1
|
Cedrone E, Ishaq A, Grabarnik E, Edmondson E, Skoczen S, Neun BW, Freer M, Shuttleworth S, Sviland L, Dickinson A, Dobrovolskaia MA. In vitro assessment of nanomedicines' propensity to cause palmar-plantar erythrodysesthesia: A Doxil vs. doxorubicin case study. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 62:102780. [PMID: 39181221 PMCID: PMC11513236 DOI: 10.1016/j.nano.2024.102780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024]
Abstract
Palmar-plantar erythrodysesthesia (PPE), also known as hand and foot syndrome, is a condition characterized by inflammation-mediated damage to the skin on the palms and soles of the hands and feet. PPE limits the successful therapeutic applications of anticancer drugs. However, identifying this toxicity during preclinical studies is challenging due to the lack of accurate in vitro and in vivo animal-based models. Therefore, there is a need for reliable models that would allow the detection of this toxicity early during the drug development process. Herein, we describe the use of an in vitro skin explant assay to assess traditional DXR, Doxil reference listed drug (RLD) and two generic PEGylated liposomal DXR formulations for their abilities to cause inflammation and skin damage. We demonstrate that the results obtained with the in vitro skin explant assay model for traditional DXR and Doxil correlate with the clinical data.
Collapse
Affiliation(s)
- Edward Cedrone
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, 8560 Progress Drive, Frederick, MD 21701, USA
| | - Abbas Ishaq
- Alcyomics Ltd., Biosphere Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, United Kingdom
| | - Emma Grabarnik
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, 8560 Progress Drive, Frederick, MD 21701, USA
| | - Elijah Edmondson
- Molecular Histopathology Laboratory, Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, 8560 Progress Drive, Frederick, MD 21701, USA
| | - Sarah Skoczen
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, 8560 Progress Drive, Frederick, MD 21701, USA
| | - Barry W Neun
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, 8560 Progress Drive, Frederick, MD 21701, USA
| | - Matthew Freer
- Alcyomics Ltd., Biosphere Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, United Kingdom
| | - Siannah Shuttleworth
- Alcyomics Ltd., Biosphere Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, United Kingdom
| | - Lisbet Sviland
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Norway
| | - Anne Dickinson
- Alcyomics Ltd., Biosphere Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, United Kingdom.
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, 8560 Progress Drive, Frederick, MD 21701, USA.
| |
Collapse
|
2
|
Anzai T, Myatt GJ, Hall F, Finney B, Nakagawa K, Iwata H, Anzai R, Dickinson A, Freer M, Nakae D, Onodera H, Matsuyama T. Drug review process advancement and required manufacturer and contract research oraganization responses. J Toxicol Pathol 2024; 37:45-53. [PMID: 38584971 PMCID: PMC10995435 DOI: 10.1293/tox.2023-0106] [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: 09/08/2023] [Accepted: 11/09/2023] [Indexed: 04/09/2024] Open
Abstract
The United States Senate passed the "FDA Modernization Act 2.0." on September 29, 2022. Although the effectiveness of this Bill, which aims to eliminate the mandatory use of laboratory animals in new drug development, is limited, it represents a significant trend that will change the shape of drug applications in the United States and other countries. However, pharmaceutical companies have not taken major steps towards the complete elimination of animal testing from the standpoint of product safety, where they prioritize patient safety. Nonetheless, society is becoming increasingly opposed to animal testing, and efforts will be made to use fewer animals and conduct fewer animal tests as a natural and reasonable response. These changes eventually alter the shape of new drug applications. Based on the assumption that fewer animal tests will be conducted or fewer animals will be used in testing, this study explored bioinformatics and new technologies as alternatives to compensate for reduced information and provide a picture of how future new drug applications may look. The authors also discuss the directions that pharmaceutical companies and nonclinical contract research organizations should adopt to promote the replacement, reduction, and refinement of animals used in research, teaching, testing, and exhibitions.
Collapse
Affiliation(s)
- Takayuki Anzai
- Showa University School of Medicine, 1-5-8 Hatanodai,
Shinagawa, Tokyo 142-0064, Japan
| | - Glenn J. Myatt
- Instem, Diamond Way, Stone Business Park, Stone,
Staffordshire, ST150SD, United Kingdom
| | - Frances Hall
- Instem, Diamond Way, Stone Business Park, Stone,
Staffordshire, ST150SD, United Kingdom
| | - Brenda Finney
- Instem, Diamond Way, Stone Business Park, Stone,
Staffordshire, ST150SD, United Kingdom
| | - Kenshi Nakagawa
- Showa University School of Medicine, 1-5-8 Hatanodai,
Shinagawa, Tokyo 142-0064, Japan
| | - Hijiri Iwata
- LunaPath Laboratory of Toxicologic Pathology, 3-5-1
Aoihigashi, Naka-ku Hamamatsu-shi, Shizuoka 433-8114, Japan
| | - Reo Anzai
- Tokyo University of the Arts, 5000 Omonma, Toride-shi,
Ibaraki 302-0001, Japan
| | - Anne Dickinson
- Newcastle University, Newcastle upon Tyne, NE1 7RU, United
Kingdom
- Alcyomics, The Biosphere, Draymans Way, Newcastle upon Tyne
NE4 5BX, United Kingdom
| | - Matthew Freer
- Alcyomics, The Biosphere, Draymans Way, Newcastle upon Tyne
NE4 5BX, United Kingdom
| | - Dai Nakae
- Teikyo Heisei University, 4-1 Uruido-Minami, Ichihara-shi,
Chiba 290-0193, Japan
| | - Hiroshi Onodera
- National Institute of Health Sciences, 3-25-26 Tono-machi,
Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Takaaki Matsuyama
- Showa University School of Medicine, 1-5-8 Hatanodai,
Shinagawa, Tokyo 142-0064, Japan
| |
Collapse
|
3
|
Maurer K, Antin JH. The graft versus leukemia effect: donor lymphocyte infusions and cellular therapy. Front Immunol 2024; 15:1328858. [PMID: 38558819 PMCID: PMC10978651 DOI: 10.3389/fimmu.2024.1328858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for many hematologic malignancies as well as non-malignant conditions. Part of the curative basis underlying HSCT for hematologic malignancies relies upon induction of the graft versus leukemia (GVL) effect in which donor immune cells recognize and eliminate residual malignant cells within the recipient, thereby maintaining remission. GVL is a clinically evident phenomenon; however, specific cell types responsible for inducing this effect and molecular mechanisms involved remain largely undefined. One of the best examples of GVL is observed after donor lymphocyte infusions (DLI), an established therapy for relapsed disease or incipient/anticipated relapse. DLI involves infusion of peripheral blood lymphocytes from the original HSCT donor into the recipient. Sustained remission can be observed in 20-80% of patients treated with DLI depending upon the underlying disease and the intrinsic burden of targeted cells. In this review, we will discuss current knowledge about mechanisms of GVL after DLI, experimental strategies for augmenting GVL by manipulation of DLI (e.g. neoantigen vaccination, specific cell type selection/depletion) and research outlook for improving DLI and cellular immunotherapies for hematologic malignancies through better molecular definition of the GVL effect.
Collapse
Affiliation(s)
| | - Joseph H. Antin
- Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Teshima T, Hashimoto D. Separation of GVL from GVHD -location, location, location. Front Immunol 2023; 14:1296663. [PMID: 38116007 PMCID: PMC10728488 DOI: 10.3389/fimmu.2023.1296663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is a curative therapy for various hematologic malignancies. However, alloimmune response is a double-edged sword that mediates both beneficial graft-versus-leukemia (GVL) effects and harmful graft-versus-host disease (GVHD). Separation of GVL effects from GVHD has been a topic of intense research to improve transplant outcomes, but reliable clinical strategies have not yet been established. Target tissues of acute GVHD are the skin, liver, and intestine, while leukemic stem cells reside in the bone marrow. Tissue specific effector T-cell migration is determined by a combination of inflammatory and chemotactic signals that interact with specific receptors on T cells. Specific inhibition of donor T cell migration to GVHD target tissues while preserving migration to the bone marrow may represent a novel strategy to separate GVL from GVHD. Furthermore, tissue specific GVHD therapy, promoting tissue tolerance, and targeting of the tumor immune microenvironment may also help to separate GVHD and GVL.
Collapse
Affiliation(s)
- Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | | |
Collapse
|
5
|
Jaing TH, Chang TY, Chiu CC. Harnessing and honing mesenchymal stem/stromal cells for the amelioration of graft-versus-host disease. World J Stem Cells 2023; 15:221-234. [PMID: 37180998 PMCID: PMC10173808 DOI: 10.4252/wjsc.v15.i4.221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/19/2023] [Accepted: 03/21/2023] [Indexed: 04/26/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation is a deterministic curative procedure for various hematologic disorders and congenital immunodeficiency. Despite its increased use, the mortality rate for patients undergoing this procedure remains high, mainly due to the perceived risk of exacerbating graft-versus-host disease (GVHD). However, even with immunosuppressive agents, some patients still develop GVHD. Advanced mesenchymal stem/stromal cell (MSC) strategies have been proposed to achieve better therapeutic outcomes, given their immunosuppressive potential. However, the efficacy and trial designs have varied among the studies, and some research findings appear contradictory due to the challenges in characterizing the in vivo effects of MSCs. This review aims to provide real insights into this clinical entity, emphasizing diagnostic, and therapeutic considerations and generating pathophysiology hypotheses to identify research avenues. The indications and timing for the clinical application of MSCs are still subject to debate.
Collapse
Affiliation(s)
- Tang-Her Jaing
- Division of Hematology, Oncology, Department of Pediatrics, Chang Gung Children’s Hospital, Chang Gung University, Taoyuan 333, Taiwan
| | - Tsung-Yen Chang
- Department of Pediatrics, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Chia-Chi Chiu
- Department of Nursing, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| |
Collapse
|
6
|
Ash S, Askenasy N. Immunotherapy for neuroblastoma by hematopoietic cell transplantation and post-transplant immunomodulation. Crit Rev Oncol Hematol 2023; 185:103956. [PMID: 36893946 DOI: 10.1016/j.critrevonc.2023.103956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/14/2022] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroblastoma represents a relatively common childhood tumor that imposes therapeutic difficulties. High risk neuroblastoma patients have poor prognosis, display limited response to radiochemotherapy and may be treated by hematopoietic cell transplantation. Allogeneic and haploidentical transplants have the distinct advantage of reinstitution of immune surveillance, reinforced by antigenic barriers. The key factors favorable to ignition of potent anti-tumor reactions are transition to adaptive immunity, recovery from lymphopenia and removal of inhibitory signals that inactivate immune cells at the local and systemic levels. Post-transplant immunomodulation may further foster anti-tumor reactivity, with positive but transient impact of infusions of lymphocytes and natural killer cells both from the donor, the recipient or third party. The most promising approaches include introduction of antigen-presenting cells in early post-transplant stages and neutralization of inhibitory signals. Further studies will likely shed light on the nature and actions of suppressor factors within tumor stroma and at the systemic level.
Collapse
Affiliation(s)
- Shifra Ash
- Department of Pediatric Hematology-Oncology, Rambam Medical Center, Haifa, Israel; Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Nadir Askenasy
- Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
7
|
Diaz-Valencia JD, Estrada-Abreo LA, Rodríguez-Cruz L, Salgado-Aguayo AR, Patiño-López G. Class I Myosins, molecular motors involved in cell migration and cancer. Cell Adh Migr 2022; 16:1-12. [PMID: 34974807 PMCID: PMC8741282 DOI: 10.1080/19336918.2021.2020705] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 01/13/2023] Open
Abstract
Class I Myosins are a subfamily of motor proteins with ATPase activity and a characteristic structure conserved in all myosins: A N-Terminal Motor Domain, a central Neck and a C terminal Tail domain. Humans have eight genes for these myosins. Class I Myosins have different functions: regulate membrane tension, participate in endocytosis, exocytosis, intracellular trafficking and cell migration. Cell migration is influenced by many cellular components including motor proteins, like myosins. Recently has been reported that changes in myosin expression have an impact on the migration of cancer cells, the formation of infiltrates and metastasis. We propose that class I myosins might be potential markers for future diagnostic, prognostic or even as therapeutic targets in leukemia and other cancers.Abbreviations: Myo1g: Myosin 1g; ALL: Acute Lymphoblastic Leukemia, TH1: Tail Homology 1; TH2: Tail Homology 2; TH3: Tail Homology 3.
Collapse
Affiliation(s)
- Juan D. Diaz-Valencia
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| | - Laura A. Estrada-Abreo
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Leonor Rodríguez-Cruz
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Alfonso R. Salgado-Aguayo
- Rheumatic Diseases Laboratory, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Genaro Patiño-López
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| |
Collapse
|
8
|
Kay AG, Fox JM, Hewitson JP, Stone AP, Robertson S, James S, Wang XN, Kapasa E, Yang XB, Genever PG. CD317-Positive Immune Stromal Cells in Human "Mesenchymal Stem Cell" Populations. Front Immunol 2022; 13:903796. [PMID: 35734183 PMCID: PMC9207511 DOI: 10.3389/fimmu.2022.903796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/04/2022] [Indexed: 12/31/2022] Open
Abstract
Heterogeneity of bone marrow mesenchymal stromal cells (MSCs, frequently referred to as "mesenchymal stem cells") clouds biological understanding and hampers their clinical development. In MSC cultures most commonly used in research and therapy, we have identified an MSC subtype characterized by CD317 expression (CD317pos (29.77 ± 3.00% of the total MSC population), comprising CD317dim (28.10 ± 4.60%) and CD317bright (1.67 ± 0.58%) MSCs) and a constitutive interferon signature linked to human disease. We demonstrate that CD317pos MSCs induced cutaneous tissue damage when applied a skin explant model of inflammation, whereas CD317neg MSCs had no effect. Only CD317neg MSCs were able to suppress proliferative cycles of activated human T cells in vitro, whilst CD317pos MSCs increased polarization towards pro-inflammatory Th1 cells and CD317neg cell lines did not. Using an in vivo peritonitis model, we found that CD317neg and CD317pos MSCs suppressed leukocyte recruitment but only CD317neg MSCs suppressed macrophage numbers. Using MSC-loaded scaffolds implanted subcutaneously in immunocompromised mice we were able to observe tissue generation and blood vessel formation with CD317neg MSC lines, but not CD317pos MSC lines. Our evidence is consistent with the identification of an immune stromal cell, which is likely to contribute to specific physiological and pathological functions and influence clinical outcome of therapeutic MSCs.
Collapse
Affiliation(s)
- Alasdair G. Kay
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom,*Correspondence: Paul G. Genever, ; Alasdair G. Kay,
| | - James M. Fox
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - James P. Hewitson
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Andrew P. Stone
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Sophie Robertson
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Sally James
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Xiao-nong Wang
- Translational and Clinical Research Institute, Newcastle University, Newcastle, United Kingdom
| | - Elizabeth Kapasa
- Department of Oral Biology, School of Dentistry, University of Leeds, St James’s University Hospital, Leeds, United Kingdom
| | - Xuebin B. Yang
- Department of Oral Biology, School of Dentistry, University of Leeds, St James’s University Hospital, Leeds, United Kingdom
| | - Paul G. Genever
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom,*Correspondence: Paul G. Genever, ; Alasdair G. Kay,
| |
Collapse
|
9
|
Pawar S, Kutay U. The Diverse Cellular Functions of Inner Nuclear Membrane Proteins. Cold Spring Harb Perspect Biol 2021; 13:a040477. [PMID: 33753404 PMCID: PMC8411953 DOI: 10.1101/cshperspect.a040477] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The nuclear compartment is delimited by a specialized expanded sheet of the endoplasmic reticulum (ER) known as the nuclear envelope (NE). Compared to the outer nuclear membrane and the contiguous peripheral ER, the inner nuclear membrane (INM) houses a unique set of transmembrane proteins that serve a staggering range of functions. Many of these functions reflect the exceptional position of INM proteins at the membrane-chromatin interface. Recent research revealed that numerous INM proteins perform crucial roles in chromatin organization, regulation of gene expression, genome stability, and mediation of signaling pathways into the nucleus. Other INM proteins establish mechanical links between chromatin and the cytoskeleton, help NE remodeling, or contribute to the surveillance of NE integrity and homeostasis. As INM proteins continue to gain prominence, we review these advancements and give an overview on the functional versatility of the INM proteome.
Collapse
Affiliation(s)
- Sumit Pawar
- Institute of Biochemistry, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Ulrike Kutay
- Institute of Biochemistry, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
10
|
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
|
11
|
Jardine L, Cytlak U, Gunawan M, Reynolds G, Green K, Wang XN, Pagan S, Paramitha M, Lamb CA, Long AK, Hurst E, Nair S, Jackson GH, Publicover A, Bigley V, Haniffa M, Simpson AJ, Collin M. Donor monocyte-derived macrophages promote human acute graft-versus-host disease. J Clin Invest 2021; 130:4574-4586. [PMID: 32453711 PMCID: PMC7456218 DOI: 10.1172/jci133909] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 05/19/2020] [Indexed: 12/16/2022] Open
Abstract
Myelopoiesis is invariably present and contributes to pathology in animal models of graft-versus-host disease (GVHD). In humans, a rich inflammatory infiltrate bearing macrophage markers has also been described in histological studies. In order to determine the origin, functional properties, and role in pathogenesis of these cells, we isolated single-cell suspensions from acute cutaneous GVHD and subjected them to genotype, transcriptome, and in vitro functional analysis. A donor-derived population of CD11c+CD14+ cells was the dominant population of all leukocytes in GVHD. Surface phenotype and NanoString gene expression profiling indicated the closest steady-state counterpart of these cells to be monocyte-derived macrophages. In GVHD, however, there was upregulation of monocyte antigens SIRPα and S100A8/9 transcripts associated with leukocyte trafficking, pattern recognition, antigen presentation, and costimulation. Isolated GVHD macrophages stimulated greater proliferation and activation of allogeneic T cells and secreted higher levels of inflammatory cytokines than their steady-state counterparts. In HLA-matched mixed leukocyte reactions, we also observed differentiation of activated macrophages with a similar phenotype. These exhibited cytopathicity to a keratinocyte cell line and mediated pathological damage to skin explants independently of T cells. Together, these results define the origin, functional properties, and potential pathogenic roles of human GVHD macrophages.
Collapse
Affiliation(s)
- Laura Jardine
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Urszula Cytlak
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Merry Gunawan
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Gary Reynolds
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Kile Green
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Sarah Pagan
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Maharani Paramitha
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher A Lamb
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Anna K Long
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Erin Hurst
- Northern Centre for Bone Marrow Transplantation and
| | - Smeera Nair
- Northern Centre for Bone Marrow Transplantation and
| | - Graham H Jackson
- Northern Centre for Bone Marrow Transplantation and.,Northern Institute of Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Amy Publicover
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Venetia Bigley
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Muzlifah Haniffa
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - A J Simpson
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Matthew Collin
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
12
|
Poels R, Drent E, Lameris R, Katsarou A, Themeli M, van der Vliet HJ, de Gruijl TD, van de Donk NWCJ, Mutis T. Preclinical Evaluation of Invariant Natural Killer T Cells Modified with CD38 or BCMA Chimeric Antigen Receptors for Multiple Myeloma. Int J Mol Sci 2021; 22:1096. [PMID: 33499253 PMCID: PMC7865760 DOI: 10.3390/ijms22031096] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 12/16/2022] Open
Abstract
Due to the CD1d restricted recognition of altered glycolipids, Vα24-invariant natural killer T (iNKT) cells are excellent tools for cancer immunotherapy with a significantly reduced risk for graft-versus-host disease when applied as off-the shelf-therapeutics across Human Leukocyte Antigen (HLA) barriers. To maximally harness their therapeutic potential for multiple myeloma (MM) treatment, we here armed iNKT cells with chimeric antigen receptors (CAR) directed against the MM-associated antigen CD38 and the plasma cell specific B cell maturation antigen (BCMA). We demonstrate that both CD38- and BCMA-CAR iNKT cells effectively eliminated MM cells in a CAR-dependent manner, without losing their T cell receptor (TCR)-mediated cytotoxic activity. Importantly, iNKT cells expressing either BCMA-CARs or affinity-optimized CD38-CARs spared normal hematopoietic cells and displayed a Th1-like cytokine profile, indicating their therapeutic utility. While the costimulatory domain of CD38-CARs had no influence on the cytotoxic functions of iNKT cells, CARs containing the 4-1BB domain showed a better expansion capacity. Interestingly, when stimulated only via CD1d+ dendritic cells (DCs) loaded with α-galactosylceramide (α-GalCer), both CD38- and BCMA-CAR iNKT cells expanded well, without losing their CAR- or TCR-dependent cytotoxic activities. This suggests the possibility of developing an off-the-shelf therapy with CAR iNKT cells, which might even be boostable in vivo by administration α-GalCer pulsed DCs.
Collapse
Affiliation(s)
- Renée Poels
- Cancer Center Amsterdam, Department of Haematology, Amsterdam UMC, VU Amsterdam, 1081 HV Amsterdam, The Netherlands; (R.P.); (E.D.); (A.K.); (M.T.); (N.W.C.J.v.d.D.)
| | - Esther Drent
- Cancer Center Amsterdam, Department of Haematology, Amsterdam UMC, VU Amsterdam, 1081 HV Amsterdam, The Netherlands; (R.P.); (E.D.); (A.K.); (M.T.); (N.W.C.J.v.d.D.)
| | - Roeland Lameris
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, VU Amsterdam, 1081 HV Amsterdam, The Netherlands; (R.L.); (H.J.v.d.V.); (T.D.d.G.)
| | - Afroditi Katsarou
- Cancer Center Amsterdam, Department of Haematology, Amsterdam UMC, VU Amsterdam, 1081 HV Amsterdam, The Netherlands; (R.P.); (E.D.); (A.K.); (M.T.); (N.W.C.J.v.d.D.)
| | - Maria Themeli
- Cancer Center Amsterdam, Department of Haematology, Amsterdam UMC, VU Amsterdam, 1081 HV Amsterdam, The Netherlands; (R.P.); (E.D.); (A.K.); (M.T.); (N.W.C.J.v.d.D.)
| | - Hans J. van der Vliet
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, VU Amsterdam, 1081 HV Amsterdam, The Netherlands; (R.L.); (H.J.v.d.V.); (T.D.d.G.)
- Lava Therapeutics, 3584 CM Utrecht, The Netherlands
| | - Tanja D. de Gruijl
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, VU Amsterdam, 1081 HV Amsterdam, The Netherlands; (R.L.); (H.J.v.d.V.); (T.D.d.G.)
| | - Niels W. C. J. van de Donk
- Cancer Center Amsterdam, Department of Haematology, Amsterdam UMC, VU Amsterdam, 1081 HV Amsterdam, The Netherlands; (R.P.); (E.D.); (A.K.); (M.T.); (N.W.C.J.v.d.D.)
| | - Tuna Mutis
- Cancer Center Amsterdam, Department of Haematology, Amsterdam UMC, VU Amsterdam, 1081 HV Amsterdam, The Netherlands; (R.P.); (E.D.); (A.K.); (M.T.); (N.W.C.J.v.d.D.)
| |
Collapse
|
13
|
Mondino A, Manzo T. To Remember or to Forget: The Role of Good and Bad Memories in Adoptive T Cell Therapy for Tumors. Front Immunol 2020; 11:1915. [PMID: 32973794 PMCID: PMC7481451 DOI: 10.3389/fimmu.2020.01915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
The generation of immunological memory is a hallmark of adaptive immunity by which the immune system "remembers" a previous encounter with an antigen expressed by pathogens, tumors, or normal tissues; and, upon secondary encounters, mounts faster and more effective recall responses. The establishment of T cell memory is influenced by both cell-intrinsic and cell-extrinsic factors, including genetic, epigenetic and environmental triggers. Our current knowledge of the mechanisms involved in memory T cell differentiation has instructed new opportunities to engineer T cells with enhanced anti-tumor activity. The development of adoptive T cell therapy has emerged as a powerful approach to cure a subset of patients with advanced cancers. Efficacy of this approach often requires long-term persistence of transferred T cell products, which can vary according to their origin and manufacturing conditions. Host preconditioning and post-transfer supporting strategies have shown to promote their engraftment and survival by limiting the competition with a hostile tumor microenvironment and between pre-existing immune cell subsets. Although in the general view pre-existing memory can confer a selective advantage to adoptive T cell therapy, here we propose that also "bad memories"-in the form of antigen-experienced T cell subsets-co-evolve with consequences on newly transferred lymphocytes. In this review, we will first provide an overview of selected features of memory T cell subsets and, then, discuss their putative implications for adoptive T cell therapy.
Collapse
Affiliation(s)
- Anna Mondino
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Teresa Manzo
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milan, Italy
| |
Collapse
|
14
|
Denholtz M, Zhu Y, He Z, Lu H, Isoda T, Döhrmann S, Nizet V, Murre C. Upon microbial challenge, human neutrophils undergo rapid changes in nuclear architecture and chromatin folding to orchestrate an immediate inflammatory gene program. Genes Dev 2020; 34:149-165. [PMID: 31919189 PMCID: PMC7000913 DOI: 10.1101/gad.333708.119] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/12/2019] [Indexed: 01/03/2023]
Abstract
Differentiating neutrophils undergo large-scale changes in nuclear morphology. How such alterations in structure are established and modulated upon exposure to microbial agents is largely unknown. Here, we found that prior to encounter with bacteria, an armamentarium of inflammatory genes was positioned in a transcriptionally passive environment suppressing premature transcriptional activation. Upon microbial exposure, however, human neutrophils rapidly (<3 h) repositioned the ensemble of proinflammatory genes toward the transcriptionally permissive compartment. We show that the repositioning of genes was closely associated with the swift recruitment of cohesin across the inflammatory enhancer landscape, permitting an immediate transcriptional response upon bacterial exposure. We found that activated enhancers, marked by increased deposition of H3K27Ac, were highly enriched for cistromic elements associated with PU.1, CEBPB, TFE3, JUN, and FOSL2 occupancy. These data reveal how upon microbial challenge the cohesin machinery is recruited to an activated enhancer repertoire to instruct changes in chromatin folding, nuclear architecture, and to activate an inflammatory gene program.
Collapse
Affiliation(s)
- Matthew Denholtz
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92039, USA
| | - Yina Zhu
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92039, USA
| | - Zhaoren He
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92039, USA
| | - Hanbin Lu
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92039, USA
| | - Takeshi Isoda
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92039, USA
| | - Simon Döhrmann
- Department of Pediatrics, University of California at San Diego School of Medicine, La Jolla, California 92093, USA
| | - Victor Nizet
- Department of Pediatrics, University of California at San Diego School of Medicine, La Jolla, California 92093, USA
- Skaggs School of Pharmaceutical Sciences, University of California at San Diego, La Jolla, California 92093, USA
| | - Cornelis Murre
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92039, USA
| |
Collapse
|
15
|
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
|
16
|
Arshad R, Sohail MF, Sarwar HS, Saeed H, Ali I, Akhtar S, Hussain SZ, Afzal I, Jahan S, Anees-ur-Rehman, Shahnaz G. ZnO-NPs embedded biodegradable thiolated bandage for postoperative surgical site infection: In vitro and in vivo evaluation. PLoS One 2019; 14:e0217079. [PMID: 31170179 PMCID: PMC6553718 DOI: 10.1371/journal.pone.0217079] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/05/2019] [Indexed: 11/19/2022] Open
Abstract
Post-operative surgical site infections (SSI) present a serious threat and may lead to complications. Currently available dressings for SSI lack mucoadhesion, safety, efficacy and most importantly patient compliance. We aimed to address these concerns by developing a bioactive thiolated chitosan-alginate bandage embedded with zinc oxide nanoparticles (ZnO-NPs) for localized topical treatment of SSI. The FTIR, XRD, DSC and TGA of bandage confirmed the compatibility of ingredients and modifications made. The porosity, swelling index and lysozyme degradation showed good properties for wound healing and biodegradation. Moreover, in-vitro antibacterial activity showed higher bactericidal effect as compared to ZnO-NPs free bandage. In-vivo wound healing in murine model showed significant improved tissue generation and speedy wound healing as compared to positive and negative controls. Over all, thiolated bandage showed potential as an advanced therapeutic agent for treating surgical site infections, meeting the required features of an ideal surgical dressing.
Collapse
Affiliation(s)
- Rabia Arshad
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Muhammad Farhan Sohail
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Riphah Institute of Pharmaceutical Sciences (RIPS), Riphah International University, Lahore Campus, Lahore, Pakistan
| | - Hafiz Shoaib Sarwar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Riphah Institute of Pharmaceutical Sciences (RIPS), Riphah International University, Lahore Campus, Lahore, Pakistan
| | - Hamid Saeed
- University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Imran Ali
- Department of Entomology, University College of Agriculture & Environmental Sciences, The Islamia University, Bahawalpur, Pakistan
| | | | - Syed Zajif Hussain
- Department of Chemistry and Chemical Engineering, SBA School of Science and Engineering (SBA-SSE), Lahore University of Management Sciences (LUMS), Lahore, Pakistan
| | - Iqra Afzal
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Sarwat Jahan
- Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Anees-ur-Rehman
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- School of Pharmaceutical Sciences, Universiti Sains Malaysia (USM), Pulau Pinang, Malaysia
| | - Gul Shahnaz
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
17
|
Ahmed SS, Whritenour J, Ahmed MM, Bibby L, Darby L, Wang XN, Watson J, Dickinson AM. Evaluation of a human in vitro skin test for predicting drug hypersensitivity reactions. Toxicol Appl Pharmacol 2019; 369:39-48. [PMID: 30768973 DOI: 10.1016/j.taap.2019.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 02/04/2019] [Accepted: 02/11/2019] [Indexed: 11/16/2022]
Abstract
The occurrence of drug hypersensitivity reactions (DHRs) following administration of low molecular weight (LMW) drugs is an important health concern. However, in vivo animal models which could be used as tools for the prediction of DHRs are lacking. As a result, research has focused on development of in vitro tools for predicting DHRs. In this study a novel human in vitro pre-clinical skin explant test was used to predict T cell-mediated hypersensitivity responses induced by LMW drugs. Responses in the skin explant test for 12 LMW drugs associated with T cell-mediated hypersensitivity in the clinic (abacavir, amoxicillin, carbamazepine, diclofenac, lamotrigine, lapatinib, lumiracoxib, nevirapine, ofloxacin, phenytoin, propranolol, sulfamethoxazole) were compared with responses for 5 drugs with few/no reports of T cell-mediated hypersensitivity reactions (acetaminophen, cimetidine, flecainide, metformin, verapamil). Changes in skin histology following in vitro exposure to the drugs as well as T cell proliferation and interferon gamma (IFNγ) production were studied. The results of the skin explant assays showed a good positive correlation (r = 0.77, p < .001) between the test outcome (prediction of positive or negative) and the clinical classification of the tested drugs. The T cell proliferation assay showed a correlation of r = 0.60 (p < .01) and the IFNγ assay r = 0.51 (p < .04). The data suggest that the skin explant model could be a useful tool to predict the potential of LMW drugs to induce DHRs.
Collapse
Affiliation(s)
- S S Ahmed
- Alcyomics Ltd, Bulman House, Regent Centre, Gosforth, Newcastle-upon-Tyne NE3 3LS, United Kingdom
| | - J Whritenour
- Pfizer Inc., Drug Safety Research and Development, Eastern Point Rd, Groton, CT 06340, USA
| | - M M Ahmed
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - L Bibby
- Alcyomics Ltd, Bulman House, Regent Centre, Gosforth, Newcastle-upon-Tyne NE3 3LS, United Kingdom; Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - L Darby
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - X N Wang
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - J Watson
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne NE2 4HH, United Kingdom
| | - A M Dickinson
- Alcyomics Ltd, Bulman House, Regent Centre, Gosforth, Newcastle-upon-Tyne NE3 3LS, United Kingdom; Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne NE2 4HH, United Kingdom.
| |
Collapse
|
18
|
van Grinsven E, Textor J, Hustin LSP, Wolf K, Koenderman L, Vrisekoop N. Immature Neutrophils Released in Acute Inflammation Exhibit Efficient Migration despite Incomplete Segmentation of the Nucleus. THE JOURNAL OF IMMUNOLOGY 2018; 202:207-217. [PMID: 30504419 DOI: 10.4049/jimmunol.1801255] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/28/2018] [Indexed: 11/19/2022]
Abstract
Acute inflammation recruits neutrophils with a band-shaped nucleus to the circulation. This neutrophil population was recently shown to have superior antibacterial capacity. Early recruitment of banded neutrophils to an infection site will likely improve the outcome of the immune response, yet it critically depends on efficient migration. However, the current dogma states that the segmentation of the mature neutrophil nucleus has evolved to favor migration through narrow pores as found between endothelial cells and in the interstitium. Therefore, we hypothesized that banded neutrophils migrate less efficiently than neutrophils with segmented nuclei, whereas recently described neutrophils with hypersegmented nuclei would in turn migrate more efficiently. Acute inflammation was evoked in a human model of experimental endotoxemia to recruit neutrophil subsets with different nuclear segmentation to the circulation. To simulate migration toward an infection site, migration of the subsets was studied in in vitro models of transendothelial migration or interstitial chemokinesis and chemotaxis. In both models, nuclear segmentation did not increase migration speed. In dense collagen matrices, the speed of the hypersegmented neutrophils was even reduced compared with the banded neutrophils. Fluorescence microscopy suggested that the hypersegmented neutrophils displayed reduced rear release and deposited more membrane vesicles. Vice versa, migration through narrow pores did not induce nuclear segmentation in the neutrophils. In conclusion, like neutrophils with a segmented nucleus, the banded subset exhibited efficient migration through narrow pores. These findings suggest that the nucleus does not preclude the banded subset from reaching an infection site.
Collapse
Affiliation(s)
- Erinke van Grinsven
- Department of Respiratory Medicine, University Medical Center Utrecht, 3508 AB Utrecht, the Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, 3508 AB Utrecht, the Netherlands
| | - Johannes Textor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands; and
| | - Lucie S P Hustin
- Department of Respiratory Medicine, University Medical Center Utrecht, 3508 AB Utrecht, the Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, 3508 AB Utrecht, the Netherlands
| | - Katarina Wolf
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Leo Koenderman
- Department of Respiratory Medicine, University Medical Center Utrecht, 3508 AB Utrecht, the Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, 3508 AB Utrecht, the Netherlands
| | - Nienke Vrisekoop
- Department of Respiratory Medicine, University Medical Center Utrecht, 3508 AB Utrecht, the Netherlands; .,Laboratory of Translational Immunology, University Medical Center Utrecht, 3508 AB Utrecht, the Netherlands
| |
Collapse
|
19
|
Alloantigen expression on malignant cells and healthy host tissue influences graft-versus-tumor reactions after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2018; 53:807-819. [PMID: 29362503 DOI: 10.1038/s41409-017-0071-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 11/08/2022]
Abstract
Durable remissions of hematological malignancies regularly observed following allogeneic hematopoietic stem cell transplantation (aHSCT) are due to the conditioning regimen, as well as an immunological phenomenon called graft-versus-leukemia (GVL) or graft-versus-tumor (GVT) effect. The development of GVL is closely linked to graft-versus-host disease (GVHD), the main side effect associated with aHSCT. Both, GVHD and GVL are mediated by donor T cells that are initially activated by antigen-presenting cells that present recipient-derived alloantigens in the context of either matched or mismatched MHC class I molecules. Using murine models of aHSCT we show that ubiquitously expressed minor histocompatibility alloantigens (mHAg) are no relevant target for GVT effects. Interestingly, certain ubiquitously expressed MHC alloantigens augmented GVT effects early after transplantation, while others did not. The magnitude of GVT effects correlated with tumor infiltration by CD8+ cytotoxic T cells and tumor cell apoptosis. Furthermore, the immune response underlying GVHD and GVT was oligoclonal, highlighting that immunodominance is an important factor during alloimmune responses. These results emphasize that alloantigen expression on non-hematopoietic tissues can influence GVT effects in a previously unrecognized fashion. These findings bear significance for harnessing optimal GVL effects in patients receiving aHSCT.
Collapse
|
20
|
Reddy P, Ferrara JL. Graft-Versus-Host Disease and Graft-Versus-Leukemia Responses. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
21
|
Maciocia PM, Wawrzyniecka PA, Philip B, Ricciardelli I, Akarca AU, Onuoha SC, Legut M, Cole DK, Sewell AK, Gritti G, Somja J, Piris MA, Peggs KS, Linch DC, Marafioti T, Pule MA. Targeting the T cell receptor β-chain constant region for immunotherapy of T cell malignancies. Nat Med 2017; 23:1416-1423. [PMID: 29131157 DOI: 10.1038/nm.4444] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/18/2017] [Indexed: 12/19/2022]
Abstract
Mature T cell cancers are typically aggressive, treatment resistant and associated with poor prognosis. Clinical application of immunotherapeutic approaches has been limited by a lack of target antigens that discriminate malignant from healthy (normal) T cells. Unlike B cell depletion, pan-T cell aplasia is prohibitively toxic. We report a new targeting strategy based on the mutually exclusive expression of T cell receptor β-chain constant domains 1 and 2 (TRBC1 and TRBC2). We identify an antibody with unique TRBC1 specificity and use it to demonstrate that normal and virus-specific T cell populations contain both TRBC1+ and TRBC2+ compartments, whereas malignancies are restricted to only one. As proof of concept for anti-TRBC immunotherapy, we developed anti-TRBC1 chimeric antigen receptor (CAR) T cells, which recognized and killed normal and malignant TRBC1+, but not TRBC2+, T cells in vitro and in a disseminated mouse model of leukemia. Unlike nonselective approaches targeting the entire T cell population, TRBC-targeted immunotherapy could eradicate a T cell malignancy while preserving sufficient normal T cells to maintain cellular immunity.
Collapse
Affiliation(s)
| | | | - Brian Philip
- Cancer Institute, University College London, London, UK
| | - Ida Ricciardelli
- Institute of Child Health, University College London, London, UK
| | - Ayse U Akarca
- Cancer Institute, University College London, London, UK
| | | | - Mateusz Legut
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - David K Cole
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Andrew K Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Giuseppe Gritti
- Hematology and Bone Marrow Transplant Units, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Joan Somja
- Department of Anatomy and Cellular Pathology, University of Liège, Liège, Belgium
| | - Miguel A Piris
- Department of Pathology, Fundación Jiménez Díaz, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Karl S Peggs
- Cancer Institute, University College London, London, UK
| | - David C Linch
- Cancer Institute, University College London, London, UK
| | | | - Martin A Pule
- Cancer Institute, University College London, London, UK.,Autolus, Ltd., London, UK
| |
Collapse
|
22
|
Development of T-cell immunotherapy for hematopoietic stem cell transplantation recipients at risk of leukemia relapse. Blood 2017; 131:108-120. [PMID: 29051183 DOI: 10.1182/blood-2017-07-791608] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/01/2017] [Indexed: 12/13/2022] Open
Abstract
Leukemia relapse remains the major cause of allogeneic hematopoietic stem cell transplantation (HCT) failure, and the prognosis for patients with post-HCT relapse is poor. There is compelling evidence that potent selective antileukemic effects can be delivered by donor T cells specific for particular minor histocompatibility (H) antigens. Thus, T-cell receptors (TCRs) isolated from minor H antigen-specific T cells represent an untapped resource for developing targeted T-cell immunotherapy to manage post-HCT leukemic relapse. Recognizing that several elements may be crucial to the efficacy and safety of engineered T-cell immunotherapy, we developed a therapeutic transgene with 4 components: (1) a TCR specific for the hematopoietic-restricted, leukemia-associated minor H antigen, HA-1; (2) a CD8 coreceptor to promote function of the class I-restricted TCR in CD4+ T cells; (3) an inducible caspase 9 safety switch to enable elimination of the HA-1 TCR T cells in case of toxicity; and (4) a CD34-CD20 epitope to facilitate selection of the engineered cell product and tracking of transferred HA-1 TCR T cells. The T-cell product includes HA-1 TCR CD4+ T cells to augment the persistence and function of the HA-1 TCR CD8+ T cells and includes only memory T cells; naive T cells are excluded to limit the potential for alloreactivity mediated by native TCR coexpressed by HA-1 TCR T cells. We describe the development of this unique immunotherapy and demonstrate functional responses to primary leukemia by CD4+ and CD8+ T cells transduced with a lentiviral vector incorporating the HA-1 TCR transgene construct.
Collapse
|
23
|
Baygan A, Aronsson-Kurttila W, Moretti G, Tibert B, Dahllöf G, Klingspor L, Gustafsson B, Khoein B, Moll G, Hausmann C, Svahn BM, Westgren M, Remberger M, Sadeghi B, Ringden O. Safety and Side Effects of Using Placenta-Derived Decidual Stromal Cells for Graft-versus-Host Disease and Hemorrhagic Cystitis. Front Immunol 2017; 8:795. [PMID: 28744284 PMCID: PMC5504152 DOI: 10.3389/fimmu.2017.00795] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/22/2017] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are increasingly used in regenerate medicine. Placenta-derived decidual stromal cells (DSCs) are a novel therapy for acute graft-versus-host-disease (GVHD) and hemorrhagic cystitis (HC) after allogeneic hematopoietic stem cell transplantation (HSCT). DSCs are more immunosuppressive than MSCs. We assessed adverse events and safety using DSCs among 44 treated patients and 40 controls. The median dose of infused cells was 1.5 (range 0.9–2.9) × 106 DSCs/kg. The patients were given 2 (1–5) doses, with a total of 82 infusions. Monitoring ended 3 months after the last DSC infusion. Three patients had transient reactions during DSC infusion. Laboratory values, hemorrhages, and transfusions were similar in the two groups. The frequency of leukemic relapse (2/2, DSC/controls) and invasive fungal infections (6/6) were the same in the two groups. Causes of death were those seen in HSCT patients: infections (5/3), respiratory failure (1/1), circulatory failure (3/1), thromboembolism (1/0), multiorgan failure (0/1), and GVHD and others (2/7). One-year survival for the DSC patients with GVHD was 67%, which was significantly better than achieved previously at our center. One-year survival was 90% in the DSC-treated HC group. DSC infusions appear safe. Randomized studies are required to prove efficacy.
Collapse
Affiliation(s)
- Arjang Baygan
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Wictor Aronsson-Kurttila
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Gianluca Moretti
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Babylonia Tibert
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Göran Dahllöf
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lena Klingspor
- Department of Microbiology, Uppsala University Hospital, Uppsala, Sweden
| | - Britt Gustafsson
- Department of Pediatrics, Uppsala University Hospital, Uppsala, Sweden
| | - Bita Khoein
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Guido Moll
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden.,Charité Universitätsmedizin, Berlin, Germany
| | - Charlotta Hausmann
- Center for Allogeneic Stem Cell Transplantation, Department of Pathology/Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Britt-Marie Svahn
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Westgren
- Department of Obstetrics and Gynecology, Karolinska University Hospital, Stockholm, Sweden
| | - Mats Remberger
- Center for Allogeneic Stem Cell Transplantation, Department of Pathology/Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Behnam Sadeghi
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Olle Ringden
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Cytomegalovirus-Specific T Cells Isolated by IFN-γ Secretion Assay Do Not Induce Significant Graft-Versus-Host Reactions In Vitro. Transplantation 2017; 100:2352-2361. [PMID: 27152919 DOI: 10.1097/tp.0000000000001219] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Graft-versus-host (GvH) disease (GvHD) remains a serious concern for patients undergoing antiviral cellular therapy. Despite the major improvements in cellular immunotherapy, the immunogenicity of virus-specific T cells has not yet been fully defined. This present study aims to examine how cytomegalovirus (CMV)-specific cytotoxic T lymphocytes (CTLs) respond to allogeneic antigen stimulation and whether they give rise to GvHD target tissue damage. METHODS Cytomegalovirus-specific CTLs were isolated by the IFN-γ secretion assay (gamma-catch) from healthy seropositive volunteers and expanded in vitro. The levels of intracellular IFN-γ, cytotoxic activity, IFN-γ and granzyme B secretion, and CD25 expression were measured using flow cytometry (fluorescence-activated cell sorting). The ability of CMV-CTLs to induce GvHD target tissue damage was evaluated using the human in vitro skin explant assay (skin explant assay). RESULTS Cytomegalovirus-specific CTLs responded specifically to CMV-phosphoprotein 65 stimulation by secreting IFN-γ and killing virus peptide loaded autologous phytohemagglutinin (PHA) blasts. Compared with unselected peripheral blood mononuclear cells, CMV-CTLs induced significantly less severe cutaneous GvH tissue damage. This observation coincided with low levels of CD25 expression, as well as IFN-γ and granzyme B secretion after allogeneic antigen stimulation in both the mixed lymphocyte reaction and in the skin explant assay. CONCLUSIONS Cytomegalovirus-specific CTLs isolated by the IFN-γ secretion assay from HLA-unmatched healthy donors exhibited a high level of anti-CMV potency without inducing significant cutaneous GvH tissue damage in vitro. This finding provides novel evidence supporting the safe use of in vitro expanded CMV-CTLs as an antiviral therapy in transplant patients with refractory CMV infections.
Collapse
|
25
|
Barton BM, Xu R, Wherry EJ, Porrett PM. Pregnancy promotes tolerance to future offspring by programming selective dysfunction in long-lived maternal T cells. J Leukoc Biol 2017; 101:975-987. [PMID: 27810945 DOI: 10.1189/jlb.1a0316-135r] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 08/31/2016] [Accepted: 10/11/2016] [Indexed: 11/24/2022] Open
Abstract
Fetal antigen available during pregnancy induces the proliferation of maternal T cells. It is unknown, however, whether these antigen-activated T cells differentiate into long-lived memory T cells that are capable of mediating rapid-recall responses to tissue antigens. To test the hypothesis that pregnancy induces an alternative fate in fetal-specific maternal T cells, we used a murine model to track longitudinally fetal-specific T cells in pregnant and postpartum animals and test the response of these cells when challenged with the same antigen during sequential pregnancy or skin transplantation. Fetal-specific CD8+ T cells were robustly primed during pregnancy but failed to acquire robust effector functions. These primed cells persisted long term in postpartum animals, frequently maintained a programmed death 1 (PD-1)+ phenotype, and failed to expand or produce cytokines robustly in response to second pregnancy or skin transplantation. However, whereas there was no impact on second pregnancy as a result of the persistence of fetal-primed memory CD8+ T cells in the mother, skin grafts bearing the same antigen were rejected more rapidly. Altogether, our data suggest that fetal antigen exposure during pregnancy induces the differentiation of long-lived maternal CD8+ T cells with context-dependent, selective effector dysfunction. This programmed effector dysfunction provides temporal and systemic restraint of maternal anti-fetal alloreactivity to promote reproductive fitness efficiently, while preserving potentially protective effector T cell responses.
Collapse
Affiliation(s)
- Brendan M Barton
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Rong Xu
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; and
| | - E John Wherry
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paige M Porrett
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; and
| |
Collapse
|
26
|
Staffas A, Burgos da Silva M, van den Brink MRM. The intestinal microbiota in allogeneic hematopoietic cell transplant and graft-versus-host disease. Blood 2017; 129:927-933. [PMID: 27940475 PMCID: PMC5324712 DOI: 10.1182/blood-2016-09-691394] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 12/05/2016] [Indexed: 12/14/2022] Open
Abstract
Hematopoietic cell transplantation (HCT) is a critical treatment of patients with high-risk hematopoietic malignancies, hematological deficiencies, and other immune diseases. In allogeneic HCT (allo-HCT), donor-derived T cells recognize host tissues as foreign, causing graft-versus-host disease (GVHD) which is a main contributor to morbidity and mortality. The intestine is one of the organs most severely affected by GVHD and research has recently highlighted the importance of bacteria, particularly the gut microbiota, in HCT outcome and in GVHD development. Loss of intestinal bacterial diversity is common during the course of HCT and is associated with GVHD development and treatment with broad-spectrum antibiotics. Loss of intestinal diversity and outgrowth of opportunistic pathogens belonging to the phylum Proteobacteria and Enterococcus genus have also been linked to increased treatment-related mortality including GVHD, infections, and organ failure after allo-HCT. Experimental studies in allo-HCT animal models have shown some promising results for prebiotic and probiotic strategies as prophylaxis or treatment of GVHD. Continuous research will be important to define the relation of cause and effect for these associations between microbiota features and HCT outcomes. Importantly, studies focused on geographic and cultural differences in intestinal microbiota are necessary to define applicability of new strategies targeting the intestinal microbiota.
Collapse
Affiliation(s)
- Anna Staffas
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marina Burgos da Silva
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marcel R M van den Brink
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine and
- Department of Immunology, Weill Medical College of Cornell University, New York, NY; and
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
27
|
Sugita S, Iwasaki Y, Makabe K, Kamao H, Mandai M, Shiina T, Ogasawara K, Hirami Y, Kurimoto Y, Takahashi M. Successful Transplantation of Retinal Pigment Epithelial Cells from MHC Homozygote iPSCs in MHC-Matched Models. Stem Cell Reports 2016; 7:635-648. [PMID: 27641649 PMCID: PMC5063629 DOI: 10.1016/j.stemcr.2016.08.010] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/14/2016] [Accepted: 08/16/2016] [Indexed: 01/12/2023] Open
Abstract
There is an ongoing controversy as to whether major histocompatibility complex (MHC) matching is a solution for allogeneic stem cell transplantation. In the present study, we established retinal pigment epithelial (RPE) cells from induced pluripotent stem cells (iPSCs) in MHC homozygote donors. We observed no rejection signs in iPSC-derived RPE allografts of MHC-matched animal models without immunosuppression, whereas there were immune attacks around the graft and retinal tissue damage in MHC-mismatched models. In an immunohistochemical examination of MHC-mismatched allografts, the transplanted RPE sheets/cells were located in the subretinal space, but the RPE exhibited inflammatory and hypertrophic changes, and many inflammatory cells, e.g., Iba1+ cells, MHC class II+ cells, and CD3+ T cells, invaded the graft area. Conversely, these inflammatory cells poorly infiltrated the area around the transplanted retina if MHC-matched allografts were used. Thus, cells derived from MHC homozygous donors could be used to treat retinal diseases in histocompatible recipients.
Collapse
Affiliation(s)
- Sunao Sugita
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Yuko Iwasaki
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan; Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University Graduate School of Medicine and Dental Sciences, Tokyo 113-8519, Japan
| | - Kenichi Makabe
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Hiroyuki Kamao
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan; Department of Ophthalmology, Kawasaki Medical School, Okayama 701-0192, Japan
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Takashi Shiina
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, Kanagawa 259-1193, Japan
| | - Kazumasa Ogasawara
- Department of Pathology, Shiga University of Medical Science, Ohtsu 520-2192, Japan
| | - Yasuhiko Hirami
- Department of Ophthalmology, Institute of Biomedical Research and Innovation Hospital, Kobe 650-0047, Japan; Department of Ophthalmology, Kobe City Medical Center General Hospital, Kobe 650-0047, Japan
| | - Yasuo Kurimoto
- Department of Ophthalmology, Institute of Biomedical Research and Innovation Hospital, Kobe 650-0047, Japan; Department of Ophthalmology, Kobe City Medical Center General Hospital, Kobe 650-0047, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| |
Collapse
|
28
|
Lange J, Weil F, Riegler C, Groeber F, Rebhan S, Kurdyn S, Alb M, Kneitz H, Gelbrich G, Walles H, Mielke S. Interactions of donor sources and media influence the histo-morphological quality of full-thickness skin models. Biotechnol J 2016; 11:1352-1361. [PMID: 27599760 DOI: 10.1002/biot.201600360] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/02/2016] [Accepted: 09/05/2016] [Indexed: 12/25/2022]
Abstract
Human artificial skin models are increasingly employed as non-animal test platforms for research and medical purposes. However, the overall histopathological quality of such models may vary significantly. Therefore, the effects of manufacturing protocols and donor sources on the quality of skin models built-up from fibroblasts and keratinocytes derived from juvenile foreskins is studied. Histo-morphological parameters such as epidermal thickness, number of epidermal cell layers, dermal thickness, dermo-epidermal adhesion and absence of cellular nuclei in the corneal layer are obtained and scored accordingly. In total, 144 full-thickness skin models derived from 16 different donors, built-up in triplicates using three different culture conditions were successfully generated. In univariate analysis both media and donor age affected the quality of skin models significantly. Both parameters remained statistically significant in multivariate analyses. Performing general linear model analyses we could show that individual medium-donor-interactions influence the quality. These observations suggest that the optimal choice of media may differ from donor to donor and coincides with findings where significant inter-individual variations of growth rates in keratinocytes and fibroblasts have been described. Thus, the consideration of individual medium-donor-interactions may improve the overall quality of human organ models thereby forming a reproducible test platform for sophisticated clinical research.
Collapse
Affiliation(s)
- Julia Lange
- Department of Internal Medicine II, Center for Allogeneic Stem Cell Transplantation, Würzburg University Medical Center, Würzburg, Germany
| | - Frederik Weil
- Department of Internal Medicine II, Center for Allogeneic Stem Cell Transplantation, Würzburg University Medical Center, Würzburg, Germany
| | - Christoph Riegler
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
| | - Florian Groeber
- Department of Tissue Engineering and Regenerative Medicine, Würzburg University Medical Center, Würzburg, Germany; Translational Center Würzburg, Regenerative Therapies in Oncology and Musculoskeletal Disease, Würzburg Branch of the Fraunhofer-Institute Interfacial Engineering and Biotechnology, IGB, Würzburg, Germany
| | - Silke Rebhan
- Department of Internal Medicine II, Center for Allogeneic Stem Cell Transplantation, Würzburg University Medical Center, Würzburg, Germany
| | - Szymon Kurdyn
- Department of Tissue Engineering and Regenerative Medicine, Würzburg University Medical Center, Würzburg, Germany; Translational Center Würzburg, Regenerative Therapies in Oncology and Musculoskeletal Disease, Würzburg Branch of the Fraunhofer-Institute Interfacial Engineering and Biotechnology, IGB, Würzburg, Germany
| | - Miriam Alb
- Department of Internal Medicine II, Center for Allogeneic Stem Cell Transplantation, Würzburg University Medical Center, Würzburg, Germany
| | - Hermann Kneitz
- Department of Dermatology, Section for Histopathology, Würzburg University Medical Center, Würzburg, Germany
| | - Götz Gelbrich
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
| | - Heike Walles
- Department of Tissue Engineering and Regenerative Medicine, Würzburg University Medical Center, Würzburg, Germany; Translational Center Würzburg, Regenerative Therapies in Oncology and Musculoskeletal Disease, Würzburg Branch of the Fraunhofer-Institute Interfacial Engineering and Biotechnology, IGB, Würzburg, Germany
| | - Stephan Mielke
- Department of Internal Medicine II, Center for Allogeneic Stem Cell Transplantation, Würzburg University Medical Center, Würzburg, Germany.
| |
Collapse
|
29
|
Malu K, Garhwal R, Pelletier MGH, Gotur D, Halene S, Zwerger M, Yang ZF, Rosmarin AG, Gaines P. Cooperative Activity of GABP with PU.1 or C/EBPε Regulates Lamin B Receptor Gene Expression, Implicating Their Roles in Granulocyte Nuclear Maturation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:910-22. [PMID: 27342846 PMCID: PMC5022553 DOI: 10.4049/jimmunol.1402285] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 05/30/2016] [Indexed: 01/26/2023]
Abstract
Nuclear segmentation is a hallmark feature of mammalian neutrophil differentiation, but the mechanisms that control this process are poorly understood. Gene expression in maturing neutrophils requires combinatorial actions of lineage-restricted and more widely expressed transcriptional regulators. Examples include interactions of the widely expressed ETS transcription factor, GA-binding protein (GABP), with the relatively lineage-restricted E-twenty-six (ETS) factor, PU.1, and with CCAAT enhancer binding proteins, C/EBPα and C/EBPε. Whether such cooperative interactions between these transcription factors also regulate the expression of genes encoding proteins that control nuclear segmentation is unclear. We investigated the roles of ETS and C/EBP family transcription factors in regulating the gene encoding the lamin B receptor (LBR), an inner nuclear membrane protein whose expression is required for neutrophil nuclear segmentation. Although C/EBPε was previously shown to bind the Lbr promoter, surprisingly, we found that neutrophils derived from Cebpe null mice exhibited normal Lbr gene and protein expression. Instead, GABP provided transcriptional activation through the Lbr promoter in the absence of C/EBPε, and activities supported by GABP were greatly enhanced by either C/EBPε or PU.1. Both GABP and PU.1 bound Ets sites in the Lbr promoter in vitro, and in vivo within both early myeloid progenitors and differentiating neutrophils. These findings demonstrate that GABP, PU.1, and C/EBPε cooperate to control transcription of the gene encoding LBR, a nuclear envelope protein that is required for the characteristic lobulated morphology of mature neutrophils.
Collapse
Affiliation(s)
- Krishnakumar Malu
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts, Lowell, MA 01854
| | - Rahul Garhwal
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts, Lowell, MA 01854
| | - Margery G H Pelletier
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts, Lowell, MA 01854
| | - Deepali Gotur
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts, Lowell, MA 01854
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06520
| | - Monika Zwerger
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland; and
| | - Zhong-Fa Yang
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Alan G Rosmarin
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Peter Gaines
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts, Lowell, MA 01854;
| |
Collapse
|
30
|
Lomvardas S, Maniatis T. Histone and DNA Modifications as Regulators of Neuronal Development and Function. Cold Spring Harb Perspect Biol 2016; 8:8/7/a024208. [PMID: 27371659 DOI: 10.1101/cshperspect.a024208] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
DNA and histone modifications, together with constraints imposed by nuclear architecture, contribute to the transcriptional regulatory landscape of the nervous system. Here, we provide select examples showing how these regulatory layers, often referred to as epigenetic, contribute to neuronal differentiation and function. We describe the interplay between DNA methylation and Polycomb-mediated repression during neuronal differentiation, the role of DNA methylation and long-range enhancer-promoter interactions in Protocadherin promoter choice, and the contribution of heterochromatic silencing and nuclear organization in singular olfactory receptor expression. Finally, we explain how the activity-dependent expression of a histone variant determines the longevity of olfactory sensory neurons.
Collapse
Affiliation(s)
- Stavros Lomvardas
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York 10032
| | - Tom Maniatis
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
31
|
Ahmed SS, Wang XN, Fielding M, Kerry A, Dickinson I, Munuswamy R, Kimber I, Dickinson AM. An in vitro human skin test for assessing sensitization potential. J Appl Toxicol 2016; 36:669-84. [PMID: 26251951 DOI: 10.1002/jat.3197] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/07/2015] [Accepted: 05/22/2015] [Indexed: 01/08/2023]
Abstract
Sensitization to chemicals resulting in an allergy is an important health issue. The current gold-standard method for identification and characterization of skin-sensitizing chemicals was the mouse local lymph node assay (LLNA). However, for a number of reasons there has been an increasing imperative to develop alternative approaches to hazard identification that do not require the use of animals. Here we describe a human in-vitro skin explant test for identification of sensitization hazards and the assessment of relative skin sensitizing potency. This method measures histological damage in human skin as a readout of the immune response induced by the test material. Using this approach we have measured responses to 44 chemicals including skin sensitizers, pre/pro-haptens, respiratory sensitizers, non-sensitizing chemicals (including skin-irritants) and previously misclassified compounds. Based on comparisons with the LLNA, the skin explant test gave 95% specificity, 95% sensitivity, 95% concordance with a correlation coefficient of 0.9. The same specificity and sensitivity were achieved for comparison of results with published human sensitization data with a correlation coefficient of 0.91. The test also successfully identified nickel sulphate as a human skin sensitizer, which was misclassified as negative in the LLNA. In addition, sensitizers and non-sensitizers identified as positive or negative by the skin explant test have induced high/low T cell proliferation and IFNγ production, respectively. Collectively, the data suggests the human in-vitro skin explant test could provide the basis for a novel approach for characterization of the sensitizing activity as a first step in the risk assessment process.
Collapse
Affiliation(s)
- S S Ahmed
- Alcyomics Ltd, Bulman House, Regent Centre, Gosforth, Newcastle-upon-Tyne, NE3 3LS, UK
| | - X N Wang
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - M Fielding
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - A Kerry
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - I Dickinson
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - R Munuswamy
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - I Kimber
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - A M Dickinson
- Alcyomics Ltd, Bulman House, Regent Centre, Gosforth, Newcastle-upon-Tyne, NE3 3LS, UK
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| |
Collapse
|
32
|
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
|
33
|
Leventhal J, Mathew J, Salomon D, Kurian S, Friedewald J, Gallon L, Konieczna I, Tambur A, charette J, Levitsky J, Jie C, Kanwar YS, Abecassis MM, Miller J. Nonchimeric HLA-Identical Renal Transplant Tolerance: Regulatory Immunophenotypic/Genomic Biomarkers. Am J Transplant 2016; 16:221-34. [PMID: 26227106 PMCID: PMC4718825 DOI: 10.1111/ajt.13416] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/14/2015] [Accepted: 06/02/2015] [Indexed: 01/25/2023]
Abstract
We previously described early results of a nonchimeric operational tolerance protocol in human leukocyte antigen (HLA)-identical living donor renal transplants and now update these results. Recipients given alemtuzumab, tacrolimus/MPA with early sirolimus conversion were multiply infused with donor hematopoietic CD34(+) stem cells. Immunosuppression was withdrawn by 24 months. Twelve months later, operational tolerance was confirmed by rejection-free transplant biopsies. Five of the first eight enrollees were initially tolerant 1 year off immunosuppression. Biopsies of three others after total withdrawal showed Banff 1A acute cellular rejection without renal dysfunction. With longer follow-up including 5-year posttransplant biopsies, four of the five tolerant recipients remain without rejection while one developed Banff 1A without renal dysfunction. We now add seven new subjects (two operationally tolerant), and demonstrate time-dependent increases of circulating CD4(+) CD25(+++) CD127(-) FOXP3(+) Tregs versus losses of Tregs in nontolerant subjects (p < 0.001). Gene expression signatures, developed using global RNA expression profiling of sequential whole blood and protocol biopsy samples, were highly associative with operational tolerance as early as 1 year posttransplant. The blood signature was validated by an external Immune Tolerance Network data set. Our approach to nonchimeric operational HLA-identical tolerance reveals association with Treg immunophenotypes and serial gene expression profiles.
Collapse
Affiliation(s)
- J.R. Leventhal
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A
| | - J.M. Mathew
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A,Department of Microbiology-Immunology; Northwestern University, Chicago, Illinois, U.S.A
| | - D.R. Salomon
- Department of Molecular and Experimental Medicine; The Scripps Research Institute, La Jolla, California, U.S.A
| | - S.M. Kurian
- Department of Molecular and Experimental Medicine; The Scripps Research Institute, La Jolla, California, U.S.A
| | - J.J. Friedewald
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Medicine-Nephrology; Northwestern University, Chicago, Illinois, U.S.A
| | - L. Gallon
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Medicine-Nephrology; Northwestern University, Chicago, Illinois, U.S.A
| | - I. Konieczna
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A
| | - A.R. Tambur
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A
| | - j. charette
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A
| | - J. Levitsky
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Medicine-Hepatology; Northwestern University, Chicago, Illinois, U.S.A
| | - C. Jie
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A
| | - Y. S. Kanwar
- Department of Pathology; Northwestern University, Chicago, Illinois, U.S.A
| | - M. M. Abecassis
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A,Department of Microbiology-Immunology; Northwestern University, Chicago, Illinois, U.S.A
| | - J. Miller
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A
| |
Collapse
|
34
|
Ayan MS, Abdelrahman AAM, Khanal N, Elsallabi OS, Birch NC. Case of acquired or pseudo-Pelger-Huët anomaly. Oxf Med Case Reports 2015; 2015:248-50. [PMID: 26634137 PMCID: PMC4664885 DOI: 10.1093/omcr/omv025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/08/2015] [Accepted: 02/26/2015] [Indexed: 11/12/2022] Open
Abstract
Pelger-Huët anomaly (PHA) is a rare benign autosomal-dominant anomaly with an incidence of ∼1 in 6000. It does not cause neutrophilia, but it can cause a false increase in band forms. It should be differentiated from acquired or pseudo-Pelger-Huët anomaly (PPHA), which has similar morphology, however; it is associated with different pathological states like Myelodysplastic syndrome, as well as with certain infections and drugs. We report a case of a 67-year-old Caucasian gentleman with past medical history of rheumatoid arthritis, type II diabetes mellitus and hypothyroidism, who presented with 1 day history of fever (101°F) and night sweats. Medications include ibuprofen, methotrexate, hydroxychloroquine and levothyroxine. Patient denied any other symptoms. His work-up showed normal WBC count (8.6) and increase in bands (24%). The patient was admitted for further evaluation. During the next 2 days, the patient did not have any fever or any new symptoms. Peripheral blood smear was done as part of his work-up for bandemia, showed findings suggestive of PHA. Ibuprofen was discontinued. Follow-up few weeks later showed normal blood smear. Diagnosis of PPHA was made. The presented case showed that we should think of PHA\PPHA in any case with normal total WBC count and significant shift to the lift with no apparent explanation. Looking at smears directly under the microscopes is crucial to make diagnosis.
Collapse
Affiliation(s)
- Mohamed S Ayan
- Internal Medicine Department , Creighton University Medical Center , Omaha, NE , USA
| | | | - Nabin Khanal
- Internal Medicine Department , Creighton University Medical Center , Omaha, NE , USA
| | - Osama S Elsallabi
- Internal Medicine Department , Creighton University Medical Center , Omaha, NE , USA
| | - Nathan C Birch
- Internal Medicine Department , Creighton University Medical Center , Omaha, NE , USA
| |
Collapse
|
35
|
Impact of Leukocyte Function-Associated Antigen-1 Blockade on Endogenous Allospecific T Cells to Multiple Minor Histocompatibility Antigen Mismatched Cardiac Allograft. Transplantation 2015; 99:2485-93. [DOI: 10.1097/tp.0000000000000805] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
van Halteren AGS, Dierselhuis MP, Netelenbos T, Fechter M. Donor parity no longer a barrier for female-to-male hematopoietic stem cell transplantation. CHIMERISM 2015; 5:56-8. [PMID: 24933732 DOI: 10.4161/chim.29562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a widely applied treatment for disorders mainly involving the hematopoietic system. The success of this treatment depends on many different patient- and donor-specific factors. Based on higher CD34+ yields and superior clinical outcomes associated with the use of male donors, males are generally seen as the preferred HSCT donor. In addition, female donors are notorious for bearing memory type lymphocytes induced by previous pregnancies; such alloimmune cells may provoke unwanted immune reactions such as graft-vs.-host disease in transplant recipients. Consequently, many transplant centers try to avoid parous donors, particularly when searching the best unrelated donor for a male patient. We recently showed that parous women with female offspring have an anti-male directed tolerogenic immune status comparable to that of nulliparous donors. As discussed in this article addendum, the sex of the donor's offspring combined with the presence of HY-specific T regulator cells are possibly better selection criteria than parity status per se.
Collapse
Affiliation(s)
- Astrid G S van Halteren
- Immunology Laboratory; Willem Alexander Children's Hospital/Leiden University Medical Center; Leiden, the Netherlands
| | - Miranda P Dierselhuis
- Department of Pediatrics; Willem Alexander Children's Hospital/Leiden University Medical Center; Leiden, the Netherlands
| | - Tanja Netelenbos
- Department of Immunohematology & Blood Transfusion/Leiden University Medical Center; Leiden, the Netherlands
| | | |
Collapse
|
37
|
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
|
38
|
van der Torren CR, van Hensbergen Y, Luther S, Aghai Z, Rychnavská ZS, Slot M, Scherjon S, Kröger N, Ganser A, Weissinger EM, Goulmy E, Hambach L. Possible role of minor h antigens in the persistence of donor chimerism after stem cell transplantation; relevance for sustained leukemia remission. PLoS One 2015; 10:e0119595. [PMID: 25774796 PMCID: PMC4361395 DOI: 10.1371/journal.pone.0119595] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 01/14/2015] [Indexed: 12/22/2022] Open
Abstract
Persistent complete donor chimerism is an important clinical indicator for remissions of hematological malignancies after HLA-matched allogeneic stem cell transplantation (SCT). However, the mechanisms mediating the persistence of complete donor chimerism are poorly understood. The frequent coincidence of complete donor chimerism with graft-versus-leukemia effects and graft-versus-host disease suggests that immune responses against minor histocompatibility antigens (mHags) are playing an important role in suppressing the host hematopoiesis after allogeneic SCT. Here, we investigated a possible relationship between donor immune responses against the hematopoiesis-restricted mHag HA-1 and the long-term kinetics of host hematopoietic chimerism in a cohort of 10 patients after allogeneic HLA-matched, HA-1 mismatched SCT. Functional HA-1 specific CTLs (HA-1 CTLs) were detectable in 6/10 patients lysing host-type hematopoietic cells in vitro. Presence of HA-1 CTLs in the peripheral blood coincided with low host hematopoiesis levels quantified by highly sensitive mHag specific PCR. Additionally, co-incubation of host type CD34+ cells with HA-1 CTLs isolated after allogeneic SCT prevented progenitor and cobblestone area forming cell growth in vitro and human hematopoietic engraftment in immunodeficient mice. Conversely, absence or loss of HA-1 CTLs mostly coincided with high host hematopoiesis levels and/or relapse. In summary, in this first study, presence of HA-1 CTLs paralleled low host hematopoiesis levels. This coincidence might be supported by the capacity of HA-1 CTLs isolated after allogeneic SCT to specifically eliminate host type hematopoietic stem/progenitor cells. Additional studies involving multiple mismatched mHags in more patients are required to confirm this novel characteristic of mHag CTLs as factor for the persistence of complete donor chimerism and leukemia remission after allogeneic SCT.
Collapse
Affiliation(s)
- Cornelis R. van der Torren
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Yvette van Hensbergen
- Sanquin Blood Supply Foundation, Division of Research, Department of Transfusion Medicine, Leiden, The Netherlands
| | - Susanne Luther
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Zohara Aghai
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Zuzana Stachová Rychnavská
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Manon Slot
- Sanquin Blood Supply Foundation, Division of Research, Department of Transfusion Medicine, Leiden, The Netherlands
| | - Sicco Scherjon
- Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Nicolaus Kröger
- Department of Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Eva M. Weissinger
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Els Goulmy
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Lothar Hambach
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
39
|
van Essen TH, Roelen DL, Williams KA, Jager MJ. Matching for Human Leukocyte Antigens (HLA) in corneal transplantation - to do or not to do. Prog Retin Eye Res 2015; 46:84-110. [PMID: 25601193 DOI: 10.1016/j.preteyeres.2015.01.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 01/05/2015] [Accepted: 01/07/2015] [Indexed: 12/15/2022]
Abstract
As many patients with severe corneal disease are not even considered as candidates for a human graft due to their high risk of rejection, it is essential to find ways to reduce the chance of rejection. One of the options is proper matching of the cornea donor and recipient for the Human Leukocyte Antigens (HLA), a subject of much debate. Currently, patients receiving their first corneal allograft are hardly ever matched for HLA and even patients undergoing a regraft usually do not receive an HLA-matched graft. While anterior and posterior lamellar grafts are not immune to rejection, they are usually performed in low risk, non-vascularized cases. These are the cases in which the immune privilege due to the avascular status and active immune inhibition is still intact. Once broken due to infection, sensitization or trauma, rejection will occur. There is enough data to show that when proper DNA-based typing techniques are being used, even low risk perforating corneal transplantations benefit from matching for HLA Class I, and high risk cases from HLA Class I and probably Class II matching. Combining HLA class I and class II matching, or using the HLAMatchmaker could further improve the effect of HLA matching. However, new techniques could be applied to reduce the chance of rejection. Options are the local or systemic use of biologics, or gene therapy, aiming at preventing or suppressing immune responses. The goal of all these approaches should be to prevent a first rejection, as secondary grafts are usually at higher risk of complications including rejections than first grafts.
Collapse
Affiliation(s)
- T H van Essen
- Department of Ophthalmology, J3-S, Leiden University Medical Center (LUMC), Leiden, The Netherlands.
| | - D L Roelen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - K A Williams
- Department of Ophthalmology, Flinders University, Adelaide, Australia
| | - M J Jager
- Department of Ophthalmology, J3-S, Leiden University Medical Center (LUMC), Leiden, The Netherlands; Schepens Eye Research Institute, Massachusetts Eye & Ear Infirmary and Harvard Medical School, Boston, USA; Peking University Eye Center, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
40
|
Yarkoni S, Stein J, Yaniv I, Askenasy N. Antigen-Specific Priming is Dispensable in Depletion of Apoptosis-Sensitive T Cells for GvHD Prophylaxis. Front Immunol 2014; 5:215. [PMID: 24904571 PMCID: PMC4032906 DOI: 10.3389/fimmu.2014.00215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 04/29/2014] [Indexed: 01/02/2023] Open
Abstract
Prophylactic approaches to graft versus host disease (GvHD) have employed both phenotypic reduction of T cells and selective elimination of host-primed donor T cells in vitro and in vivo. An additional approach to GvHD prophylaxis by functional depletion of apoptosis-sensitive donor T cells without host-specific sensitization ex vivo showed remarkable reduction in GHD incidence and severity. We address the role and significance of antigen-specific sensitization of donor T cells and discuss the mechanisms of functional T cell purging by apoptosis for GvHD prevention. Host-specific sensitization is dispensable because migration is antigen-independent and donor T cell sensitization is mediated by multiple and redundant mechanisms of presentation of major and minor histocompatibility complex and tissue antigens by donor and host antigen-presenting cells. Our data suggest that potential murine and human GvH effectors reside within subsets of preactivated T cells susceptible to negative regulation by apoptosis prior to encounter of and sensitization to specific antigens.
Collapse
Affiliation(s)
| | - Jerry Stein
- Bone Marrow Transplant Unit, Department of Pediatric Hematology-Oncology, Schneider Children's Medical Center of Israel , Petah Tikva , Israel
| | - Isaac Yaniv
- Bone Marrow Transplant Unit, Department of Pediatric Hematology-Oncology, Schneider Children's Medical Center of Israel , Petah Tikva , Israel
| | - Nadir Askenasy
- Frankel Laboratory, Center for Stem Cell Research, Schneider Children's Medical Center of Israel , Petah Tikva , Israel
| |
Collapse
|
41
|
van der Zouwen B, Kruisselbrink AB, Frederik Falkenburg JH, Jedema I. Collateral damage of nonhematopoietic tissue by hematopoiesis-specific T cells results in graft-versus-host disease during an ongoing profound graft-versus-leukemia reaction. Biol Blood Marrow Transplant 2014; 20:760-9. [PMID: 24607556 DOI: 10.1016/j.bbmt.2014.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 03/01/2014] [Indexed: 11/19/2022]
Abstract
After allogeneic stem cell transplantation (allo-SCT), donor T cells may recognize minor histocompatibility antigens (MiHA) specifically expressed on cells of the recipient. It has been hypothesized that T cells recognizing hematopoiesis-restricted MiHA mediate specific graft-versus-leukemia (GVL) activity without inducing graft-versus-host disease (GVHD), whereas T cells recognizing ubiquitously expressed MiHA induce both GVL and GVHD reactivity. It also has been hypothesized that alloreactive CD4 T cells are capable of mediating specific GVL reactivity due to the hematopoiesis-restricted expression of HLA class II. However, clinical observations suggest that an overt GVL response, associated with expansion of T cells specific for hematopoiesis-restricted antigens, is often associated with GVHD reactivity. Therefore, we developed in vitro models to investigate whether alloreactive T cells recognizing hematopoiesis-restricted antigens induce collateral damage to surrounding nonhematopoietic tissues. We found that collateral damage to MiHA-negative fibroblasts was induced by misdirection of cytotoxic granules released from MiHA-specific T cells activated by MiHA-positive hematopoietic cells, resulting in granzyme-B-mediated activation of apoptosis in the surrounding fibroblasts. We demonstrated that direct contact between the activated T cell and the fibroblast is a prerequisite for this collateral damage to occur. Our data suggest that hematopoiesis-restricted T cells actively participate in an overt GVL response and may contribute to GVHD via induction of collateral damage to nonhematopoietic targets.
Collapse
Affiliation(s)
- Boris van der Zouwen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Inge Jedema
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
42
|
Lashley LEELO, van der Hoorn MLP, Haasnoot GW, Roelen DL, Claas FHJ. Uncomplicated oocyte donation pregnancies are associated with a higher incidence of human leukocyte antigen alloantibodies. Hum Immunol 2014; 75:555-60. [PMID: 24530746 DOI: 10.1016/j.humimm.2014.02.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/03/2014] [Accepted: 02/04/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND Fetuses in pregnancies conceived after oocyte donation (OD) have a higher degree of antigeneic dissimilarity with the mother compared to semi-allogeneic fetuses after natural conception. We questioned whether this leads to higher level of HLA antibody formation in OD pregnancies. METHOD Uncomplicated pregnancies after OD were compared with pregnancies conceived either spontaneously or by IVF. We calculated the number of HLA- and epitope mismatches. Maternal sera were screened for HLA antibodies with ELISA; child HLA specific antibody production was determined using CDC and Luminex with single antigen beads for class I and II. RESULTS A significantly (p<0.0001) higher incidence of HLA antibody production was observed in women conceiving after OD (69%) compared to non-donor pregnancies (24-25%). The antibody formation was positively correlated with the number of fetomaternal antigen (Spearman's rho 0.95, p<0.0001) and epitope mismatches (Spearman's rho 0.91, p<0.0001). The number of HLA-DR mismatches between women and child was an independent risk factor for the production of HLA class I specific alloantibodies. CONCLUSION Women conceiving after OD have a higher risk of developing child-specific HLA antibodies; the higher the number of immunogenetic differences, the higher the chance these antibodies are formed. The high incidence of antibody production also strongly depends upon the number of HLA-DR mismatches. Despite the stronger antibody response, OD was associated with uncomplicated pregnancy in cases included in this study.
Collapse
Affiliation(s)
| | | | - Geert W Haasnoot
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Dave L Roelen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Frans H J Claas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
43
|
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
|
44
|
O'Keefe CL, Risitano AM, Maciejewski JP. Clinical Implications of T Cell Receptor Repertoire Analysis after Allogeneic Stem Cell Transplantation. Hematology 2013; 9:189-98. [PMID: 15204100 DOI: 10.1080/10245330410001701530] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Stem cell transplantation (SCT) constitutes a major challenge to the immune system. Long-term impairment of immunity against various common infectious stimuli leads to increased susceptibility to infectious diseases; in contrast, an immune response against the recipient may cause the devastating graft-versus-host disease (GvHD). Recovery of the immune system (both qualitative and quantitative) after SCT is perhaps the most important factor in determining the clinical outcome. Consequently, immune reconstitution has been extensively studied using different approaches, including quantitative analysis of immune cells as well as their phenotypic characterization. Analysis of diversity and clonality is an important tool in determining competence of the immune system, assuming that a broad diversity assures efficient response to different stimuli and clonal dominance reflects ongoing, potentially relevant immune responses. Detailed analysis of the immune repertoire through the flow cytometric and molecular study of the T cell receptor repertoire has been applied to gain quantitative and qualitative insights about the T cell immune competence and responsiveness. After SCT, a contraction of the T cell pool and a reduction in T cell receptor diversity is clearly associated with clinical immunodeficiency. Reconstitution of the immune system is often characterized by dominance of oligoclonal T cell populations, reflecting specific antigen-driven immune responses. Detailed characterization of T lymphocytes by T cell receptor analysis is possible, and may lead to the identification of individual clones involved in specific immune reactions, such as alloresponses in GvHD, the closely related graft-versus-leukemia effect and opportunistic viral agents such as CMV or EBV.
Collapse
Affiliation(s)
- Christine L O'Keefe
- Experimental Hematology and Hematopoiesis Section, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | | |
Collapse
|
45
|
Spierings E, Kim YH, Hendriks M, Borst E, Sergeant R, Canossi A, Oudshoorn M, Loiseau P, Dolstra H, Markiewicz M, Leffell MS, Pereira N, Kircher B, Turpeinen H, Eliaou JF, Gervais T, Laurin D, Enczmann J, Martinetti M, Thomson J, Oguz F, Santarone S, Partanen J, Siekiera U, Alessandrino EP, Kalayoglu S, Brand R, Goulmy E. Multicenter analyses demonstrate significant clinical effects of minor histocompatibility antigens on GvHD and GvL after HLA-matched related and unrelated hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2013; 19:1244-53. [PMID: 23756210 DOI: 10.1016/j.bbmt.2013.06.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/02/2013] [Indexed: 12/28/2022]
Abstract
The effect of minor H antigen mismatching on the occurrence of graft-versus-host disease (GvHD) and graft-versus-leukemia (GvL) after HLA-matched hematopoietic stem cell transplantation (HSCT) has mainly been demonstrated in single-center studies. Yet, the International Histocompatibility and Immunogenetics Workshops (IHIW) provide a collaborative platform to execute crucial large studies. In collaboration with 20 laboratories of the IHIW, the roles of 10 autosomal and 10 Y chromosome-encoded minor H antigens were investigated on GvHD and relapse incidence in 639 HLA-identical related donor (IRD) and 210 HLA-matched unrelated donor (MUD) HSCT recipients. Donor and recipient DNA samples were genotyped for the minor H antigens HA-1, HA-2, HA-3, HA-8, HB-1, ACC-1, ACC-2, SP110, PANE1, UGT2B17, and HY. The correlations with the primary outcomes GvHD (acute or chronic GvHD), survival, and relapse were statistically analyzed. The results of these multicenter analyses show that none of the HLA class I-restricted HY antigens were found to be associated with any of the primary outcomes. Interestingly, of the HLA class II-restricted HY antigens analyzed, HLA-DQ5 positive recipients showed a significantly increased GvHD-free survival in female-to-male HSCT compared with male-to-female HSCT (P = .013). Yet, analysis of the overall gender effect, thus independent of the known HY antigens, between the gender groups demonstrated an increased GvHD incidence in the female-to-male transplantations (P < .005) and a decreased GvHD-free survival in the female-to-male transplantations (P < .001). Of all autosomally encoded minor H antigens, only mismatching for the broadly expressed minor H antigen HA-8 increased the GvHD incidence in IRD HSCT (Hazard ratio [HR] = 5.28, P < .005), but not in MUD HSCT. Most striking was the influence of hematopoietic restricted minor H antigens on GvL as mismatching for hematopoietic minor H antigens correlated with lower relapse rates (P = .078), higher relapse-free survival (P = .029), and higher overall survival (P = .032) in recipients with GvHD, but not in those without GvHD. In conclusion, the significant GvHD effect of the broadly expressed minor H antigen HA-8 favors matching for HA-8 in IRD, but not in MUD, patient/donor pairs. The GvHD-GvL association demonstrating a significant lower relapse in hematopoietic minor H antigen mismatched patient/donor pairs underlines their clinical applicability for adoptive immunotherapy, enhancing the GvL effect in a GvHD controllable manner.
Collapse
Affiliation(s)
- Eric Spierings
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bund D, Buhmann R, Gökmen F, Zorn J, Kolb HJ, Schmetzer HM. Minor histocompatibility antigen UTY as target for graft-versus-leukemia and graft-versus-haematopoiesis in the canine model. Scand J Immunol 2013; 77:39-53. [PMID: 23126655 DOI: 10.1111/sji.12011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 10/25/2012] [Indexed: 12/12/2022]
Abstract
Male patients with female-stem-cell donors have better prognosis compared to female-to-male combinations due to Y-encoded minor histocompatibility antigens recognized by female-alloimmune-effector lymphocytes in the context of a graft-versus-leukemia (GvL) effect. We provide data in a dog-model that the minor histocompatibility antigen UTY might be a promising target to further improve GvL-immune reactions after allogeneic-stem-cell transplantations. Female-canine-UTY-specific T cells (CTLs) were stimulated in vitro using autologous-DCs loaded with three HLA-A2-restricted-UTY-derived peptides (3-fold-expansion), and specific T cell responses were determined in 3/6 female dogs. CTLs specifically recognized/lysed autologous-female-peptide-loaded DCs, but not naïve-autologous-female DCs and monocytes. They mainly recognized bone-marrow (BM) and to a lower extent DCs, monocytes, PBMCs and B-cells from DLA-identical-male littermates and peptide-loaded T2-cells in an MHC-I-restricted manner. A UTY-/male-specific reactivity was also obtained in vivo after stimulation of a female dog with DLA-identical-male PBMCs. In summary, we demonstrated natural UTY processing and presentation in dogs. We showed that female-dog CTLs were specifically stimulated by HLA-A2-restricted-UTY peptides, thereby enabling recognition of DLA-identical-male cells, mainly BM cells. These observations suggest UTY as a promising candidate-antigen to improve GvL-reactions in the course of immunotherapy.
Collapse
Affiliation(s)
- D Bund
- Medical Department III, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany.
| | | | | | | | | | | |
Collapse
|
47
|
Zinöcker S, Dressel R, Wang XN, Dickinson AM, Rolstad B. Immune reconstitution and graft-versus-host reactions in rat models of allogeneic hematopoietic cell transplantation. Front Immunol 2012; 3:355. [PMID: 23226148 PMCID: PMC3510360 DOI: 10.3389/fimmu.2012.00355] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 11/08/2012] [Indexed: 12/28/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (alloHCT) extends the lives of thousands of patients who would otherwise succumb to hematopoietic malignancies such as leukemias and lymphomas, aplastic anemia, and disorders of the immune system. In alloHCT, different immune cell types mediate beneficial graft-versus-tumor (GvT) effects, regulate detrimental graft-versus-host disease (GvHD), and are required for protection against infections. Today, the “good” (GvT effector cells and memory cells conferring protection) cannot be easily separated from the “bad” (GvHD-causing cells), and alloHCT remains a hazardous medical modality. The transplantation of hematopoietic stem cells into an immunosuppressed patient creates a delicate environment for the reconstitution of donor blood and immune cells in co-existence with host cells. Immunological reconstitution determines to a large extent the immune status of the allo-transplanted host against infections and the recurrence of cancer, and is critical for long-term protection and survival after clinical alloHCT. Animal models continue to be extremely valuable experimental tools that widen our understanding of, for example, the dynamics of post-transplant hematopoiesis and the complexity of immune reconstitution with multiple ways of interaction between host and donor cells. In this review, we discuss the rat as an experimental model of HCT between allogeneic individuals. We summarize our findings on lymphocyte reconstitution in transplanted rats and illustrate the disease pathology of this particular model. We also introduce the rat skin explant assay, a feasible alternative to in vivo transplantation studies. The skin explant assay can be used to elucidate the biology of graft-versus-host reactions, which are known to have a major impact on immune reconstitution, and to perform genome-wide gene expression studies using controlled combinations of minor and major histocompatibility between the donor and the recipient.
Collapse
Affiliation(s)
- Severin Zinöcker
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo Oslo, Norway ; Department of Immunology, Oslo University Hospital - Rikshospitalet Oslo, Norway
| | | | | | | | | |
Collapse
|
48
|
Regulatory T cells inhibit CD8(+) T-cell tissue invasion in human skin graft-versus-host reactions. Transplantation 2012; 94:456-64. [PMID: 22890131 DOI: 10.1097/tp.0b013e31826205d6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Regulatory T cells (Tregs) effectively ameliorate graft-versus-host disease (GVHD). The mechanisms underlying Treg therapeutic effect on GVHD are not fully elucidated. This study investigates whether Treg prevention of GVH tissue damage is associated with blocking CD8 effector T-cell tissue invasion, a question not yet addressed in humans. METHOD Tissue-infiltrating T cells and histopathology scores were detected using an in vitro human GVHD skin explant model, together with immunohistochemistry, cytometric bead array, functional adhesion and migration assays, flow cytometry, and quantitative real-time polymerase chain reaction. RESULTS Treg intervention during priming significantly decreased effector T-cell infiltration into target tissue (P<0.01) resulting in a striking reduction in the histopathology score of tissue injury (P<0.0001). These results were coupled with reduced CXCR3 and cutaneous lymphocyte antigen expression by effector T cells, together with decreased CXCL10 and CXCL11 expression in target tissue. Treg intervention also impaired the functional interaction of CXCR3 and cutaneous lymphocyte antigen with their specific ligands (P<0.01) and suppressed the secretion of CXCL9, CXCL10, and interferon-γ (P<0.01, P<0.05, and P<0.001, respectively). Late addition of Tregs into the effector phase abolished their ability to suppress effector T-cell tissue invasion, resulting in a total loss of their ability to ameliorate GVH tissue damage. CONCLUSION Preventing effector T-cell tissue invasion is a critical mechanistic event leading to Treg attenuation of GVH tissue damage. This therapeutic effect is associated with a failure of CD8 T cells to increase tissue homing receptors after allo-stimulation, together with a breakdown of interferon-γ-induced chemoattractant expression in the target tissue.
Collapse
|
49
|
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
|
50
|
Tsirigotis P, Or R, Resnick IB, Shapira MY. Immunotherapeutic approaches to improve graft-versus-tumor effect and reduce graft-versus-host disease. Immunotherapy 2012; 4:407-24. [PMID: 22512635 DOI: 10.2217/imt.12.14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The therapeutic efficacy of allogeneic stem cell transplantation is mainly based on the alloreactive immune response of the graft against the host. However, the graft-versus-host process can be viewed as a double-edged sword since it is responsible for both the beneficial graft-versus-tumor effect and the deleterious graft-versus-host disease. During the last two decades, intensive research has been focused on the development of novel immunotherapeutic methods aimed to dissociate graft-versus-host disease from graft-versus-tumor effect. A brief description of these efforts is discussed in this review.
Collapse
Affiliation(s)
- Panagiotis Tsirigotis
- Department of Bone Marrow Transplantation & Cancer Immunotherapy, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| | | | | | | |
Collapse
|