1
|
Olsen KS, Jadi O, Dexheimer S, Bortone DS, Vensko SP, Bennett S, Tang H, Diiorio M, Saran T, Dingfelder D, Zhu Q, Wang Y, Haiman CA, Pooler L, Sheng X, Webb A, Pasquini MC, McCarthy PL, Spellman SR, Weimer E, Hahn T, Sucheston-Campbell L, Armistead PM, Vincent BG. Shared graft-versus-leukemia minor histocompatibility antigens in DISCOVeRY-BMT. Blood Adv 2023; 7:1635-1649. [PMID: 36477467 PMCID: PMC10182302 DOI: 10.1182/bloodadvances.2022008863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/07/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022] Open
Abstract
T-cell responses to minor histocompatibility antigens (mHAs) mediate graft-versus-leukemia (GVL) effects and graft-versus-host disease (GVHD) in allogeneic hematopoietic cell transplantation. Therapies that boost T-cell responses improve allogeneic hematopoietic cell transplant (alloHCT) efficacy but are limited by concurrent increases in the incidence and severity of GVHD. mHAs with expression restricted to hematopoietic tissue (GVL mHAs) are attractive targets for driving GVL without causing GVHD. Prior work to identify mHAs has focused on a small set of mHAs or population-level single-nucleotide polymorphism-association studies. We report the discovery of a large set of novel GVL mHAs based on predicted immunogenicity, tissue expression, and degree of sharing among donor-recipient pairs (DRPs) in the DISCOVeRY-BMT data set of 3231 alloHCT DRPs. The total number of predicted mHAs varied by HLA allele, and the total number and number of each class of mHA significantly differed by recipient genomic ancestry group. From the pool of predicted mHAs, we identified the smallest sets of GVL mHAs needed to cover 100% of DRPs with a given HLA allele. We used mass spectrometry to search for high-population frequency mHAs for 3 common HLA alleles. We validated 24 predicted novel GVL mHAs that are found cumulatively within 98.8%, 60.7%, and 78.9% of DRPs within DISCOVeRY-BMT that express HLA-A∗02:01, HLA-B∗35:01, and HLA-C∗07:02, respectively. We confirmed the immunogenicity of an example novel mHA via T-cell coculture with peptide-pulsed dendritic cells. This work demonstrates that the identification of shared mHAs is a feasible and promising technique for expanding mHA-targeting immunotherapeutics.
Collapse
Affiliation(s)
- Kelly S. Olsen
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Microbiology and Immunology, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Othmane Jadi
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Sarah Dexheimer
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Dante S. Bortone
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Steven P. Vensko
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Sarah Bennett
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Microbiology and Immunology, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Hancong Tang
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - Marisa Diiorio
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Tanvi Saran
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - David Dingfelder
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Qianqian Zhu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Yiwen Wang
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Palo Alto, CA
| | - Christopher A. Haiman
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA
| | - Loreall Pooler
- The Center for Genetic Epidemiology, University of South California, Los Angeles, CA
| | - Xin Sheng
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA
| | - Amy Webb
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH
| | - Marcelo C. Pasquini
- Center for International Blood and Marrow Transplant Research and Medical College of Wisconsin, Milwaukee, WI
| | - Philip L. McCarthy
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Stephen R. Spellman
- National Marrow Donor Program, Center for International Blood and Marrow Transplant Research, Minneapolis, MN
| | - Eric Weimer
- Department of Pathology & Laboratory Medicine, UNC School of Medicine, Chapel Hill, NC
| | - Theresa Hahn
- Department of Cancer Prevention & Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Lara Sucheston-Campbell
- College of Pharmacy, The Ohio State University, Columbus, OH
- College of Veterinary Medicine, The Ohio State University, Columbus, OH
| | - Paul M. Armistead
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Microbiology and Immunology, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Computational Medicine Program, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
2
|
Said R, Sellami MH, Kaabi H, Hmida S. Minor histocompatibility antigens HA-8 and PANE1 in the TUNISIAN population. Mol Genet Genomic Med 2022; 10:e2050. [PMID: 36036171 PMCID: PMC9651600 DOI: 10.1002/mgg3.2050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/05/2022] [Accepted: 08/22/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Minor histocompatibility antigens (mHAgs) are endogenous immunogenic peptides initially identified due to complications detected in several contexts of HLA geno-identical hematopoietic stem cell transplantation (HSCT). In this study, we chose to examine the molecular polymorphism of the mHAgs HA-8 and PANE1 in the Tunisian population. MATERIAL AND METHODS This study was conducted on 150 healthy and unrelated individuals. The DNA extraction and Sequence-Specific Primers PCR (PCR-SSP) methods were used for the molecular genotyping of the selected SNPs: PUM3 (rs2173904) and CENPM (rs5758511). RESULTS Our results show that, 94% of Tunisians are carriers of the PANE1R allele (immunogenic variant of the PANE1 mHAg) and 68% of Tunisians are carriers of the HA-8R allele (immunogenic variant of the HA-8 mHAg). Furthermore, this study shows that about 5% of the Tunisians are carrier of the PANE1R antigen and its HLA molecule of presentation (the PANE1R/HLA-A*0301 combination). However, only 2% of Tunisians are carrier of the HA-8R/HLA-A*0201 combination, that is, the HA8 immunogenic variant and its specific HLA molecule of presentation. CONCLUSION Our results are close to those reported in Caucasian, Asiatic, and African populations, this may be explained by the historical events experienced by Tunisia for millennia. These results could be used for further clinical and anthropological studies.
Collapse
Affiliation(s)
- Rahma Said
- Department of ImmunohaematologyNational Blood Transfusion Center of TunisTunisTunisia
| | | | - Houda Kaabi
- Department of ImmunohaematologyNational Blood Transfusion Center of TunisTunisTunisia
| | - Slama Hmida
- Department of ImmunohaematologyNational Blood Transfusion Center of TunisTunisTunisia
| |
Collapse
|
3
|
Giesler S, Zeiser R. Deciphering the role of Minor histocompatibility antigens for acute graft-versus-host disease. Transplant Cell Ther 2021; 27:523-524. [PMID: 34210498 DOI: 10.1016/j.jtct.2021.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Sophie Giesler
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; Comprehensive Cancer Center Freiburg (CCCF), Medical Center- University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; Comprehensive Cancer Center Freiburg (CCCF), Medical Center- University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK) Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany; Signalling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg.
| |
Collapse
|
4
|
Mossallam GI, Fattah RA, Bokhary M, Moneer M, Mahmoud HK. LMP7 polymorphism may modify the presentation and clinical impact of minor histocompatibility antigens in matched related hematopoietic stem cell transplantation. Cell Immunol 2021; 364:104329. [PMID: 33798908 DOI: 10.1016/j.cellimm.2021.104329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/19/2021] [Accepted: 03/02/2021] [Indexed: 10/22/2022]
Abstract
Differential expression of minor histocompatibility antigens between the recipient and donor determines their disparity and can be modified by immunoproteasomes that regulate their processing and presentation. We examined the impact of HA-1 and HA-8 disparity, and immunoproteasome LMP7 polymorphism in 130 pairs. In multivariate analysis, HA-1 disparity showed a statistically significant association with an increased incidence of acute graft-versus-host disease (aGVHD) II-IV (p = 0.043, HR: 3.71, 95%CI = 1.04-13.26), while LMP7-Q/Q showed a trend toward increased incidence of aGVHD compared to LMP7-Q/K and K/K genotypes (p = 0.087, HR: 2.36, 95%CI = 0.88-6.31). All HA-1 and HA-8 disparate patients who developed aGVHD had the LMP7-Q/Q genotype. No significant association could be detected between HA-1, HA-8, or LMP7 and chronic GVHD, relapse-free survival (RFS), overall survival (OS), or transplant-related mortality (TRM). In conclusion, we suggested an association between the HA-1 disparity and the risk of developing aGVHD with a possible modifying effect of LMP7.
Collapse
Affiliation(s)
- Ghada I Mossallam
- Bone Marrow Transplantation Laboratory Unit, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Raafat Abdel Fattah
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt; Bone Marrow Transplantation Unit, Nasser Institute Hospital for Research and Treatment, Cairo, Egypt
| | - Mahmoud Bokhary
- Bone Marrow Transplantation Unit, Nasser Institute Hospital for Research and Treatment, Cairo, Egypt
| | - Manar Moneer
- Department of Epidemiology and Biostatistics, National Cancer Institute, Cairo University, Egypt
| | - Hossam K Mahmoud
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt; Bone Marrow Transplantation Unit, Nasser Institute Hospital for Research and Treatment, Cairo, Egypt
| |
Collapse
|
5
|
Zhang J, Teramoto T, Qiu C, Wine RN, Gonzalez LE, Baserga SJ, Tanaka Hall TM. Nop9 recognizes structured and single-stranded RNA elements of preribosomal RNA. RNA (NEW YORK, N.Y.) 2020; 26:1049-1059. [PMID: 32371454 PMCID: PMC7373996 DOI: 10.1261/rna.075416.120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/29/2020] [Indexed: 05/04/2023]
Abstract
Nop9 is an essential factor in the processing of preribosomal RNA. Its absence in yeast is lethal, and defects in the human ortholog are associated with breast cancer, autoimmunity, and learning/language impairment. PUF family RNA-binding proteins are best known for sequence-specific RNA recognition, and most contain eight α-helical repeats that bind to the RNA bases of single-stranded RNA. Nop9 is an unusual member of this family in that it contains eleven repeats and recognizes both RNA structure and sequence. Here we report a crystal structure of Saccharomyces cerevisiae Nop9 in complex with its target RNA within the 20S preribosomal RNA. This structure reveals that Nop9 brings together a carboxy-terminal module recognizing the 5' single-stranded region of the RNA and a bifunctional amino-terminal module recognizing the central double-stranded stem region. We further show that the 3' single-stranded region of the 20S target RNA adds sequence-independent binding energy to the RNA-Nop9 interaction. Both the amino- and carboxy-terminal modules retain the characteristic sequence-specific recognition of PUF proteins, but the amino-terminal module has also evolved a distinct interface, which allows Nop9 to recognize either single-stranded RNA sequences or RNAs with a combination of single-stranded and structured elements.
Collapse
Affiliation(s)
- Jun Zhang
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Takamasa Teramoto
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Chen Qiu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Robert N Wine
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Lauren E Gonzalez
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Susan J Baserga
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06520, USA
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Traci M Tanaka Hall
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
6
|
Mutis T, Xagara A, Spaapen RM. The Connection Between Minor H Antigens and Neoantigens and the Missing Link in Their Prediction. Front Immunol 2020; 11:1162. [PMID: 32670277 PMCID: PMC7326952 DOI: 10.3389/fimmu.2020.01162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/12/2020] [Indexed: 12/26/2022] Open
Abstract
For hundreds of thousands of years, the human genome has extensively evolved, resulting in genetic variations in almost every gene. Immunological reflections of these genetic variations become clearly visible after an allogeneic stem cell transplantation (allo-SCT) as minor Histocompatibility (H) antigens. Minor H antigens are peptides cleaved from genetically encoded variable protein regions after which they are presented at the cell surface by HLA molecules. After allo-SCT with minor H antigen mismatches between donor and recipient, donor T cells recognize the minor H antigens of the recipient as foreign, evoking strong alloreactive immune responses. Studies in the late eighties have discovered that a subset of minor H antigens are encoded by hematopoietic system-specific genes. After allo-SCT, this subset is strictly expressed on the hematopoietic malignant cells and was therefore the first well-defined highly immunogenic group of tumor-specific antigens. In the last decade, neoantigens derived from genetic mutations in tumors have been identified as another group of immunogenic tumor-specific antigens. Therefore, hematopoietic minor H antigens and neoantigens are therapeutic equivalents. This review will connect our current knowledge about the immune biology and identification of minor H antigens and neoantigens leading to novel conclusions on their prediction.
Collapse
Affiliation(s)
- Tuna Mutis
- Department of Hematology, Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands
| | - Anastasia Xagara
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
7
|
Fuchs KJ, Honders MW, van der Meijden ED, Adriaans AE, van der Lee DI, Pont MJ, Monajemi R, Kielbasa SM, 't Hoen PAC, van Bergen CAM, Falkenburg JHF, Griffioen M. Optimized Whole Genome Association Scanning for Discovery of HLA Class I-Restricted Minor Histocompatibility Antigens. Front Immunol 2020; 11:659. [PMID: 32362897 PMCID: PMC7180171 DOI: 10.3389/fimmu.2020.00659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/23/2020] [Indexed: 12/21/2022] Open
Abstract
Patients undergoing allogeneic stem cell transplantation as treatment for hematological diseases face the risk of Graft-versus-Host Disease as well as relapse. Graft-versus-Host Disease and the favorable Graft-versus-Leukemia effect are mediated by donor T cells recognizing polymorphic peptides, which are presented on the cell surface by HLA molecules and result from single nucleotide polymorphism alleles that are disparate between patient and donor. Identification of polymorphic HLA-binding peptides, designated minor histocompatibility antigens, has been a laborious procedure, and the number and scope for broad clinical use of these antigens therefore remain limited. Here, we present an optimized whole genome association approach for discovery of HLA class I minor histocompatibility antigens. T cell clones isolated from patients who responded to donor lymphocyte infusions after HLA-matched allogeneic stem cell transplantation were tested against a panel of 191 EBV-transformed B cells, which have been sequenced by the 1000 Genomes Project and selected for expression of seven common HLA class I alleles (HLA-A∗01:01, A∗02:01, A∗03:01, B∗07:02, B∗08:01, C∗07:01, and C∗07:02). By including all polymorphisms with minor allele frequencies above 0.01, we demonstrated that the new approach allows direct discovery of minor histocompatibility antigens as exemplified by seven new antigens in eight different HLA class I alleles including one antigen in HLA-A∗24:02 and HLA-A∗23:01, for which the method has not been originally designed. Our new whole genome association strategy is expected to rapidly augment the repertoire of HLA class I-restricted minor histocompatibility antigens that will become available for donor selection and clinical use to predict, follow or manipulate Graft-versus-Leukemia effect and Graft-versus-Host Disease after allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Kyra J Fuchs
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - M Willy Honders
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Edith D van der Meijden
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands.,Department of Internal Medicine, Hematology and Internal Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Alwin E Adriaans
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Margot J Pont
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands.,Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Ramin Monajemi
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| | - Szymon M Kielbasa
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| | - Peter A C 't Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands.,Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | | | | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
8
|
Kremer AN, Bausenwein J, Lurvink E, Kremer AE, Rutten CE, van Bergen CAM, Kretschmann S, van der Meijden E, Honders MW, Mazzeo D, Watts C, Mackensen A, Falkenburg JHF, Griffioen M. Discovery and Differential Processing of HLA Class II-Restricted Minor Histocompatibility Antigen LB-PIP4K2A-1S and Its Allelic Variant by Asparagine Endopeptidase. Front Immunol 2020; 11:381. [PMID: 32218783 PMCID: PMC7078166 DOI: 10.3389/fimmu.2020.00381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/17/2020] [Indexed: 12/03/2022] Open
Abstract
Minor histocompatibility antigens are the main targets of donor-derived T-cells after allogeneic stem cell transplantation. Identification of these antigens and understanding their biology are a key requisite for more insight into how graft vs. leukemia effect and graft vs. host disease could be separated. We here identified four new HLA class II-restricted minor histocompatibility antigens using whole genome association scanning. For one of the new antigens, i.e., LB-PIP4K2A-1S, we measured strong T-cell recognition of the donor variant PIP4K2A-1N when pulsed as exogenous peptide, while the endogenously expressed variant in donor EBV-B cells was not recognized. We showed that lack of T-cell recognition was caused by intracellular cleavage by a protease named asparagine endopeptidase (AEP). Furthermore, microarray gene expression analysis showed that PIP4K2A and AEP are both ubiquitously expressed in a wide variety of healthy tissues, but that expression levels of AEP were lower in primary acute myeloid leukemia (AML). In line with that, we confirmed low activity of AEP in AML cells and demonstrated that HLA-DRB1*03:01 positive primary AML expressing LB-PIP4K2A-1S or its donor variant PIP4K2A-1N were both recognized by specific T-cells. In conclusion, LB-PIP4K2A-1S not only represents a novel minor histocompatibility antigen but also provides evidence that donor T-cells after allogeneic stem cell transplantation can target the autologous allelic variant as leukemia-associated antigen. Furthermore, it demonstrates that endopeptidases can play a role in cell type-specific intracellular processing and presentation of HLA class II-restricted antigens, which may be explored in future immunotherapy of AML.
Collapse
Affiliation(s)
- Anita N. Kremer
- Department of Internal Medicine 5, Hematology/Oncology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Judith Bausenwein
- Department of Internal Medicine 5, Hematology/Oncology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Ellie Lurvink
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Andreas E. Kremer
- Department of Internal Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Caroline E. Rutten
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Sascha Kretschmann
- Department of Internal Medicine 5, Hematology/Oncology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Edith van der Meijden
- Department of Internal Medicine 5, Hematology/Oncology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Maria W. Honders
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Daniela Mazzeo
- Division of Cell Signaling & Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Colin Watts
- Division of Cell Signaling & Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology/Oncology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | | | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
9
|
Zong S, Mi T, Flores LG, Alpert A, Olivares S, Patel K, Maiti S, Mcnamara G, Cooper LJN, Torikai H. Very rapid cloning, expression and identifying specificity of T-cell receptors for T-cell engineering. PLoS One 2020; 15:e0228112. [PMID: 32040512 PMCID: PMC7010234 DOI: 10.1371/journal.pone.0228112] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/07/2020] [Indexed: 11/29/2022] Open
Abstract
Neoantigens can be predicted and in some cases identified using the data obtained from the whole exome sequencing and transcriptome sequencing of tumor cells. These sequencing data can be coupled with single-cell RNA sequencing for the direct interrogation of the transcriptome, surfaceome, and pairing of αβ T-cell receptors (TCRαβ) from hundreds of single T cells. Using these 2 large datasets, we established a platform for identifying antigens recognized by TCRαβs obtained from single T cells. Our approach is based on the rapid expression of cloned TCRαβ genes as Sleeping Beauty transposons and the determination of the introduced TCRαβs’ antigen specificity and avidity using a reporter cell line. The platform enables the very rapid identification of tumor-reactive TCRs for the bioengineering of T cells with redirected specificity.
Collapse
Affiliation(s)
- Shan Zong
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Tiejuan Mi
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Leo G. Flores
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Amir Alpert
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Simon Olivares
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Krina Patel
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sourindra Maiti
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - George Mcnamara
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Laurence J. N. Cooper
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Ziopharm Oncology, Inc., Boston, Massachusetts, United States of America
| | - Hiroki Torikai
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
10
|
Summers C, Sheth VS, Bleakley M. Minor Histocompatibility Antigen-Specific T Cells. Front Pediatr 2020; 8:284. [PMID: 32582592 PMCID: PMC7283489 DOI: 10.3389/fped.2020.00284] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/06/2020] [Indexed: 01/05/2023] Open
Abstract
Minor Histocompatibility (H) antigens are major histocompatibility complex (MHC)/Human Leukocyte Antigen (HLA)-bound peptides that differ between allogeneic hematopoietic stem cell transplantation (HCT) recipients and their donors as a result of genetic polymorphisms. Some minor H antigens can be used as therapeutic T cell targets to augment the graft-vs.-leukemia (GVL) effect in order to prevent or manage leukemia relapse after HCT. Graft engineering and post-HCT immunotherapies are being developed to optimize delivery of T cells specific for selected minor H antigens. These strategies have the potential to reduce relapse risk and thereby permit implementation of HCT approaches that are associated with less toxicity and fewer late effects, which is particularly important in the growing and developing pediatric patient. Most minor H antigens are expressed ubiquitously, including on epithelial tissues, and can be recognized by donor T cells following HCT, leading to graft-vs.-host disease (GVHD) as well as GVL. However, those minor H antigens that are expressed predominantly on hematopoietic cells can be targeted for selective GVL. Once full donor hematopoietic chimerism is achieved after HCT, hematopoietic-restricted minor H antigens are present only on residual recipient malignant hematopoietic cells, and these minor H antigens serve as tumor-specific antigens for donor T cells. Minor H antigen-specific T cells that are delivered as part of the donor hematopoietic stem cell graft at the time of HCT contribute to relapse prevention. However, in some cases the minor H antigen-specific T cells delivered with the graft may be quantitatively insufficient or become functionally impaired over time, leading to leukemia relapse. Following HCT, adoptive T cell immunotherapy can be used to treat or prevent relapse by delivering large numbers of donor T cells targeting hematopoietic-restricted minor H antigens. In this review, we discuss minor H antigens as T cell targets for augmenting the GVL effect in engineered HCT grafts and for post-HCT immunotherapy. We will highlight the importance of these developments for pediatric HCT.
Collapse
Affiliation(s)
- Corinne Summers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Vipul S Sheth
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Marie Bleakley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|
11
|
Romaniuk DS, Postovskaya AM, Khmelevskaya AA, Malko DB, Efimov GA. Rapid Multiplex Genotyping of 20 HLA-A *02:01 Restricted Minor Histocompatibility Antigens. Front Immunol 2019; 10:1226. [PMID: 31275297 PMCID: PMC6593292 DOI: 10.3389/fimmu.2019.01226] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
A subset of MHC-associated self-peptides presented by the recipient's cells and immunologically foreign to the donor can induce an allogeneic immune response after hematopoietic stem cell transplantation (HSCT). These immunogenic peptides originate from the genomic polymorphisms and are known as minor histocompatibility antigens (MiHA). MiHA mismatches trigger the post-transplant immune response, which could manifest in both the deleterious “graft-vs.-host” disease and the beneficial “graft-vs.-leukemia” effect. Importantly, some MiHAs are considered to be promising targets for posttransplant T-cell immunotherapy of hematopoietic malignancies. This creates a demand for a robust and fast approach to genotyping MiHA-encoding polymorphisms. We report a multiplex real-time PCR method for the genotyping of 20 polymorphisms that are encoding HLA-A*02:01-restricted MiHAs. This method uses allele-specific primers and gene-specific hydrolysis probes. In 1 h it allows for the detection of MiHA mismatches in a donor-recipient pair without the need for electrophoresis, sequencing, or other time-consuming techniques. We validated the method with Sanger and NGS sequencing and demonstrated good performance over a wide range of DNA concentrations. We propose our protocol as a fast and accurate method of identifying mismatched MiHAs. The information on the MiHA mismatches is useful for studying the allogeneic immune response following HSCT and for selecting the targets for post-transplant T-cell therapy.
Collapse
Affiliation(s)
- Dmitrii S Romaniuk
- Laboratory for Transplantation Immunology, National Research Center for Hematology, Moscow, Russia
| | - Anna M Postovskaya
- Laboratory for Transplantation Immunology, National Research Center for Hematology, Moscow, Russia
| | - Alexandra A Khmelevskaya
- Laboratory for Transplantation Immunology, National Research Center for Hematology, Moscow, Russia
| | - Dmitry B Malko
- Laboratory for Transplantation Immunology, National Research Center for Hematology, Moscow, Russia
| | - Grigory A Efimov
- Laboratory for Transplantation Immunology, National Research Center for Hematology, Moscow, Russia
| |
Collapse
|
12
|
Koyama D, Murata M, Hanajiri R, Akashi T, Okuno S, Kamoshita S, Julamanee J, Takagi E, Miyao K, Sakemura R, Goto T, Terakura S, Nishida T, Kiyoi H. Quantitative Assessment of T Cell Clonotypes in Human Acute Graft-versus-Host Disease Tissues. Biol Blood Marrow Transplant 2019; 25:417-423. [DOI: 10.1016/j.bbmt.2018.10.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/15/2018] [Indexed: 01/05/2023]
|
13
|
Bijen HM, Hassan C, Kester MGD, Janssen GMC, Hombrink P, de Ru AH, Drijfhout JW, Meiring HD, de Jong AP, Falkenburg JHF, Jimenez CR, Heemskerk MHM, van Veelen PA. Specific T Cell Responses against Minor Histocompatibility Antigens Cannot Generally Be Explained by Absence of Their Allelic Counterparts on the Cell Surface. Proteomics 2018; 18:e1700250. [PMID: 29251415 DOI: 10.1002/pmic.201700250] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/22/2017] [Indexed: 12/20/2022]
Abstract
Allogeneic stem cell transplantation has emerged as immunotherapy in the treatment of a variety of hematological malignancies. Its efficacy depends on induction of graft versus leukemia by donor lymphocytes. Both graft versus leukemia and graft versus host disease are induced by T cells reactive against polymorphic peptides, called minor histocompatibility antigens (MiHA), which differ between patient and donor and are presented in the context of self-HLA (where HLA is human leukocyte antigen). The allelic counterpart (AC) of the MiHA is generally considered to be absent at the cell surface, based on the absence of immune responses directed against the AC. To study this in detail, we evaluate the recognition, HLA-binding affinity, and cell surface expression of three selected MiHA. By quantitative MS, we demonstrate the similarly abundant expression of both MiHA and AC at the cell surface. We conclude that the absent recognition of the AC cannot generally be explained by insufficient processing and presentation at the cell surface of the AC.
Collapse
Affiliation(s)
- Helena M Bijen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Chopie Hassan
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Michel G D Kester
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - George M C Janssen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Pleun Hombrink
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arnoud H de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Wouter Drijfhout
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hugo D Meiring
- Laboratory for Vaccine Research, Unit Research and Development, Netherlands Vaccine Institute, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Ad P de Jong
- Laboratory for Vaccine Research, Unit Research and Development, Netherlands Vaccine Institute, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | | | - Connie R Jimenez
- OncoProteomics Laboratory, Medical Oncology, VU Medical Center, Amsterdam, The Netherlands
| | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
14
|
Thordardottir S, Schaap N, Louer E, Kester MGD, Falkenburg JHF, Jansen J, Radstake TRD, Hobo W, Dolstra H. Hematopoietic stem cell-derived myeloid and plasmacytoid DC-based vaccines are highly potent inducers of tumor-reactive T cell and NK cell responses ex vivo. Oncoimmunology 2017; 6:e1285991. [PMID: 28405517 PMCID: PMC5384421 DOI: 10.1080/2162402x.2017.1285991] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/14/2017] [Accepted: 01/18/2017] [Indexed: 12/28/2022] Open
Abstract
Because of the potent graft-versus-tumor (GVT) effect, allogeneic stem cell transplantation (alloSCT) can be a curative therapy for hematological malignancies. However, relapse remains the most frequent cause of treatment failure, illustrating the necessity for development of adjuvant post-transplant therapies to boost GVT immunity. Dendritic cell (DC) vaccination is a promising strategy in this respect, in particular, where distinct biologic functions of naturally occurring DC subsets, i.e. myeloid DCs (mDCs) and plasmacytoid DCs (pDCs), are harnessed. However, it is challenging to obtain high enough numbers of primary DC subsets from blood for immunotherapy due to their low frequencies. Therefore, we present here an ex vivo GMP-compliant cell culture protocol for generating different DC subsets from CD34+ hematopoietic stem and progenitor cells (HSPCs) of alloSCT donor origin. High numbers of BDCA1+ mDCs and pDCs could be generated, sufficient for multiple vaccination cycles. These HSPC-derived DC subsets were highly potent in inducing antitumor immune responses in vitro. Notably, HSPC-derived BDCA1+ mDCs were superior in eliciting T cell responses. They efficiently primed naïve T cells and robustly expanded patient-derived minor histocompatibility antigen (MiHA)-specific T cells. Though the HSPC-pDCs also efficiently induced T cell responses, they exhibited superior capacity in activating NK cells. pDC-primed NK cells highly upregulated TRAIL and possessed strong cytolytic capacity against tumor cells. Collectively, these findings indicate that HSPC-derived DC vaccines, comprising both mDCs and pDCs, may possess superior potential to boost antitumor immunity post alloSCT, due to their exceptional T cell and NK cell stimulatory capacity.
Collapse
Affiliation(s)
- Soley Thordardottir
- 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
| | - Elja Louer
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Michel G D Kester
- Department of Hematology, Leiden University Medical Center , Leiden, the Netherlands
| | | | - Joop Jansen
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Timothy R D Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, the Netherlands; Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine - Laboratory 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
|
15
|
Kanakry CG, Coffey DG, Towlerton AMH, Vulic A, Storer BE, Chou J, Yeung CCS, Gocke CD, Robins HS, O'Donnell PV, Luznik L, Warren EH. Origin and evolution of the T cell repertoire after posttransplantation cyclophosphamide. JCI Insight 2016; 1. [PMID: 27213183 DOI: 10.1172/jci.insight.86252] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Posttransplantation cyclophosphamide (PTCy) effectively prevents graft-versus-host disease (GVHD), but its immunologic impact is poorly understood. We assessed lymphocyte reconstitution via flow cytometry (n = 74) and antigen receptor sequencing (n = 35) in recipients of myeloablative, HLA-matched allogeneic BM transplantation using PTCy. Recovering T cells were primarily phenotypically effector memory with lower T cell receptor β (TRB) repertoire diversity than input donor repertoires. Recovering B cells were predominantly naive with immunoglobulin heavy chain locus (IGH) repertoire diversity similar to donors. Numerical T cell reconstitution and TRB diversity were strongly associated with recipient cytomegalovirus seropositivity. Global similarity between input donor and recipient posttransplant repertoires was uniformly low at 1-2 months after transplant but increased over the balance of the first posttransplant year. Blood TRB repertoires at ≥3 months after transplant were often dominated by clones present in the donor blood/marrow memory CD8+ compartment. Limited overlap was observed between the TRB repertoires of T cells infiltrating the skin or gastrointestinal tract versus the blood. Although public TRB sequences associated with herpesvirus- or alloantigen-specific CD8+ T cells were detected in some patients, posttransplant TRB and IGH repertoires were unique to each individual. These data define the immune dynamics occurring after PTCy and establish a benchmark against which immune recovery after other transplantation approaches can be compared.
Collapse
Affiliation(s)
- Christopher G Kanakry
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David G Coffey
- Clinical Research Division, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Andrea M H Towlerton
- Clinical Research Division, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA
| | - Ante Vulic
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Barry E Storer
- Clinical Research Division, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA
| | - Jeffrey Chou
- Clinical Research Division, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Cecilia C S Yeung
- Clinical Research Division, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA; Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Christopher D Gocke
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Harlan S Robins
- Public Health Sciences Division, FHCRC, Seattle, Washington, USA
| | - Paul V O'Donnell
- Clinical Research Division, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Leo Luznik
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Edus H Warren
- Clinical Research Division, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
16
|
Lipkin E, Strillacci MG, Eitam H, Yishay M, Schiavini F, Soller M, Bagnato A, Shabtay A. The Use of Kosher Phenotyping for Mapping QTL Affecting Susceptibility to Bovine Respiratory Disease. PLoS One 2016; 11:e0153423. [PMID: 27077383 PMCID: PMC4831767 DOI: 10.1371/journal.pone.0153423] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 03/29/2016] [Indexed: 02/06/2023] Open
Abstract
Bovine respiratory disease (BRD) is the leading cause of morbidity and mortality in feedlot cattle, caused by multiple pathogens that become more virulent in response to stress. As clinical signs often go undetected and various preventive strategies failed, identification of genes affecting BRD is essential for selection for resistance. Selective DNA pooling (SDP) was applied in a genome wide association study (GWAS) to map BRD QTLs in Israeli Holstein male calves. Kosher scoring of lung adhesions was used to allocate 122 and 62 animals to High (Glatt Kosher) and Low (Non-Kosher) resistant groups, respectively. Genotyping was performed using the Illumina BovineHD BeadChip according to the Infinium protocol. Moving average of -logP was used to map QTLs and Log drop was used to define their boundaries (QTLRs). The combined procedure was efficient for high resolution mapping. Nineteen QTLRs distributed over 13 autosomes were found, some overlapping previous studies. The QTLRs contain polymorphic functional and expression candidate genes to affect kosher status, with putative immunological and wound healing activities. Kosher phenotyping was shown to be a reliable means to map QTLs affecting BRD morbidity.
Collapse
Affiliation(s)
- Ehud Lipkin
- Department of Genetics, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Harel Eitam
- Department of Ruminant Sciences, Agricultural Research Organization (ARO), Bet-Dagan, Israel
| | - Moran Yishay
- Department of Ruminant Sciences, Agricultural Research Organization (ARO), Bet-Dagan, Israel
| | | | - Morris Soller
- Department of Genetics, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Ariel Shabtay
- Department of Ruminant Sciences, Agricultural Research Organization (ARO), Bet-Dagan, Israel
| |
Collapse
|
17
|
Griffioen M, van Bergen CAM, Falkenburg JHF. Autosomal Minor Histocompatibility Antigens: How Genetic Variants Create Diversity in Immune Targets. Front Immunol 2016; 7:100. [PMID: 27014279 PMCID: PMC4791598 DOI: 10.3389/fimmu.2016.00100] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 03/01/2016] [Indexed: 11/13/2022] Open
Abstract
Allogeneic stem cell transplantation (alloSCT) can be a curative treatment for hematological malignancies. Unfortunately, the desired anti-tumor or graft-versus-leukemia (GvL) effect is often accompanied with undesired side effects against healthy tissues known as graft-versus-host disease (GvHD). After HLA-matched alloSCT, GvL and GvHD are both mediated by donor-derived T-cells recognizing polymorphic peptides presented by HLA surface molecules on patient cells. These polymorphic peptides or minor histocompatibility antigens (MiHA) are produced by genetic differences between patient and donor. Since polymorphic peptides may be useful targets to manipulate the balance between GvL and GvHD, the dominant repertoire of MiHA needs to be discovered. In this review, the diversity of autosomal MiHA characterized thus far as well as the various molecular mechanisms by which genetic variants create immune targets and the role of cryptic transcripts and proteins as antigen sources are described. The tissue distribution of MiHA as important factor in GvL and GvHD is considered as well as possibilities how hematopoietic MiHA can be used for immunotherapy to augment GvL after alloSCT. Although more MiHA are still needed for comprehensive understanding of the biology of GvL and GvHD and manipulation by immunotherapy, this review shows insight into the composition and kinetics of in vivo immune responses with respect to specificity, diversity, and frequency of specific T-cells and surface expression of HLA-peptide complexes and other (accessory) molecules on the target cell. A complex interplay between these factors and their environment ultimately determines the spectrum of clinical manifestations caused by immune responses after alloSCT.
Collapse
Affiliation(s)
- Marieke Griffioen
- Department of Hematology, Leiden University Medical Center , Leiden , Netherlands
| | | | | |
Collapse
|
18
|
Oostvogels R, Lokhorst HM, Mutis T. Minor histocompatibility Ags: identification strategies, clinical results and translational perspectives. Bone Marrow Transplant 2015; 51:163-71. [PMID: 26501766 DOI: 10.1038/bmt.2015.256] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 08/11/2015] [Accepted: 08/15/2015] [Indexed: 12/14/2022]
Abstract
Allogeneic stem cell transplantation (allo-SCT) and donor lymphocyte infusion are effective treatment modalities for various hematological malignancies. Their therapeutic effect, the graft-versus-tumor (GvT) effect, is based mainly on an alloimmune response of donor T cells directed at tumor cells, in which differences in the expression of minor histocompatibility Ags (mHags) on the cells of the patient and donor have a crucial role. However, these differences are also responsible for induction of sometimes detrimental GvHD. As relapse and development of GvHD pose major threats for a large proportion of allotransplanted patients, additional therapeutic strategies are required. To augment the GvT response without increasing the risk of GvHD, specific mHag-directed immunotherapeutic strategies have been developed. Over the past years, much effort has been put into the identification of therapeutically relevant mHags to enable these strategies for a substantial proportion of patients. Currently, the concept of mHag-directed immunotherapy is tested in clinical trials on feasibility, safety and efficacy. In this review, we will summarize the recent developments in mHag identification and the clinical data on mHag-specific immune responses and mHag-directed therapies in patients with hematological malignancies. Finally, we will outline the current challenges and future prospectives in the field.
Collapse
Affiliation(s)
- R Oostvogels
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - H M Lokhorst
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - T Mutis
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Spierings E. Minor histocompatibility antigens: past, present, and future. ACTA ACUST UNITED AC 2015; 84:374-60. [PMID: 25262921 DOI: 10.1111/tan.12445] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Indexed: 01/02/2023]
Abstract
Minor histocompatibility (H) antigens are key molecules driving allo-immune responses in both graft-versus-host-disease (GvHD) and in graft-versus-leukemia (GvL) reactivity in human leukocyte antigen (HLA)-matched hematopoietic stem-cell transplantation (HSCT). 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 can cause both GvHD and GvL effects, while hematopoietic system-restricted minor H antigens are more prone to induce GvL responses. This phenomenon renders the latter group of minor H antigens as curative tools for HSCT-based immunotherapy of hematological malignancies and disorders, in which minor H antigen-specific responses are enhanced in order to eradicate the malignant cells. This article describes the immunogenetics of minor H antigens and methods that have been developed to identify them. Moreover, it summarizes the clinical relevance of minor H antigens in transplantation, with special regards to allogeneic HSCT and solid-organ transplantation.
Collapse
Affiliation(s)
- Eric Spierings
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
20
|
A divergent Pumilio repeat protein family for pre-rRNA processing and mRNA localization. Proc Natl Acad Sci U S A 2014; 111:18554-9. [PMID: 25512524 DOI: 10.1073/pnas.1407634112] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pumilio/feminization of XX and XO animals (fem)-3 mRNA-binding factor (PUF) proteins bind sequence specifically to mRNA targets using a single-stranded RNA-binding domain comprising eight Pumilio (PUM) repeats. PUM repeats have now been identified in proteins that function in pre-rRNA processing, including human Puf-A and yeast Puf6. This is a role not previously ascribed to PUF proteins. Here we present crystal structures of human Puf-A that reveal a class of nucleic acid-binding proteins with 11 PUM repeats arranged in an "L"-like shape. In contrast to classical PUF proteins, Puf-A forms sequence-independent interactions with DNA or RNA, mediated by conserved basic residues. We demonstrate that equivalent basic residues in yeast Puf6 are important for RNA binding, pre-rRNA processing, and mRNA localization. Thus, PUM repeats can be assembled into alternative folds that bind to structured nucleic acids in addition to forming canonical eight-repeat crescent-shaped RNA-binding domains found in classical PUF proteins.
Collapse
|
21
|
Oostvogels R, Lokhorst HM, Minnema MC, van Elk M, van den Oudenalder K, Spierings E, Mutis T, Spaapen RM. Identification of minor histocompatibility antigens based on the 1000 Genomes Project. Haematologica 2014; 99:1854-9. [PMID: 25150256 DOI: 10.3324/haematol.2014.109801] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Minor histocompatibility antigens are highly immunogeneic polymorphic peptides playing crucial roles in the clinical outcome of HLA-identical allogeneic stem cell transplantation. Although the introduction of genome-wide association-based strategies significantly has accelerated the identification of minor histocompatibility antigens over the past years, more efficient, rapid and robust identification techniques are required for a better understanding of the immunobiology of minor histocompatibility antigens and for their optimal clinical application in the treatment of hematologic malignancies. To develop a strategy that can overcome the drawbacks of all earlier strategies, we now integrated our previously developed genetic correlation analysis methodology with the comprehensive genomic databases from the 1000 Genomes Project. We show that the data set of the 1000 Genomes Project is suitable to identify all of the previously known minor histocompatibility antigens. Moreover, we demonstrate the power of this novel approach by the identification of the new HLA-DP4 restricted minor histocompatibility antigen UTDP4-1, which despite extensive efforts could not be identified using any of the previously developed biochemical, molecular biological or genetic strategies. The 1000 Genomes Project-based identification of minor histocompatibility antigens thus represents a very convenient and robust method for the identification of new targets for cancer therapy after allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Rimke Oostvogels
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht; Department of Hematology, University Medical Center Utrecht, Utrecht
| | - Henk M Lokhorst
- Department of Hematology, University Medical Center Utrecht, Utrecht; Department of Hematology, VU University Medical Center, Amsterdam
| | - Monique C Minnema
- Department of Hematology, University Medical Center Utrecht, Utrecht
| | - Maureen van Elk
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht
| | | | - Eric Spierings
- Department of Immunology, University Medical Center Utrecht, Utrecht
| | - Tuna Mutis
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht; Department of Hematology, VU University Medical Center, Amsterdam;
| | - Robbert M Spaapen
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht; Department of Immunopathology, Sanquin Research, Amsterdam; Department of Cell Biology II, The Netherlands Cancer Institute, Amsterdam; Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, the Netherlands
| |
Collapse
|
22
|
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: 154] [Impact Index Per Article: 14.0] [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
|
23
|
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
|
24
|
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
|
25
|
Fan CC, Lee LY, Yu MY, Tzen CY, Chou C, Chang MS. Upregulated hPuf-A promotes breast cancer tumorigenesis. Tumour Biol 2013; 34:2557-64. [PMID: 23625657 DOI: 10.1007/s13277-013-0801-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 04/09/2013] [Indexed: 12/11/2022] Open
Abstract
hPuf-A is a member of RNA-binding PUF family that regulates mRNA translation. Redistribution of hPuf-A from the nucleolus to the nucleoplasm upon genotoxic stress modulates the poly(ADP-ribosyl)ation activity of PARP-1. Here, we report a novel function of hPuf-A involved in promoting breast cancer progression. Immunohistochemical studies showed higher expression levels of hPuf-A in stage I, II, III, and IV breast cancer specimens in contrast with those of hPuf-A in ductal carcinoma in situ. The presence of hPuf-A is highly associated with colony formation capacities in breast cancer T47D and MDA-MB-231 cells. Xenograft growth of hPuf-A-silenced and hPuf-A overexpressing MDA-MB-231 cells in nude mice was substantially in concert with colony formation capacities. This promoting effect of hPuf-A in tumorigenesis might be correlated with the regulation of its associated mRNAs, such as RbAp48 and DDX3. Collectively, hPuf-A may have diagnostic values in breast cancer progression.
Collapse
Affiliation(s)
- Chi-Chen Fan
- Department of Physiology, Mackay Memorial Hospital, Taipei, Taiwan,
| | | | | | | | | | | |
Collapse
|
26
|
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
|
27
|
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
|
28
|
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
|
29
|
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
|
30
|
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
|
31
|
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
|
32
|
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
|
33
|
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
|
34
|
Tykodi SS, Sandmaier BM, Warren EH, Thompson JA. Allogeneic hematopoietic cell transplantation for renal cell carcinoma: ten years after. Expert Opin Biol Ther 2011; 11:763-73. [PMID: 21417772 DOI: 10.1517/14712598.2011.566855] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The first series of patients with metastatic renal cell carcinoma (RCC) treated by non-myeloablative allogeneic hematopoietic cell transplantation (HCT) was reported in 2000 and demonstrated an allogeneic graft-versus-tumor (GVT) effect that encouraged further investigation of this approach. However, the past 10 years have also witnessed profound changes in the medical management of metastatic RCC with the introduction of targeted therapies directed against VEGF or mammalian target of rapamycin (mTOR) signaling pathways creating uncertainty about a continued role for allogeneic HCT in the treatment of RCC. AREAS COVERED A total of 21 published reports describing clinical outcomes for 398 patients with metastatic RCC treated by allogeneic HCT compiled herein provide a composite overview of the world wide experience for key efficacy and toxicity outcomes. Review of correlative studies that identify donor-derived T cells as mediators of RCC-specific GVT effects offers insight into both the potential as well as the technical barriers to the delivery of antigen-specific post-transplant cellular therapy or vaccination designed to augment the allogeneic GVT effect. EXPERT OPINION The future development of non-myeloablative allogeneic HCT for metastatic RCC will require novel treatment protocols designed to augment and sustain post-transplant GVT effects against RCC to generate renewed enthusiasm for this approach.
Collapse
Affiliation(s)
- Scott S Tykodi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | | | | | |
Collapse
|
35
|
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: 93] [Impact Index Per Article: 6.6] [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
|
36
|
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
|
37
|
Chang HY, Fan CC, Chu PC, Hong BE, Lee HJ, Chang MS. hPuf-A/KIAA0020 modulates PARP-1 cleavage upon genotoxic stress. Cancer Res 2011; 71:1126-34. [PMID: 21266351 DOI: 10.1158/0008-5472.can-10-1831] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Human hPuf-A/KIAA0020 was first identified as a new minor histocompatibility antigen in 2001. Its zebrafish orthologue contains six Pumilio-homology RNA-binding domains and has been shown to participate in the development of eyes and primordial germ cells, but the cellular function of hPuf-A remains unclear. In this report, we showed that hPuf-A predominantly localized in the nucleoli with minor punctate signals in the nucleoplasm. The nucleolar localization of hPuf-A would redistribute to the nucleoplasm after the treatment of RNA polymerase inhibitors (actinomycin D and 5,6-dichlorobenzimidazole riboside) and topoisomerase inhibitors [camptothecin (CPT) and etoposide]. Interestingly, knockdown of hPuf-A sensitized cells to CPT and UV treatment and cells constitutively overexpressing hPuf-A became more resistant to genotoxic exposure. Affinity gel pull-down coupled with mass spectrometric analysis identified PARP-1 as one of the hPuf-A interacting proteins. hPuf-A specifically interacts with the catalytic domain of PARP-1 and inhibits poly(ADP-ribosyl)ation of PARP-1 in vitro. Depletion of hPuf-A increased the cleaved PARP-1 and overexpression of hPuf-A lessened PARP-1 cleavage when cells were exposed to CPT and UV light. Collectively, hPuf-A may regulate cellular response to genotoxic stress by inhibiting PARP-1 activity and thus preventing PARP-1 degradation by caspase-3.
Collapse
Affiliation(s)
- Hao-Yen Chang
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
38
|
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]
|
39
|
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
|
40
|
Park MJ, Choi HB, Kim TG. Improved genotyping of the human minor histocompatibility antigen HB-1 by polymerase chain reaction with sequence-specific primers using a complementary oligonucleotide. ACTA ACUST UNITED AC 2010; 76:482-6. [PMID: 20718936 DOI: 10.1111/j.1399-0039.2010.01546.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Single nucleotide polymorphisms of minor histocompatibility antigens (mHags) have been genotyped by allele-specific polymerase chain reaction with sequence-specific primers (PCR-SSP). Because discriminating the genotype of HB-1 Y by PCR-SSP under various PCR conditions was difficult, we optimized the use of oligonucleotides complementary to the allele-specific forward primer to improve the specificity of the HB-1 Y PCR-SSP. Specific allele discrimination was possible with an annealing temperature between 61°C and 63°C and in the presence of a threefold excess of a 15-bp complementary oligonucleotide. In conclusion, the inclusion of a complementary oligonucleotide in the PCR-SSP assay may improve its specificity and selectivity for genotyping several mHags for which optimizing PCR conditions have been difficult.
Collapse
Affiliation(s)
- M-J Park
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seocho-Gu, Seoul, Korea
| | | | | |
Collapse
|
41
|
Alyea EP, DeAngelo DJ, Moldrem J, Pagel JM, Przepiorka D, Sadelin M, Young JW, Giralt S, Bishop M, Riddell S. NCI First International Workshop on The Biology, Prevention and Treatment of Relapse after Allogeneic Hematopoietic Cell Transplantation: report from the committee on prevention of relapse following allogeneic cell transplantation for hematologic malignancies. Biol Blood Marrow Transplant 2010; 16:1037-69. [PMID: 20580849 PMCID: PMC3235046 DOI: 10.1016/j.bbmt.2010.05.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2010] [Accepted: 05/14/2010] [Indexed: 10/19/2022]
Abstract
Prevention of relapse after allogeneic hematopoietic stem cell transplantation is the most likely approach to improve survival of patients treated for hematologic malignancies. Herein we review the limits of currently available transplant therapies and the innovative strategies being developed to overcome resistance to therapy or to fill therapeutic modalities not currently available. These novel strategies include nonimmunologic therapies, such as targeted preparative regimens and posttransplant drug therapy, as well as immunologic interventions, including graft engineering, donor lymphocyte infusions, T cell engineering, vaccination, and dendritic cell-based approaches. Several aspects of the biology of the malignant cells as well as the host have been identified that obviate success of even these newer strategies. To maximize the potential for success, we recommend pursuing research to develop additional targeted therapies to be used in the preparative regimen or as maintenance posttransplant, better characterize the T cell and dendritic cells subsets involved in graft-versus-host disease and the graft-versus-leukemia/tumor effect, identify strategies for timing immunologic or nonimmunologic therapies to eliminate the noncycling cancer stem cell, identify more targets for immunotherapies, develop new vaccines that will not be limited by HLA, and develop methods to identify populations at very high risk for relapse to accelerate clinical development and avoid toxicity in patients not at risk for relapse.
Collapse
Affiliation(s)
- Edwin P Alyea
- Dana Farber Cancer Institute, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ofran Y, Kim HT, Brusic V, Blake L, Mandrell M, Wu CJ, Sarantopoulos S, Bellucci R, Keskin DB, Soiffer RJ, Antin JH, Ritz J. Diverse patterns of T-cell response against multiple newly identified human Y chromosome-encoded minor histocompatibility epitopes. Clin Cancer Res 2010; 16:1642-51. [PMID: 20160060 DOI: 10.1158/1078-0432.ccr-09-2701] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Donor T cells respond to minor histocompatibility antigens (mHA), resulting in both graft-versus-host disease and graft versus leukemia after allogeneic hematopoietic stem cell transplantation. Because relatively few mHAs are known, we developed a new approach to predict and subsequently validate candidate mHA. EXPERIMENTAL DESIGN We developed an algorithm based on genetic disparities between Y chromosome-encoded and X chromosome-encoded proteins and known requirements for binding to HLA class I molecules to predict Y chromosome-derived, HLA A*0201-restricted peptides (HY) and ranked peptides based on potential immunogenicity. We evaluated T-cell responses to 41 candidate peptides in 28 male recipients with female donors (FM), 22 male recipients with male donors (MM), and 26 normal individuals. All patients and donors were HLA A*0201 positive. RESULTS Thirteen peptides derived from five proteins elicited significantly greater T-cell responses in FM patients compared with MM patients and in normal females compared with normal males. Six peptides were more immunogenic than the only previously known HLA A*0201-restricted Y-encoded mHA. Twenty-seven of 28 FM patients responded to at least one HY peptide, but despite a common Y chromosome mismatch and expression of HLA A*0201, each patient responded to a unique set of peptides. CONCLUSIONS Novel HLA A*0201-restricted HY epitopes can be predicted and validated in patients after allogeneic hematopoietic stem cell transplantation. Highly diverse patterns of T-cell response against these epitopes have been identified. Prospective monitoring of responses to large panels of immunogenic peptides can facilitate the identification of clinically relevant targets of graft-versus-host disease and graft versus leukemia.
Collapse
Affiliation(s)
- Yishai Ofran
- Division of Hematologic Malignancies, Cancer Vaccine Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Identification of 4 new HLA-DR–restricted minor histocompatibility antigens as hematopoietic targets in antitumor immunity. Blood 2009; 114:3684-92. [DOI: 10.1182/blood-2009-03-208017] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Potent graft-versus-leukemia (GVL) effects can be mediated by donor-derived T cells recognizing minor histocompatibility antigens (mHags) in patients treated with donor lymphocyte infusion (DLI) for relapsed hematologic malignancies after HLA-matched allogeneic stem cell transplantation (alloSCT). Donor-derived T cells, however, may not only induce GVL, but also mediate detrimental graft-versus-host disease (GVHD). Because HLA-class II is under noninflammatory conditions predominantly expressed on hematopoietic cells, CD4+ T cells administered late after alloSCT may selectively confer GVL without GVHD. Although a broad range of different HLA-class I–restricted mHags have been identified, the first 2 autosomal HLA-class II–restricted mHags have only recently been characterized. By screening a recombinant bacteria cDNA expression library, we identified 4 new HLA-class II–restricted mHags recognized by CD4+ T cells induced in a patient with relapsed chronic myeloid leukemia who achieved long-term complete remission and experienced only mild GVHD of the skin after DLI. All CD4+ T cells were capable of recognizing the mHags presented by HLA-DR surface molecules on primary hematopoietic cells, but not on skin-derived (cytokine-treated) fibroblasts. The selective recognition of hematopoietic cells as well as the balanced population frequencies and common HLA-DR restriction elements make the novel mHags possible targets for development of immunotherapeutic strategies.
Collapse
|
44
|
Laurin D, Hannani D, Pernollet M, Moine A, Plumas J, Bensa JC, Cahn JY, Garban F. Immunomonitoring of graft-versus-host minor histocompatibility antigen correlates with graft-versus-host disease and absence of relapse after graft. Transfusion 2009; 50:418-28. [PMID: 19843288 DOI: 10.1111/j.1537-2995.2009.02440.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND After HLA-identical hematopoietic stem cell transplantation, minor histocompatibility (mH) antigen alloreactivity plays a dominant role in the development of graft-versus-host disease (GVHD) and graft versus leukemia (GVL). STUDY DESIGN AND METHODS We have analyzed the mH alloreactivity (enzyme-linked immunospot [ELISpot] for interferon-gamma[IFN-gamma] assay) from 24 donor/recipient pairs over a period of 2 years of follow-up and correlated such alloreactivity with the development of GVHD or absence of relapse. Circulating specific T cells anti-mH with multimer HLA-peptides were also studied. RESULTS We show by ELISpot IFN-gamma assay that alloreactivity during the first 3 months from donor versus recipient or donor versus mismatched identified mH antigens is associated with acute GVHD and GVL effect. In addition, we demonstrate that the donor-versus-recipient reactivity observed after the third month is highly associated with chronic GVHD and GVL (p = 0.0007). Finally, we show by multimer HLA-peptide assay that mH epitope-specific T cells present after 3 months are statistically related to the GVL effect. CONCLUSIONS Our results provide a robust method to monitor mH antigen graft-versus-host reaction and suggest that current identified mH have predictive value on GVHD and GVL.
Collapse
Affiliation(s)
- David Laurin
- Etablissement Français du Sang Rhône-Alpes, and Immunobiologie et Immunothérapie des Cancers, Centre de Recherche INSERM Albert Bonniot, La Tronche, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Spellman S, Warden MB, Haagenson M, Pietz BC, Goulmy E, Warren EH, Wang T, Ellis TM. Effects of mismatching for minor histocompatibility antigens on clinical outcomes in HLA-matched, unrelated hematopoietic stem cell transplants. Biol Blood Marrow Transplant 2009; 15:856-63. [PMID: 19539218 DOI: 10.1016/j.bbmt.2009.03.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Accepted: 03/20/2009] [Indexed: 11/30/2022]
Abstract
Several studies in HLA-matched sibling hematopoietic stem cell transplantation (HSCT) have reported an association between mismatches in minor histocompatibility antigens (mHAg) and outcomes. We assessed whether single and multiple minor mHAg mismatches are associated with outcomes in 730 unrelated donor, HLA-A, B, C, DRB1, and DQB1 allele-matched hematopoietic stem cell transplants (HSCT) facilitated by the National Marrow Donor Program (NMDP) between 1996 and 2003. Patients had acute and chronic leukemia or myelodysplastic syndrome (MDS), received myeloablative conditioning regimens and calcineurin inhibitor-based graft-versus-host-disease (GVHD) prophylaxis, and most received bone marrow (BM; 85%). Donor and recipient DNA samples were genotyped for mHAg including: HA-1, HA-2, HA-3, HA-8, HB-1 and CD31(125/563). Primary outcomes included grades III-IV acute GVHD (aGVHD) and survival; secondary outcomes included chronic GVHD (cGVHD), engraftment, and relapse. Single disparities at HA-1, HA-2, HA-3, HA-8, and HB-1 were not significantly associated with any of the outcomes analyzed. In HLA-A2-positive individuals, single CD31(563) or multiple mHAg mismatches in the HVG vector were associated with lower risk of grades III-IV aGVHD. Based on these data, we conclude that mHAg incompatibility at HA-1, HA-2, HA-3, HA-8, HB-1, and CD31 has no detectable effect on the outcome of HLA matched unrelated donor HSCT.
Collapse
|
46
|
Fanning SL, Appel MY, Berger SA, Korngold R, Friedman TM. The immunological impact of genetic drift in the B10.BR congenic inbred mouse strain. THE JOURNAL OF IMMUNOLOGY 2009; 183:4261-72. [PMID: 19752227 DOI: 10.4049/jimmunol.0900971] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The MHC-matched, minor histocompatibility Ag (miHA)-mismatched B10.BR-->CBA strain combination has been used to elucidate the immunobiology of graft-vs-host disease (GVHD) following allogeneic bone marrow transplantation. Studies conducted in the 1980s had established that B10.BR CD8+ T cells were capable of mediating GVHD in the absence of CD4+ T cells, and that CD4+ T cells were unable to induce lethal disease. In more recent studies with this GVHD model, we detected etiological discrepancies with the previously published results, which suggested that genetic drift might have occurred within the B10.BR strain. In particular, there was increased allorecognition of CBA miHA by B10.BR CD4+ T cells, as determined by both TCR Vbeta spectratype analysis and the induction of lethal GVHD in CBA recipients. Additionally, alloreactivity was observed between the genetically drifted mice (B10.BR/Jdrif) and mice rederived from frozen embryos of the original strain (B10.BR/Jrep) using Vbeta spectratype analysis and IFN-gamma ELISPOT assays, suggesting that new miHA differences had arisen between the mice. Furthermore, T cell-depleted B10.BR/Jdrif bone marrow cells were unable to provide long-term survival following either allogeneic or syngeneic bone marrow transplantation. Gene expression analysis revealed several genes involved in hematopoiesis that were overexpressed in the lineage-negative fraction of B10.BR/Jdrif bone marrow, as compared with B10.BR/Jrep mice. Taken together, these results suggest that genetic drift in the B10.BR strain has significantly impacted the immune alloreactive response in the GVHD model by causing altered expression of miHA and diminished capacity for survival following transplantation into lethally irradiated recipients.
Collapse
Affiliation(s)
- Stacey L Fanning
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | | | | | | | | |
Collapse
|
47
|
Ringdén O, Karlsson H, Olsson R, Omazic B, Uhlin M. The allogeneic graft-versus-cancer effect. Br J Haematol 2009; 147:614-33. [PMID: 19735262 DOI: 10.1111/j.1365-2141.2009.07886.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Allogeneic haematological stem cell transplantation (HSCT) has developed into immunotherapy. Donor CD4+, CD8+ and natural killer (NK) cells have been reported to mediate graft-versus-leukaemia (GVL) effects, using Fas-dependent killing and perforin degranulation to eradicate malignant cells. Cytokines, such as interleukin-2, interferon-gamma and tumour necrosis factor-alpha potentiate the GVL effect. Post-transplant adoptive therapy of cytotoxic T-cells (CTL) against leukaemia-specific antigens, minor histocompatibility antigens, or T-cell receptor genes may constitute successful approaches to induce anti-tumour effects. Clinically, a significant GVL effect is induced by chronic rather than acute graft-versus-host disease (GVHD). An anti-tumour effect has also been reported for myeloma, lymphoma and solid tumours. Reduced intensity conditioning enables HSCT in older and disabled patients and relies on the graft-versus-tumour effect. Donor lymphocyte infusions promote the GVL effect and can be given as escalating doses with response monitored by minimal residual disease. A high CD34+ cell dose of peripheral blood stem cells increases GVL. There is a balance between effective immunosuppression, low incidence of GVHD and relapse. For instance, T-cell depletion of the graft increases the risk of relapse. This paper reviews the current knowledge in graft-versus-cancer effects. Future directions, such as immunotherapy using leukaemia-specific CTLs, allo-depleted T-cells and suicide gene manipulated T-cells, are presented.
Collapse
Affiliation(s)
- Olle Ringdén
- Centre for Allogeneic Stem Cell Transplantation and Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
48
|
Neumann F, Sturm C, Hülsmeyer M, Dauth N, Guillaume P, Luescher IF, Pfreundschuh M, Held G. Fab antibodies capable of blocking T cells by competitive binding have the identical specificity but a higher affinity to the MHC-peptide-complex than the T cell receptor. Immunol Lett 2009; 125:86-92. [DOI: 10.1016/j.imlet.2009.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 05/16/2009] [Accepted: 06/03/2009] [Indexed: 10/20/2022]
|
49
|
Abstract
The curative potential of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for many hematologic malignancies derives in large part from reconstitution of normal donor immunity and the development of a potent graft-versus-leukemia (GVL) immune response capable of rejecting tumor cell in vivo. Elucidation of the mechanisms of GVL by studies of animal models and analysis of clinical data has yielded important insights into how clinically effective tumor immunity is generated following allo-HSCT. These studies have identified NK cells and B cells as well as T cells as important mediators of the GVL response. A variety of antigenic targets of the GVL response have also been identified, and include tumor-associated antigens as well as minor histocompatibility antigens. The principles of effective GVL can now be applied to the development of novel therapies that enhance the therapeutic benefit of allogeneic HSCT while minimizing the toxicities associated with treatment. Moreover, many components of this approach that result in elimination of tumor cells following allogeneic HSCT can potentially be adapted to enhance the effectiveness of tumor immunity in the autologous setting.
Collapse
Affiliation(s)
- Catherine J Wu
- Cancer Vaccine Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | |
Collapse
|
50
|
Spierings E, Gras S, Reiser JB, Mommaas B, Almekinders M, Kester MGD, Chouquet A, Le Gorrec M, Drijfhout JW, Ossendorp F, Housset D, Goulmy E. Steric Hindrance and Fast Dissociation Explain the Lack of Immunogenicity of the Minor Histocompatibility HA-1Arg Null Allele. THE JOURNAL OF IMMUNOLOGY 2009; 182:4809-16. [DOI: 10.4049/jimmunol.0803911] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|