft-versus-leukemia effect after HLA-matched allogeneic stem cell transplantation. mHags are HLA-restricted polymorphic peptides expressed on normal and leukemia cells. Vaccination with hematopoiesis-restricted mHag peptides, such as HA-1, may boost the graft-versus-leukemia effect. However, some animal studies indicate that peptides exactly reflecting immunogenic T cell epitopes (short peptides [SPs]) induce tolerance that is potentially due to systemic Ag spreading. Peptide length extension (long peptides [LPs]) may optimize immune responses by restricting and prolonging Ag presentation on dendritic cells (DCs). In this study, we compared the in vitro characteristics and T cell-stimulatory capacities of a human 30-mer HA-1 LP with the 9-mer HA-1 SP. DCs presented the HA-1 LP and SP and expanded HA-1-specific cytotoxic T cell lines. As hypothesized, HA-1 LP presentation, but not SP presentation, was largely restricted to activated DCs and was nearly absent on other hematopoietic cells. However, DCs presented the HA-1 LP 2-3 log levels less efficiently than the SP. Finally, the decay of HA-1 LP and SP presentation on DCs was comparable. We conclude that HA-1 LP and SP differ in their in vitro characteristics and that only comparative clinical studies after allogeneic stem cell transplantation may reveal the optimal HA-1 vaccine.
Collapse
Affiliation(s)
- Lothar Hambach
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
99
|
Lio HY, Tang JL, Wu J, Wu SJ, Lin CY, Yang YC. Minor histocompatibility antigen HA-1 and HA-2 polymorphisms in Taiwan: frequency and application in hematopoietic stem cell transplantation. Clin Chem Lab Med 2010; 48:1287-93. [PMID: 20509834 DOI: 10.1515/cclm.2010.246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Minor histocompatibility antigens influence the occurrence of graft-vs.-host disease and graft-vs.-leukemia effects after hematopoietic stem cell transplantation (HSCT). We determined the population frequencies of HA-1 and HA-2 alleles in Taiwan and exploited their potential applications in allogeneic HSCT. METHODS HA-1 and HA-2 were genotyped using polymerase chain reaction and restriction fragment length polymorphism in healthy controls (221 for HA-1 and 306 for HA-2) and HLA-matched donor-recipient sibling pairs with HSCT (92 for HA-1 and 38 for HA-2). The association of genetic polymorphisms with HSCT outcome was evaluated by univariate and multivariate analyses. RESULTS The allele frequencies in controls were 35.3% and 64.7% for HA-1(H) and HA-1(R), and 89.0% and 11.0% for HA-2(V) and HA-2(M), respectively. HA-1 disparity was denoted in 16.3% of HLA-matched donor-recipient sibling pairs, while it was not associated with HSCT outcome. HA-2 disparity was not observed in the donor-recipient pairs studied. The possibilities of using HA-1 and HA-2 variabilities as molecular markers for hematopoietic chimerism after HSCT were 39.2% and 18.4%, respectively. CONCLUSIONS Our data provide the information on allele and genotype frequencies of HA-1 and HA-2 in a Taiwanese population, and suggest that prospective genomic typing for HA-1 and HA-2 alleles of the donor and recipient could be a useful approach for molecular identification of hematopoietic chimerism after HSCT, rather than prognosis of clinical outcome.
Collapse
Affiliation(s)
- Hoi-Yan Lio
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
100
|
O'Reilly RJ, Dao T, Koehne G, Scheinberg D, Doubrovina E. Adoptive transfer of unselected or leukemia-reactive T-cells in the treatment of relapse following allogeneic hematopoietic cell transplantation. Semin Immunol 2010; 22:162-72. [PMID: 20537908 DOI: 10.1016/j.smim.2010.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 02/15/2010] [Indexed: 10/19/2022]
Abstract
Adoptive transfer of in vivo generated antigen-specific donor-derived T-cells is increasingly recognized as an effective approach for the treatment or prevention of EBV lymphomas and cytomegalovirus infections complicating allogeneic hematopoietic cell transplants. This review examines evidence from preclinical experiments and initial clinical trials to critically assess both the potential and current limitations of adoptive transfer of donor T-cells sensitized to selected minor alloantigens of the host or to peptide epitopes of proteins, differentially expressed by clonogenic leukemia cells, such as the Wilms tumor protein, WT-1, as a strategy to treat or prevent recurrence of leukemia in the post-transplant period.
Collapse
Affiliation(s)
- Richard J O'Reilly
- The Transplantation and Leukemia Service of the Department of Medicine and the Immunology and Molecular Pharmacology Programs at Memorial Sloan Kettering Cancer Center, United States.
| | | | | | | | | |
Collapse
|