1
|
Zhang Y, Ye C, Zhu H, Zhuang Y, Chen S, Weng Y, Ren J, Luo X, Zheng J, Zheng X, Li J, Lan L, Xie Y, Han Z, Hu J, Yang T. Association of iKIR-mismatch model and donor aKIRs with better outcome in haploidentical hematopoietic stem cell transplantation for acute myeloid leukemia. Front Immunol 2023; 13:1091188. [PMID: 36761162 PMCID: PMC9904326 DOI: 10.3389/fimmu.2022.1091188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 12/09/2022] [Indexed: 01/26/2023] Open
Abstract
Objectives Killer cell immunoglobulin like receptor (KIR) can trigger the alloreactivity of NK cells. However, there is no clear consensus as to their function. Here, we investigated the potential influence of KIR mismatch and KIR alleles on the outcome of haploidentical hematopoietic stem cell transplantation (haplo-HSCT) in acute myeloid leukemia (AML) patients. Method Data from 79 AML patients treated with haplo-HSCT were retrospectively analyzed. HLA-C genotyping was determined by the PCR-rSSO method. KIR, HLA-A and HLA-B genotyping was performed by the PCR-SSP method. Cox proportional hazards model and Kaplan-Meier survival curves were used for analysis. Results Both KIR ligand mismatch (KLM) group and KIR receptor-ligand mismatch (RLM) group were associated with a decreased risk in aGVHD and relapse rate (RR), and better overall survival (OS) compared to the KIR ligand matching and receptor-ligand matching groups, respectively (aGVHD: KLM: p=0.047, HR:0.235; RLM: p<0.001, HR:0.129; RR: KLM: p=0.049, HR:0.686, RLM: p=0.017, HR:0.200;OS:KLM: p=0.012, HR: 0.298, RLM: p=0.021, HR:0.301). RLM was more accurate at predicting relapse and aGVHD compared with KLM (aGVHD: p=0.009; RR: p=0.039). Patients with greater number of donor activating KIRs (aKIR) had a lower incidence of aGVHD and relapse, and the benefits correlated with the increase in the number of donor aKIRs (aGVHD: p=0.019, HR:0.156; RR: p=0.037, HR:0.211). Patients with RLM and the highest number of donor aKIRs had the lowest RR, lowest incidence of aGVHD and best OS. Conclusions Both KLM and RLM reduced the risk of aGVHD and relapse after haplo-HSCT in AML patients, and RLM showed superiority in predicting HSCT outcome. The synergistic effects of RLM and donor aKIRs can provide a better donor selection strategy to improve haplo-HSCT outcome in AML patients.
Collapse
Affiliation(s)
- Yu Zhang
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Chenjing Ye
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Haojie Zhu
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Youran Zhuang
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney, Australia
| | - Shaozhen Chen
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Yingxi Weng
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Jinhua Ren
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Xiaofeng Luo
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Jing Zheng
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Xiaoyun Zheng
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Jing Li
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Lingqiong Lan
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Yongxin Xie
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Zhongchao Han
- Chinese Academy of Medical Sciences, State Key Laboratory Experimental Hematology, Tianjin, China
| | - Jianda Hu
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| | - Ting Yang
- Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology Fuzhou, Fujian, China
| |
Collapse
|
2
|
Lu Y. Natural Killer (NK) cells in immunotherapy and perspectives in antitumour approaches. BIO WEB OF CONFERENCES 2022. [DOI: 10.1051/bioconf/20225501005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Natural Killer (NK) cells comprise a group of specialized innate lymphoid cells endowed with multiple cytotoxicity mechanisms while also harnessed with the ability to enhance other immune cells with cytokine production. This exclusive advantage of them to recognize and eliminate virally infected cells and tumour cells has been unmasked for decades, and previous clinical trials are also successfully tested for both efficacy and safety. With the emerging strategies in CAR-T cell therapy, such technologies can also be capable of further enhancing the viability of NK cell immunotherapy through cytokine armouring, chimeric antigen receptor (CAR) transduction, checkpoint inhibition, and co-stimulatory signals. Other than the conventional approach of engineering CAR to target tumour antigens, they are also capable of acting as blockers to the inhibitory compartments on tumour cells within the harsh environment to reduce the negative effects. Despite all these aspects, the tumour microenvironment (TME) is another essential facet when discussing cancer therapy owing to its characteristic setting that contributes immensely to immune evasion and immune function inhibition. In this review, I introduce the foundational mechanism for NK cytotoxicity and its signalling routes, discuss the impacts of TME on immune cells and their antitumour effects, evaluate possible strategies that overcome the current challenges, and propose a few potentially adoptive measures for future research in general immunotherapy from a perspective of molecular biology.
Collapse
|
3
|
Chander V, Gangenahalli G. Emerging strategies for enhancing the homing of hematopoietic stem cells to the bone marrow after transplantation. Exp Cell Res 2020; 390:111954. [PMID: 32156602 DOI: 10.1016/j.yexcr.2020.111954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/26/2022]
Abstract
Bone marrow failure is the primary cause of death after nuclear accidents or intentional exposure to high or low doses of ionizing radiation. Hematopoietic stem cell transplantation is the most potent treatment procedure for patients suffering from several hematopoietic malignancies arising after radiation injuries. Successful hematopoietic recovery after transplantation depends on efficient homing and subsequent engraftment of hematopoietic stem cells in specific niches within the bone marrow. It is a rapid and coordinated process in which circulating cells actively enter the bone marrow through the process known as transvascular migration, which involves the tightly regulated relay of events that finally leads to homing of cells in the bone marrow. Various adhesion molecules, chemokines, glycoproteins, integrins, present both on the surface of stem cells and sinusoidal endothelium plays a critical role in transvascular migration. But despite having an in-depth knowledge of homing and engraftment and the key events that regulate it, we are still not completely able to avoid graft failures and post-transplant mortalities. This deems it necessary to design a flawless plan for successful transplantation. Here, in this review, we will discuss the current clinical methods used to overcome graft failures and their flaws. We will also discuss, what are the new approaches developed in the past 10-12 years to selectively deliver the hematopoietic stem cells in the bone marrow by adopting proper targeting strategies that can help revolutionize the field of regenerative and translational medicine.
Collapse
Affiliation(s)
- Vikas Chander
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences, Delhi, 110054, India
| | - Gurudutta Gangenahalli
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences, Delhi, 110054, India.
| |
Collapse
|
4
|
Mechanistic understanding of β-cryptoxanthin and lycopene in cancer prevention in animal models. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158652. [PMID: 32035228 DOI: 10.1016/j.bbalip.2020.158652] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/26/2020] [Accepted: 01/28/2020] [Indexed: 02/07/2023]
Abstract
To better understand the potential function of carotenoids in the chemoprevention of cancers, mechanistic understanding of carotenoid action on genetic and epigenetic signaling pathways is critically needed for human studies. The use of appropriate animal models is the most justifiable approach to resolve mechanistic issues regarding protective effects of carotenoids at specific organs and tissue sites. While the initial impetus for studying the benefits of carotenoids in cancer prevention was their antioxidant capacity and pro-vitamin A activity, significant advances have been made in the understanding of the action of carotenoids with regards to other mechanisms. This review will focus on two common carotenoids, provitamin A carotenoid β-cryptoxanthin and non-provitamin A carotenoid lycopene, as promising chemopreventive agents or chemotherapeutic compounds against cancer development and progression. We reviewed animal studies demonstrating that β-cryptoxanthin and lycopene effectively prevent the development or progression of various cancers and the potential mechanisms involved. We highlight recent research that the biological functions of β-cryptoxanthin and lycopene are mediated, partially via their oxidative metabolites, through their effects on key molecular targeting events, such as NF-κB signaling pathway, RAR/PPARs signaling, SIRT1 signaling pathway, and p53 tumor suppressor pathways. The molecular targets by β-cryptoxanthin and lycopene, offer new opportunities to further our understanding of common and distinct mechanisms that involve carotenoids in cancer prevention. This article is part of a Special Issue entitled Carotenoids recent advances in cell and molecular biology edited by Johannes von Lintig and Loredana Quadro.
Collapse
|
5
|
Donor Selection Based on Killer Cell Immunoglobulin–Like Receptor (KIR) Genotype May Improve Outcome After T-Cell–Replete Haploidentical Transplantation. Transplant Proc 2018; 50:679-682. [DOI: 10.1016/j.transproceed.2017.09.070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 09/21/2017] [Indexed: 02/01/2023]
|
6
|
Jäger MD, Vondran FWR, Ramackers W, Röseler T, Schlitt HJ, Bektas H, Klempnauer J, Timrott K. A Depleting Anti-CD45 Monoclonal Antibody as Isolated Conditioning for Bone Marrow Transplantation in the Rat. PLoS One 2016; 11:e0154682. [PMID: 27139494 PMCID: PMC4854389 DOI: 10.1371/journal.pone.0154682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 04/18/2016] [Indexed: 11/19/2022] Open
Abstract
Objective A monoclonal antibody (mAb) against the leukocyte common antigen CD45 (RT7 in rats) could facilitate bone marrow transplantation (BMT). This study in rats evaluates a depletive rat anti-RT7a mAb as isolated tool for BMT conditioning without using irradiation or any chemotherapeutic / immunosuppressive agent. Methods The model used a CD45 di-allelic polymorphism (RT7a/RT7b). The anti-RT7a mAb was intravenously administered to LEW.1W rats (RT1uRT7a) at 5, 10 and 15 mg/kg. 1x108 BM cells of MHC syngeneic (RT1u), MHC disparate (RT1l) or MHC haploidentical (RT1u/l) donors were transplanted. All BM donor strains carried the RT7b allele so that their CD45+ cells were not affected by the anti-RT7a mAb. Recipients were monitored for reconstitution and donor-chimerism in blood leukocytes. Results mAb dosages of 5 or 10 mg/kg were myelosuppressive, whereas 15 mg/kg was myeloablative. Multi-lineage donor-chimerism at day 100 indicated engraftment of MHC syngeneic BM after any used mAb dosage (5 mg/kg: 46+/-7%; 10 mg/kg: 62+/-5%; 15 mg/kg: 80+/-4%). MHC disparate BM resulted in autologous reconstitution after conditioning by 10 mg/kg of the mAb and caused transient chimerism ending up in death associated with aplasia after conditioning by 15 mg/kg of the mAb. MHC haploidentical BM (F1 to parental) engrafted only after conditioning by 15 mg/kg (chimerism at day 100: 78+/-7%). Abandonment of α/β TCR+ cell depletion from BM grafts impaired the engraftment process after conditioning using 15 mg/kg of the mAb in the MHC syngeneic setting (2 of 6 recipients failed to engraft) and the MHC haploidentical setting (3 of 6 recipients failed). Conclusion This depletive anti-RT7a mAb is myelosuppressive and conditions for engraftment of MHC syngeneic BM. The mAb also facilitates engraftment of MHC haploidentical BM, if a myeloablative dose is used. RT7b expressing, BM-seeded α/β TCR+ cells seem to impair the engraftment process after myeloablative mAb conditioning.
Collapse
Affiliation(s)
- Mark D. Jäger
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Medizinische Hochschule Hannover, Hannover, Germany
- * E-mail:
| | - Florian W. R. Vondran
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Wolf Ramackers
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Tilmann Röseler
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Hans J. Schlitt
- Klinik und Poliklinik für Chirurgie, Klinikum der Universität Regensburg, Regensburg, Germany
| | - Hüseyin Bektas
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Jürgen Klempnauer
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Kai Timrott
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Medizinische Hochschule Hannover, Hannover, Germany
| |
Collapse
|
7
|
Mahr B, Unger L, Hock K, Pilat N, Baranyi U, Schwarz C, Maschke S, Farkas AM, Wekerle T. IL-2/α-IL-2 Complex Treatment Cannot Be Substituted for the Adoptive Transfer of Regulatory T cells to Promote Bone Marrow Engraftment. PLoS One 2016; 11:e0146245. [PMID: 26731275 PMCID: PMC4701413 DOI: 10.1371/journal.pone.0146245] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 12/15/2015] [Indexed: 12/16/2022] Open
Abstract
Cell therapy with recipient Tregs achieves engraftment of allogeneic bone marrow (BM) without the need for cytoreductive conditioning (i.e., without irradiation or cytotoxic drugs). Thereby mixed chimerism and transplantation tolerance are established in recipients conditioned solely with costimulation blockade and rapamycin. However, clinical translation would be substantially facilitated if Treg-stimulating pharmaceutical agents could be used instead of individualized cell therapy. Recently, it was shown that interleukin-2 (IL-2) complexed with a monoclonal antibody (mAb) (clone JES6-1A12) against IL-2 (IL-2 complexes) potently expands and activates Tregs in vivo. Therefore, we investigated whether IL-2 complexes can replace Treg therapy in a costimulation blockade-based and irradiation-free BM transplantation (BMT) model. Unexpectedly, the administration of IL-2 complexes at the time of BMT (instead of Tregs) failed to induce BM engraftment in non-irradiated recipients (0/6 with IL-2 complexes vs. 3/4 with Tregs, p<0.05). Adding IL-2 complexes to an otherwise effective regimen involving recipient irradiation (1Gy) but no Treg transfer indeed actively triggered donor BM rejection at higher doses (0/8 with IL-2 complexes vs. 9/11 without, p<0.01) and had no detectable effect at two lower doses (3/5 vs. 9/11, p>0.05). CD8 T cells and NK cells of IL-2 complex-treated naïve mice showed an enhanced proliferative response towards donor antigens in vitro despite the marked expansion of Tregs. However, IL-2 complexes also expanded conventional CD4 T cells, CD8 T cells, NK cells, NKT cells and notably even B cells, albeit to a lesser extent. Notably, IL-2 complex expanded Tregs featured less potent suppressive functions than in vitro activated Tregs in terms of T cell suppression in vitro and BM engraftment in vivo. In conclusion, these data suggest that IL-2 complexes are less effective than recipient Tregs in promoting BM engraftment and in contrast actually trigger BM rejection, as their effect is not sufficiently restricted to Tregs but rather extends to several other lymphocyte populations.
Collapse
Affiliation(s)
- Benedikt Mahr
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18–20, 1090, Vienna, Austria
| | - Lukas Unger
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18–20, 1090, Vienna, Austria
| | - Karin Hock
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18–20, 1090, Vienna, Austria
| | - Nina Pilat
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18–20, 1090, Vienna, Austria
| | - Ulrike Baranyi
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18–20, 1090, Vienna, Austria
| | - Christoph Schwarz
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18–20, 1090, Vienna, Austria
| | - Svenja Maschke
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18–20, 1090, Vienna, Austria
| | - Andreas Michael Farkas
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18–20, 1090, Vienna, Austria
| | - Thomas Wekerle
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18–20, 1090, Vienna, Austria
- * E-mail:
| |
Collapse
|
8
|
Ames E, Canter RJ, Grossenbacher SK, Mac S, Chen M, Smith RC, Hagino T, Perez-Cunningham J, Sckisel GD, Urayama S, Monjazeb AM, Fragoso RC, Sayers TJ, Murphy WJ. NK Cells Preferentially Target Tumor Cells with a Cancer Stem Cell Phenotype. THE JOURNAL OF IMMUNOLOGY 2015; 195:4010-9. [PMID: 26363055 DOI: 10.4049/jimmunol.1500447] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/10/2015] [Indexed: 01/02/2023]
Abstract
Increasing evidence supports the hypothesis that cancer stem cells (CSCs) are resistant to antiproliferative therapies, able to repopulate tumor bulk, and seed metastasis. NK cells are able to target stem cells as shown by their ability to reject allogeneic hematopoietic stem cells but not solid tissue grafts. Using multiple preclinical models, including NK coculture (autologous and allogeneic) with multiple human cancer cell lines and dissociated primary cancer specimens and NK transfer in NSG mice harboring orthotopic pancreatic cancer xenografts, we assessed CSC viability, CSC frequency, expression of death receptor ligands, and tumor burden. We demonstrate that activated NK cells are capable of preferentially killing CSCs identified by multiple CSC markers (CD24(+)/CD44(+), CD133(+), and aldehyde dehydrogenase(bright)) from a wide variety of human cancer cell lines in vitro and dissociated primary cancer specimens ex vivo. We observed comparable effector function of allogeneic and autologous NK cells. We also observed preferential upregulation of NK activation ligands MICA/B, Fas, and DR5 on CSCs. Blocking studies further implicated an NKG2D-dependent mechanism for NK killing of CSCs. Treatment of orthotopic human pancreatic cancer tumor-bearing NSG mice with activated NK cells led to significant reductions in both intratumoral CSCs and tumor burden. Taken together, these data from multiple preclinical models, including a strong reliance on primary human cancer specimens, provide compelling preclinical evidence that activated NK cells preferentially target cancer cells with a CSC phenotype, highlighting the translational potential of NK immunotherapy as part of a combined modality approach for refractory solid malignancies.
Collapse
Affiliation(s)
- Erik Ames
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Robert J Canter
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Steven K Grossenbacher
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Stephanie Mac
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Mingyi Chen
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Rachel C Smith
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Takeshi Hagino
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Jessica Perez-Cunningham
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Gail D Sckisel
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Shiro Urayama
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817
| | - Arta M Monjazeb
- Department of Radiation Oncology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Ruben C Fragoso
- Department of Radiation Oncology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Thomas J Sayers
- Basic Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD 21702; and
| | - William J Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95817; Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817
| |
Collapse
|
9
|
Kim N, Lee H, Shin J, Nam YS, Im KI, Lim JY, Lee ES, Kang YN, Park SH, Cho SG. Immune Reconstitution Kinetics following Intentionally Induced Mixed Chimerism by Nonmyeloablative Transplantation. PLoS One 2015; 10:e0126318. [PMID: 25961559 PMCID: PMC4427480 DOI: 10.1371/journal.pone.0126318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/31/2015] [Indexed: 11/18/2022] Open
Abstract
Establishing mixed chimerism is a promising approach for inducing donor-specific transplant tolerance. The establishment and maintenance of mixed chimerism may enable long-term engraftment of organ transplants while minimizing the use of immunosuppressants. Several protocols for inducing mixed chimerism have been reported; however, the exact mechanism underlying the development of immune tolerance remains to be elucidated. Therefore, understanding the kinetics of engraftment during early post-transplant period may provide insight into establishing long-term mixed chimerism and permanent transplant tolerance. In this study, we intentionally induced allogeneic mixed chimerism using a nonmyeloablative regimen by host natural killer (NK) cell depletion and T cell-depleted bone marrow (BM) grafts in a major histocompatibility complex (MHC)-mismatched murine model and analyzed the kinetics of donor (C57BL/6) and recipient (BALB/c) engraftment in the weeks following transplantation. Donor BM cells were well engrafted and stabilized without graft-versus-host disease (GVHD) as early as one week post-bone marrow transplantation (BMT). Donor-derived thymic T cells were reconstituted four weeks after BMT; however, the emergence of newly developed T cells was more obvious at the periphery as early as two weeks after BMT. Also, the emergence and changes in ratio of recipient- and donor-derived NKT cells and antigen presenting cells (APCs) including dendritic cells (DCs) and B cells were noted after BMT. Here, we report a longitudinal analysis of the development of donor- and recipient-originated hematopoietic cells in various lymphatic tissues of intentionally induced mixed chimerism mouse model during early post-transplant period. Through the understanding of immune reconstitution at early time points after nonmyeloablative BMT, we suggest guidelines on intentionally inducing durable mixed chimerism.
Collapse
Affiliation(s)
- Nayoun Kim
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul, Korea
- Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Hyunji Lee
- School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Junghoon Shin
- School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Young-Sun Nam
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul, Korea
- Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Keon-Il Im
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul, Korea
- Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Jung-Yeon Lim
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul, Korea
- Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Eun-Sol Lee
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul, Korea
- Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Young-Nam Kang
- Department of Radiation Oncology, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Se-Ho Park
- School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Seok-Goo Cho
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul, Korea
- Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine, Seoul, Korea
- Catholic Blood and Marrow Transplantation Center, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
10
|
Enforced fucosylation of cord blood hematopoietic cells accelerates neutrophil and platelet engraftment after transplantation. Blood 2015; 125:2885-92. [PMID: 25778529 DOI: 10.1182/blood-2015-01-607366] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 02/27/2015] [Indexed: 12/30/2022] Open
Abstract
Delayed engraftment is a major limitation of cord blood transplantation (CBT), due in part to a defect in the cord blood (CB) cells' ability to home to the bone marrow. Because this defect appears related to low levels of fucosylation of cell surface molecules that are responsible for binding to P- and E-selectins constitutively expressed by the marrow microvasculature, and thus for marrow homing, we conducted a first-in-humans clinical trial to correct this deficiency. Patients with high-risk hematologic malignancies received myeloablative therapy followed by transplantation with 2 CB units, one of which was treated ex vivo for 30 minutes with the enzyme fucosyltransferase-VI and guanosine diphosphate fucose to enhance the interaction of CD34(+) stem and early progenitor cells with microvessels. The results of enforced fucosylation for 22 patients enrolled in the trial were then compared with those for 31 historical controls who had undergone double unmanipulated CBT. The median time to neutrophil engraftment was 17 days (range, 12-34 days) compared with 26 days (range, 11-48 days) for controls (P = .0023). Platelet engraftment was also improved: median was 35 days (range, 18-100 days) compared with 45 days (range, 27-120 days) for controls (P = .0520). These findings support ex vivo fucosylation of multipotent CD34(+) CB cells as a clinically feasible means to improve engraftment efficiency in the double CBT setting. The trial is registered to www.clinicaltrials.gov as #NCT01471067.
Collapse
|
11
|
Yoon SR, Kim TD, Choi I. Understanding of molecular mechanisms in natural killer cell therapy. Exp Mol Med 2015; 47:e141. [PMID: 25676064 PMCID: PMC4346487 DOI: 10.1038/emm.2014.114] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 11/17/2014] [Accepted: 12/02/2014] [Indexed: 01/14/2023] Open
Abstract
Cancer cells and the immune system are closely related and thus influence each other. Although immune cells can suppress cancer cell growth, cancer cells can evade immune cell attack via immune escape mechanisms. Natural killer (NK) cells kill cancer cells by secreting perforins and granzymes. Upon contact with cancer cells, NK cells form immune synapses to deliver the lethal hit. Mature NK cells are differentiated from hematopoietic stem cells in the bone marrow. They move to lymph nodes, where they are activated through interactions with dendritic cells. Interleukin-15 (IL-15) is a key molecule that activates mature NK cells. The adoptive transfer of NK cells to treat incurable cancer is an attractive approach. A certain number of activated NK cells are required for adoptive NK cell therapy. To prepare these NK cells, mature NK cells can be amplified to obtain sufficient numbers of NK cells. Alternatively, NK cells can be differentiated and amplified from hematopoietic stem cells. In addition, the selection of donors is important to achieve maximal efficacy. In this review, we discuss the overall procedures and strategies of NK cell therapy against cancer.
Collapse
Affiliation(s)
- Suk Ran Yoon
- 1] Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea [2] Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Tae-Don Kim
- 1] Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea [2] Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Inpyo Choi
- 1] Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea [2] Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
12
|
Reprint of: Recent Advances in Cytomegalovirus: An Update on Pharmacologic and Cellular Therapies. Biol Blood Marrow Transplant 2015; 21:S19-24. [DOI: 10.1016/j.bbmt.2014.12.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 11/03/2014] [Indexed: 12/18/2022]
|
13
|
Recent advances in cytomegalovirus: an update on pharmacologic and cellular therapies. Biol Blood Marrow Transplant 2014; 21:24-9. [PMID: 25452035 DOI: 10.1016/j.bbmt.2014.11.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 11/03/2014] [Indexed: 12/19/2022]
Abstract
The 2015 Tandem American Society for Blood and Marrow Transplantation/Center for International Blood and Marrow Transplant Meetings provide an opportunity to review the current status and future perspectives on therapy for cytomegalovirus (CMV) infection in the setting of hematopoietic stem cell transplantation (HSCT). After many years during which we have seen few tangible advances in terms of new antiviral drugs, we are now experiencing an exciting period of late-stage drug development, characterized by a series of phase III trials incorporating a variety of novel agents. These trials have the potential to shift our current standard therapeutic strategies, which generally involve pre-emptive therapy based on sensitive molecular surveillance, towards the prophylactic approaches we see more generally with other herpes viruses such as herpes simplex and varicella zoster. This comes at a time when the promise of extensive preclinical research has been translated into encouraging clinical responses with several cellular immunotherapy strategies, which have also been moved towards definitive late-stage clinical trials. How these approaches will be integrated with the new wave of antiviral drugs remains open to conjecture. Although most of the focus of these cellular immunotherapy studies has been on adaptive immunity, and in particular T cells, an increasing awareness of the possible role of other cellular subsets in controlling CMV infection has developed. In particular, the role of natural killer (NK) cells is being revisited, along with that of γδ T cells. Depletion of NK cells in mice results in higher titers of murine CMV in tissues and increased mortality, whereas NK cell deficiency in humans has been linked to severe CMV disease. We will review recent progress in these areas.
Collapse
|
14
|
Advantages and clinical applications of natural killer cells in cancer immunotherapy. Cancer Immunol Immunother 2013; 63:21-8. [PMID: 23989217 DOI: 10.1007/s00262-013-1469-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 08/14/2013] [Indexed: 01/27/2023]
Abstract
The past decade has witnessed a burgeoning of research and further insight into the biology and clinical applications of natural killer (NK) cells. Once thought to be simple innate cells important only as cytotoxic effector cells, our understanding of NK cells has grown to include memory-like responses, the guidance of adaptive responses, tissue repair, and a delicate paradigm for how NK cells become activated now termed "licensing" or "arming." Although these cells were initially discovered and named for their spontaneous ability to kill tumor cells, manipulating NK cells in therapeutic settings has proved difficult and complex in part due to our emerging understanding of their biology. Therapies involving NK cells may either activate endogenous NK cells or involve transfers of exogenous cells by hematopoietic stem cell transplantation or adoptive cell therapy. Here, we review the basic biology of NK cells, highlighting characteristics which make NK cells particularly useful in cancer therapies. We also explore current treatment strategies that have been used for cancer as well as discuss potential future directions for the field.
Collapse
|
15
|
Denman CJ, Senyukov VV, Somanchi SS, Phatarpekar PV, Kopp LM, Johnson JL, Singh H, Hurton L, Maiti SN, Huls MH, Champlin RE, Cooper LJN, Lee DA. Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PLoS One 2012; 7:e30264. [PMID: 22279576 PMCID: PMC3261192 DOI: 10.1371/journal.pone.0030264] [Citation(s) in RCA: 448] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 12/12/2011] [Indexed: 01/03/2023] Open
Abstract
NK cells have therapeutic potential for a wide variety of human malignancies. However, because NK cells expand poorly in vitro, have limited life spans in vivo, and represent a small fraction of peripheral white blood cells, obtaining sufficient cell numbers is the major obstacle for NK-cell immunotherapy. Genetically-engineered artificial antigen-presenting cells (aAPCs) expressing membrane-bound IL-15 (mbIL15) have been used to propagate clinical-grade NK cells for human trials of adoptive immunotherapy, but ex vivo proliferation has been limited by telomere shortening. We developed K562-based aAPCs with membrane-bound IL-21 (mbIL21) and assessed their ability to support human NK-cell proliferation. In contrast to mbIL15, mbIL21-expressing aAPCs promoted log-phase NK cell expansion without evidence of senescence for up to 6 weeks of culture. By day 21, parallel expansion of NK cells from 22 donors demonstrated a mean 47,967-fold expansion (median 31,747) when co-cultured with aAPCs expressing mbIL21 compared to 825-fold expansion (median 325) with mbIL15. Despite the significant increase in proliferation, mbIL21-expanded NK cells also showed a significant increase in telomere length compared to freshly obtained NK cells, suggesting a possible mechanism for their sustained proliferation. NK cells expanded with mbIL21 were similar in phenotype and cytotoxicity to those expanded with mbIL15, with retained donor KIR repertoires and high expression of NCRs, CD16, and NKG2D, but had superior cytokine secretion. The mbIL21-expanded NK cells showed increased transcription of the activating receptor CD160, but otherwise had remarkably similar mRNA expression profiles of the 96 genes assessed. mbIL21-expanded NK cells had significant cytotoxicity against all tumor cell lines tested, retained responsiveness to inhibitory KIR ligands, and demonstrated enhanced killing via antibody-dependent cell cytotoxicity. Thus, aAPCs expressing mbIL21 promote improved proliferation of human NK cells with longer telomeres and less senescence, supporting their clinical use in propagating NK cells for adoptive immunotherapy.
Collapse
Affiliation(s)
- Cecele J. Denman
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
| | - Vladimir V. Senyukov
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
| | - Srinivas S. Somanchi
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
| | - Prasad V. Phatarpekar
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
- Graduate School of Biomedical Sciences, Health Science Center, The University of Texas, Houston, Texas, United States of America
| | - Lisa M. Kopp
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
| | - Jennifer L. Johnson
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
| | - Harjeet Singh
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
| | - Lenka Hurton
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
- Graduate School of Biomedical Sciences, Health Science Center, The University of Texas, Houston, Texas, United States of America
| | - Sourindra N. Maiti
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
| | - M. Helen Huls
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
| | - Richard E. Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
| | - Laurence J. N. Cooper
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
- Graduate School of Biomedical Sciences, Health Science Center, The University of Texas, Houston, Texas, United States of America
| | - Dean A. Lee
- Division of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, United States of America
- Graduate School of Biomedical Sciences, Health Science Center, The University of Texas, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
16
|
Nonobese diabetic natural killer cells: a barrier to allogeneic chimerism that can be reduced by rapamycin. Transplantation 2011; 92:977-84. [PMID: 21956197 DOI: 10.1097/tp.0b013e3182313e70] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Induction of allogeneic hematopoietic chimerism is a promising strategy to induce tolerance to donor islets for treating type 1 diabetes. Successful induction of chimerism requires overcoming host alloimmunity. In diabetes-prone nonobese diabetic (NOD) mice, this is challenging due to their general tolerance resistance. Although the adaptive alloimmunity of NOD mice is a known barrier to allogeneic chimerism, whether NOD natural killer (NK) cells are an additional barrier has not been examined. Because NOD NK cells exhibit functional defects, they may not inhibit chimerism generation. METHODS Antibody depletion of NK cells in vivo, or transplantation of F1 hybrid donor cells to eliminate the "missing-self" trigger of NK cells, was preformed to test the NK-mediated rejection of donor bone marrow cells. We also studied the capacity of rapamycin to block the NK cell response against allogeneic cells in vivo. RESULTS Depleting NK cells or rendering them unresponsive to the donor greatly improved the level of chimerism obtained in NOD mice. Rapamycin significantly reduced the resistance to allogeneic chimerism mounted by NOD NK cells; however, it was much less effective than NK cell depletion by antibodies. CONCLUSIONS Contrary to the view that NOD NK cells are defective, we found these cells to be a substantial barrier to allogeneic chimerism in the presence or absence of adaptive immunity. Moreover, rapamycin will need to be combined with other approaches to fully overcome the NK cell barrier.
Collapse
|
17
|
Barao I, Alvarez M, Redelman D, Weiss JM, Ortaldo JR, Wiltrout RH, Murphy WJ. Hydrodynamic delivery of human IL-15 cDNA increases murine natural killer cell recovery after syngeneic bone marrow transplantation. Biol Blood Marrow Transplant 2011; 17:1754-64. [PMID: 21906575 DOI: 10.1016/j.bbmt.2011.08.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 08/31/2011] [Indexed: 01/26/2023]
Abstract
Immune deficiency immediately following bone marrow transplantation (BMT) increases susceptibility to opportunistic infections as well as tumor relapse. Natural Killer (NK) cells play important roles in the resistance to virally infected and transformed cells. Interleukin (IL)-15 has been shown to be essential for NK cell development and survival. We administered human (h) IL-15 cDNA (pIL-15) via hydrodynamic delivery to murine recipients undergoing congenic BMT to determine its effects on NK cell reconstitution. Hydrodynamic pIL-15 delivery resulted in high levels of hIL-15 protein in the serum that lasted for several days and then quickly declined. The appearance of hIL-15 was followed by a significant increase of mature donor-derived NK cells within the bone marrow, spleens, and livers of the treated recipients. No accumulation of immature NK cell progenitors was observed. The NK cells from IL-15-treated recipients displayed an activated phenotype and were lytically active toward tumor targets in vitro to a similar degree as did those cells from recipients treated with control plasmid. This suggests that the predominant effect of IL-15 was a quantitative increase in total NK cell numbers and not qualitative changes in NK cell functions. No toxicities or adverse effects were observed. Studies performed in transplanted mice bearing renal carcinoma tumors demonstrated that this mode of hIL-15 gene delivery resulted in increased antitumor responses. These results support the use of cytokine gene transfer-based regimens as a platform to augment NK cell recovery after BMT.
Collapse
Affiliation(s)
- Isabel Barao
- University of Nevada, Reno, Department of Microbiology and Immunology, Reno, Nevada, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Barao I, Alvarez M, Ames E, Orr MT, Stefanski HE, Blazar BR, Lanier LL, Anderson SK, Redelman D, Murphy WJ. Mouse Ly49G2+ NK cells dominate early responses during both immune reconstitution and activation independently of MHC. Blood 2011; 117:7032-41. [PMID: 21498673 PMCID: PMC3143551 DOI: 10.1182/blood-2010-11-316653] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 03/28/2011] [Indexed: 11/20/2022] Open
Abstract
Natural killer (NK) cell subsets can be defined by the differential expression of inhibitory receptors for MHC class I molecules. Early after congenic HSCT, we found that Ly49G2(high) single-positive NK cells repopulated, displayed an activated phenotype, and were highly cytolytic. Over time, this subset was replaced with NK cells with a normal pattern of Ly49 expression. Treatment of mice with IL-2 also resulted in the rapid expansion of these Ly49G2(high) single-positive NK cells. Only the Ly49g (Klra7) Pro1 transcript was highly induced in both HSCT- and IL-2-treated recipients. MHC-independent expansion of the Ly49G2(+) subset was also observed after Listeria monocytogenes or mouse cytomegalovirus infection. Our data indicate that during reconstitution after HSCT and various activation stimuli, Ly49G2(+) NK cells represent the "first-responder" NK cells, which occur independently of NK-cell licensing via Ly49-MHC interactions. These data suggest that the inhibitory Ly49G2 receptor represents an activation marker on mouse NK cells under various conditions.
Collapse
MESH Headings
- Animals
- Biomarkers/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Cytomegalovirus/immunology
- Female
- Gene Expression Regulation
- Graft Survival
- Hematopoietic Stem Cell Transplantation
- Interleukin-2/metabolism
- Killer Cells, Natural/cytology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Listeria monocytogenes/immunology
- Lymphocyte Activation
- Major Histocompatibility Complex
- Mice
- Mice, Congenic
- Mice, Inbred Strains
- Neoplasms/immunology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Recombinant Proteins/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Isabel Barao
- Department of Microbiology and Immunology, University of Nevada, Reno, Reno, NV, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Symons HJ, Leffell MS, Rossiter ND, Zahurak M, Jones RJ, Fuchs EJ. Improved survival with inhibitory killer immunoglobulin receptor (KIR) gene mismatches and KIR haplotype B donors after nonmyeloablative, HLA-haploidentical bone marrow transplantation. Biol Blood Marrow Transplant 2009; 16:533-42. [PMID: 19961944 DOI: 10.1016/j.bbmt.2009.11.022] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2009] [Accepted: 11/24/2009] [Indexed: 12/11/2022]
Abstract
Natural killer (NK) cell alloreactivity, which may contribute to the graft-versus-leukemia (GVL) effect of allogeneic hematopoietic stem cell transplantation (HSCT), is influenced by the interaction of killer-cell immunoglobulin-like receptors (KIRs) on donor NK cells and their ligands, human leukocyte antigen (HLA) class I molecules on recipient antigen-presenting cells (APCs). Distinct models to predict NK cell alloreactivity differ in their incorporation of information from typing of recipient and donor KIR and HLA gene loci, which exist on different autosomes and are inherited independently as haplotypes. Individuals may differ in the inheritance of the 2 KIR haplotypes, A and B, or in the expression of individual KIR genes. Here, we examined the effect of KIR and HLA genotype, in both the recipient and donor, on the outcome of 86 patients with advanced hematologic malignancies who received nonmyeloablative (NMA), HLA-haploidentical HSCT with high-dose, posttransplantation cyclophosphamide (Cy). Compared to recipients of bone marrow (BM) from donors with identical KIR gene content, recipients of inhibitory KIR (iKIR) gene-mismatched BM had an improved overall survival (OS) (hazard ratio [HR]=0.37; confidence interval [CI]: 0.21-0.63; P=.0003), event-free survival (EFS) (HR=0.51; CI: 0.31-0.84; P=.01), and relapse rate (cause-specific HR, SDHR=0.53; CI: 0.31-0.93; P=.025). Patients homozygous for the KIR "A" haplotype, which encodes only 1 activating KIR, had an improved OS (HR=0.30; CI: 0.13-10.69; P=.004), EFS (HR=0.47; CI: 0.22-1.00; P=.05), and nonrelapse mortality (NRM; cause-specific HR=0.13; CI: 0.017-0.968; P=.046) if their donor expressed at least 1 KIR B haplotype that encodes several activating KIRs. Models that incorporated information from recipient HLA typing, with or without donor HLA typing, were not predictive of outcome in this patient cohort. Thus, NMA conditioning and T cell-replete, HLA-haploidentical HSCTs involving iKIR gene mismatches between donor and recipient, or KIR haplotype AA recipients of BM from KIR Bx donors, were associated with lower relapse and NRM and improved OS and EFS. These findings suggest that selection of donors based upon inhibitory KIR gene or haplotype incompatibility may be warranted.
Collapse
Affiliation(s)
- Heather J Symons
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University,1650 Orleans Street, Baltimore, MD 21231, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Tabayoyong WB, Salas JG, Bonde S, Zavazava N. HOXB4-transduced embryonic stem cell-derived Lin-c-kit+ and Lin-Sca-1+ hematopoietic progenitors express H60 and are targeted by NK cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:5449-57. [PMID: 19828634 DOI: 10.4049/jimmunol.0901807] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Embryonic stem (ES) cells are a novel source of cells, especially hematopoietic progenitor cells that can be used to treat degenerative diseases in humans. However, there is a need to determine how ES cell-derived progenitors are regulated by both the adaptive and innate immune systems post transplantation. In this study, we demonstrate that hematopoietic progenitor cells (HPCs) derived from mouse ES cells ectopically expressing HOXB4 fail to engraft long-term in the presence of NK cells. In particular, the H60-expressing Lin(-)c-kit(+) and Lin(-)Sca-1(+) subpopulations were preferentially deleted in Rag2(-/-), but not in Rag2(-/-)gamma(c)(-/-) mice. Up-regulation of class I expression on HPCs prevented their lysis by NK cells, and Ab-mediated depletion of NK cells restored long-term HPC engraftment. In contrast to the notion that ES-derived cells are immune-privileged, we show in this study that NK cells form a formidable barrier to the long-term engraftment of ES cell-derived hematopoietic progenitors.
Collapse
Affiliation(s)
- William B Tabayoyong
- Medical Scientist Training Program and Immunology Graduate Program, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
21
|
Kim HR, Lee KH, Park SJ, Kim SY, Yang YK, Tae J, Kim J. Anti-cancer activity and mechanistic features of a NK cell activating molecule. Cancer Immunol Immunother 2009; 58:1691-700. [PMID: 19259669 PMCID: PMC11030105 DOI: 10.1007/s00262-009-0680-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Accepted: 02/05/2009] [Indexed: 01/06/2023]
Abstract
Natural cytotoxicity receptors (NCRs) are major activating receptors involved in NK cytotoxicity. NCR expression varies with the activation state of NK cells, and the expression level correlates with NK cells' natural cytotoxicity. In this study, we found that Gö6983, a PKC inhibitor, induced a remarkable increase of NCR expression on primary NK cells, but other PKC inhibitors and NK cell stimulators such as IL-2 and PMA, did not. Gö6983 increased the expression of NCR in a time- and concentration-dependent manner. Furthermore, Gö6983 strongly upregulated the surface expression of death ligands FasL and TRAIL, but not cytotoxic molecules perforin and granzyme B. Unlike two other NK stimulating molecules, IL-2, and PMA, Gö6983 did not induce NK cell proliferation. Up-regulation of NCRs and death ligands on NK cells by Gö6983 resulted in a significant enhancement of NK cytotoxicity against various cancer cell lines. Most importantly, administration of Gö6983 effectively inhibited pulmonary tumor metastasis in mice in a dose-dependent manner. These results suggest that Gö6983 functions as an NK cell activating molecule (NKAM); this NKAM is a novel anti-cancer and anti-metastasis drug candidate because it enhances NK cytotoxicity against cancer cells in vivo as well as in vitro.
Collapse
Affiliation(s)
- Hyung-Ran Kim
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea
| | - Kyoung-Ho Lee
- Department of Microbiology, Yonsei University Wonju College of Medicine, Wonju, Kangwon Korea
| | - Su Jung Park
- Department of Microbiology, Yonsei University Wonju College of Medicine, Wonju, Kangwon Korea
| | - So Young Kim
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea
| | | | - Jinsung Tae
- Department of Chemistry, Yonsei University, Seoul, Korea
| | - Jongsun Kim
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemoon-gu, Seoul, 120-752 Korea
| |
Collapse
|
22
|
A chromosome 16 quantitative trait locus regulates allogeneic bone marrow engraftment in nonmyeloablated mice. Blood 2009; 114:202-10. [PMID: 19417206 DOI: 10.1182/blood-2009-03-208801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Identifying genes that regulate bone marrow (BM) engraftment may reveal molecular targets for overcoming engraftment barriers. To achieve this aim, we applied a forward genetic approach in a mouse model of nonmyeloablative BM transplantation. We evaluated engraftment of allogeneic and syngeneic BM in BALB.K and B10.BR recipients. This allowed us to partition engraftment resistance into its intermediate phenotypes, which are firstly the immune-mediated resistance and secondly the nonimmune rejection of donor BM cells. We observed that BALB.K and B10.BR mice differed with regard to each of these resistance mechanisms, thereby providing evidence that both are under genetic control. We then generated a segregating backcross (n = 200) between the BALB.K and B10.BR strains to analyze for genetic linkage to the allogeneic BM engraftment phenotype using a 127-marker genome scan. This analysis identified a novel quantitative trait locus (QTL) on chromosome 16, termed Bmgr5 (logarithm of odds 6.4, at 11.1 cM). The QTL encodes susceptibility alleles, from the BALB.K strain, that are permissive for allogeneic BM engraftment. Further identification of Bmgr5 genes by positional cloning may reveal new and effective approaches for overcoming BM engraftment obstacles.
Collapse
|
23
|
Natural killer cells in allogeneic transplantation: effect on engraftment, graft- versus-tumor, and graft-versus-host responses. Biol Blood Marrow Transplant 2009; 15:765-76. [PMID: 19539207 DOI: 10.1016/j.bbmt.2009.01.019] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 01/23/2009] [Indexed: 11/21/2022]
Abstract
Natural killer (NK) cells are effectors of the innate immune system and recognize cells transformed by viruses or neoplasia. Their response to "missing self" signals was described 3 decades ago, but the recent discovery of a panoply of activating receptors has made it clear that NK cell reactivity arises from a combination of inhibitory and activating signals. Successful clinical exploitation of NK cell reactivity was demonstrated in allogeneic transplantation for acute myelogenous leukemia from HLA-haploidentical donors when matched donors were not available. Multiple clinical studies have since attempted to use NK reactivity in the setting of both HLA-matched and -mismatched transplantation, with varying results. This review summarizes the heterogeneous clinical results and explains them based on a succinct description of NK cell biology.
Collapse
|
24
|
Suppression of graft-versus-host disease after adoptive infusion of alloreactive NK cells induced by silencing Ly49C gene in mice. Transpl Immunol 2009; 20:243-8. [DOI: 10.1016/j.trim.2008.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 10/19/2008] [Accepted: 10/21/2008] [Indexed: 11/20/2022]
|
25
|
Nakajima S, Hida S, Taki S. IL-15 inhibits pre-B cell proliferation by selectively expanding Mac-1+B220+ NK cells. Biochem Biophys Res Commun 2008; 369:1139-43. [DOI: 10.1016/j.bbrc.2008.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Accepted: 03/05/2008] [Indexed: 01/12/2023]
|
26
|
Sivula J, Volin L, Porkka K, Vettenranta K, Itälä M, Partanen J, Turpeinen H. Killer-cell immunoglobulin-like receptor ligand compatibility in the outcome of Finnish unrelated donor hematopoietic stem cell transplantation. Transpl Immunol 2007; 18:62-6. [PMID: 17584604 DOI: 10.1016/j.trim.2007.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 03/02/2007] [Accepted: 03/23/2007] [Indexed: 11/19/2022]
Abstract
Incompatibility in killer-cell immunoglobulin-like receptor (KIR) ligand between recipient and donor of hematopoietic stem cell transplantation has been reported to lead to natural killer (NK) cell activation. This activation may result in better transplantation outcome through reduced risk of graft-versus-host (GvH) disease, relapse and mortality. In the present study the effect of KIR ligand incompatibility was investigated retrospectively in 186 unrelated stem cell transplantations performed in Finland during years 1993-2004. No clear evidence for a better outcome in cases with KIR ligand incompatibility was obtained. Transplantation-related mortality was 64% in Kaplan-Meier analysis in the GvH direction KIR ligand-mismatched group and 33% in the KIR ligand-matched group. This difference was statistically non-significant. Consequently, no support could be obtained for a beneficial effect of KIR ligand incompatibility in the present set of unrelated donor transplantations.
Collapse
Affiliation(s)
- J Sivula
- Research and Development, Finnish Red Cross Blood Service, Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
27
|
Welniak LA, Blazar BR, Murphy WJ. Immunobiology of allogeneic hematopoietic stem cell transplantation. Annu Rev Immunol 2007; 25:139-70. [PMID: 17129175 DOI: 10.1146/annurev.immunol.25.022106.141606] [Citation(s) in RCA: 369] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) has evolved into an effective adoptive cellular immunotherapy for the treatment of a number of cancers. The immunobiology of allogeneic HSCT is unique in transplantation in that it involves potential immune recognition and attack between both donor and host. Much of the immunobiology of allogeneic HSCT has been gleaned from preclinical models and correlation with clinical observations. We review our current understanding of some of the issues that affect the success of this therapy, including host-versus-graft (HVG) reactions, graft-versus-host disease (GVHD), graft-versus-tumor (GVT) activity, and restoration of functional immunity to prevent transplant-related opportunistic infections. We also review new strategies to optimize the GVT and improve overall immune function while reducing GVHD and graft rejection.
Collapse
Affiliation(s)
- Lisbeth A Welniak
- Department of Microbiology and Immunology, University of Nevada, Reno, Nevada 89557, USA.
| | | | | |
Collapse
|
28
|
Keller MD, Chen DF, Condron SA, Liu N, Reinsmoen NL, Buckley RH. The effect of natural killer cell killer Ig-like receptor alloreactivity on the outcome of bone marrow stem cell transplantation for severe combined immunodeficiency (SCID). J Clin Immunol 2006; 27:109-16. [PMID: 17191149 DOI: 10.1007/s10875-006-9058-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2006] [Accepted: 11/15/2006] [Indexed: 11/30/2022]
Abstract
Natural killer (NK) cell alloreactions against recipient cells in the setting of bone marrow transplantation have been associated with decreased rates of graft-versus-host disease (GVHD) and improved survival in transplant recipients with myeloid leukemia. These alloreactions are predicted by the absence of recipient HLA class I ligands for donor inhibitory killer Ig-like receptors (KIR). We hypothesized that donor NK cell alloreactions against recipient cells may affect the development of T and B-cell functions and incidence of GVHD in infants with severe combined immunodeficiency (SCID). Of the 156 patients with SCID who had received related bone marrow transplants without pretransplant chemotherapy or posttransplant GVHD prophylaxis, 137 patient-donor pairs were evaluated for the absence of recipient HLA class I ligands for donor inhibitory KIR. Analysis showed that the absence of a KIR ligand had no effect on the incidence or severity of GVHD (RR [corrected] = 0.95, p = 0.84), development of T-cell function (RR [corrected] = 1.05, p = 0.69), production of IgA (p = 0.46) or IgM (p = 0.33), or on 5-year survival (RR [corrected] = 1.21, p = 0.10). Further, in patients possessing native NK cells, the absence of KIR ligands in donors for recipient-inhibitory KIR did not alter transplantation outcomes. This study suggests that inhibitory KIR/HLA interactions do not play a significant role in bone marrow transplantation for SCID.
Collapse
Affiliation(s)
- M D Keller
- Division of Pediatric Allergy and Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | |
Collapse
|
29
|
Passweg JR, Koehl U, Uharek L, Meyer-Monard S, Tichelli A. Natural-killer-cell-based treatment in haematopoietic stem-cell transplantation. Best Pract Res Clin Haematol 2006; 19:811-24. [PMID: 16997185 DOI: 10.1016/j.beha.2006.06.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Adoptive immunotherapy using natural killer (NK) cells is currently under investigation, especially in situations where anti-neoplastic effect is needed but infusion of T cells is considered hazardous, such as in recipients of haematopoietic stem-cell transplantation (HSCT) from haploidentical donors. NK-cell therapy is mainly but not exclusively investigated in the setting of allogeneic stem-cell transplantation. NK cells may induce potent anti-leukaemic and possibly anti-rejection activity, and may even mitigate graft-versus-host disease (GvHD). It remains to be determined whether such effects are clinically important and whether or not they are mediated mainly or exclusively by KIR-HLA class I interactions. Recent advances in graft engineering has provided methods for isolating large numbers of purified NK cells. Several groups have shown that clinical-grade NK cells at doses up to 10(7)/kg may be collected and purified for the purpose of infusion to patients. Early results in a limited number of patients show that these cell doses may be administered without adverse events and possibly without inducing GvHD. Further study is required to determine whether such infusions will be useful in preventing graft rejection, exerting graft-versus-leukaemia effects, and/or hastening immune recovery.
Collapse
Affiliation(s)
- Jakob R Passweg
- Service d'Hématologie, Departement Medecine Interne, Hôpitaux Universitaires de Genève, Genève, Switzerland.
| | | | | | | | | |
Collapse
|
30
|
Zimmerman ZF, Levy RB. MiHA reactive CD4 and CD8 T-cells effect resistance to hematopoietic engraftment following reduced intensity conditioning. Am J Transplant 2006; 6:2089-98. [PMID: 16796724 DOI: 10.1111/j.1600-6143.2006.01428.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Reduced intensity conditioning (RIC) prior to allogeneic hematopoietic cell transplantation (HCT) has shown promise in lowering the incidence of post-transplant complications including infection and graft-versus-host disease. T-cell-mediated graft rejection, however, remains a crucial factor in determining how 'mild' a level of immunosuppression can be administered. Understanding the kinetics of resistance responses as well as the role of CD4+ and CD8+ T cells underlies the development of protocols to circumvent resistance and support hematopoietic engraftment. In these studies, a major histocompatibility complex (MHC)-matched/minor histocompatibility antigen (MiHA) disparate RIC HCT model was developed in which resistance against donor hematopoietic progenitors as well as mature peripheral blood cells could be assessed. Interestingly, resistance was diminished in the absence of either host CD4+ or CD8+ T cells. However, its impairment was more severe in CD4-/- mice where resistance was not detected. Host CD4+ T cells were required for optimal expansion of specific (H60) T-cell receptor (TCR) expressing host anti-donor MiHA reactive CD8+ T cells following HCT. These observations demonstrate a critical role for host CD4+ T cells in resistance against MiHA disparate HCT. This RIC HCT resistance model will be useful for the analysis of the barrier to engraftment mediated by host T cells and the development of strategies to support engraftment.
Collapse
Affiliation(s)
- Z F Zimmerman
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | | |
Collapse
|
31
|
Zimmerman Z, Jones M, Shatry A, Komatsu M, Mammolenti M, Levy R. Cytolytic pathways used by effector cells derived from recipient naive and memory T cells and natural killer cells in resistance to allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 2006; 11:957-71. [PMID: 16338617 DOI: 10.1016/j.bbmt.2005.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Accepted: 07/11/2005] [Indexed: 11/23/2022]
Affiliation(s)
- Zachary Zimmerman
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida 33101, USA
| | | | | | | | | | | |
Collapse
|
32
|
Smith DM, Agura ED, Levy MF, Melton LB, Domiati-Saad R, Klintmalm G. Graft vs host disease following kidney transplantation using an '0 HLA antigen mismatched' donor. Nephrol Dial Transplant 2006; 21:2656-9. [PMID: 16627604 DOI: 10.1093/ndt/gfl174] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Douglas M Smith
- Transplant Immunology Laboratory, Baylor University Medical Center, 3500 Gaston Avenue, Dallas, TX 75246, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Sun JY, Gaidulis L, Dagis A, Palmer J, Rodriguez R, Miller MM, Forman SJ, Senitzer D. Killer Ig-like receptor (KIR) compatibility plays a role in the prevalence of acute GVHD in unrelated hematopoietic cell transplants for AML. Bone Marrow Transplant 2005; 36:525-30. [PMID: 16025153 DOI: 10.1038/sj.bmt.1705089] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Killer Ig-like receptor (KIR) is a major cluster of the natural killer cell receptors and may play a role in the outcome of hematopoietic cell transplants. A total of 65 AML cases transplanted with T-replete hematopoietic cells from unrelated donors were retrospectively KIR-genotyped by a multiplex PCR method of our own design. The KIR gene frequency and genotype patterns in these 130 samples were consistent with the data in the literature. Based upon overall inhibitory and activating KIR genes in both donors and patients, we developed an algorithm to calculate a compatibility score for each transplant case as plus, zero or minus. Significantly higher incidence (18/20, 90%) of acute (a) GVHD (grade II-IV) was found in the transplant cases with plus scores than that (25/45, 56%) in the cases with zero or minus scores (P < 0.01). When the scores are sorted in the opposite way, fewer cases (13/26, 50%) of aGVHD were found in the transplants with minus scores than that (30/39, 77%) in the transplants with zero or plus scores (P < 0.05). The difference of aGVHD prevalence between the plus score and minus score groups is highly significant (P < 0.01). KIR genotype compatibility calculated by this algorithm may predict aGVHD incidence and be helpful in choosing donors.
Collapse
Affiliation(s)
- J Y Sun
- Division of Hematology and Hematopoietic Cell Transplantation (HCT), City of Hope National Medical Center and Beckman Research Institute, Duarte, CA 91010, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Bignon JD, Gagne K. KIR matching in hematopoietic stem cell transplantation. Curr Opin Immunol 2005; 17:553-9. [PMID: 16085405 DOI: 10.1016/j.coi.2005.07.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2005] [Accepted: 07/21/2005] [Indexed: 10/25/2022]
Abstract
Although the key role of MHC-restricted T lymphocytes in hematopoietic stem cell transplantation (HSCT) has been known for a long time, recent data have focused on complementary or alternative effector cell populations, and in particular on NK cells. Spontaneously generated NK cell alloreactivity from stem cell grafts involves specific interactions between NK receptors, including killer immunoglobulin-like receptors (KIRs) and their MHC class I ligands. The combined effects of HLA and KIR polymorphic genes might explain discrepancies in the impact of donor-recipient matching observed in HSCT.
Collapse
Affiliation(s)
- Jean-Denis Bignon
- HLA laboratory, Etablissement Français du Sang, Pays de Loire, 44011 Nantes, France.
| | | |
Collapse
|
35
|
Rubio MT, Saito TI, Kattleman K, Zhao G, Buchli J, Sykes M. Mechanisms of the antitumor responses and host-versus-graft reactions induced by recipient leukocyte infusions in mixed chimeras prepared with nonmyeloablative conditioning: a critical role for recipient CD4+ T cells and recipient leukocyte infusion-derived IFN-gamma-producing CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:665-76. [PMID: 16002661 DOI: 10.4049/jimmunol.175.2.665] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Surprisingly, antitumor responses can occur in patients who reject donor grafts following nonmyeloablative hemopoietic cell transplantation. In murine mixed chimeras prepared with nonmyeloablative conditioning, we previously showed that recipient leukocyte infusions (RLI) induced loss of donor chimerism, IFN-gamma production, and antitumor responses against host-type tumors. However, the mechanisms behind this phenomenon remain to be determined. We now demonstrate that the effects of RLI are mediated by distinct and complex mechanisms. Donor marrow rejection is induced by RLI-derived alloactivated T cells, which activate non-RLI-derived, recipient IFN-gamma-producing cells. RLI-derived CD8 T cells induce the production of IFN-gamma by both RLI and non-RLI-derived recipient cells. The antitumor responses of RLI involve mainly RLI-derived IFN-gamma-producing CD8 T cells and recipient-derived CD4 T cells and do not involve donor T cells. The pathways of donor marrow and tumor rejection lead to the development of tumor-specific cell-mediated cytotoxic responses that are not due to bystander killing by alloreactive T cells.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/transplantation
- Cell Line, Tumor
- Cytotoxicity, Immunologic/genetics
- Down-Regulation/genetics
- Down-Regulation/immunology
- Female
- Graft Rejection/genetics
- Graft Rejection/immunology
- Host vs Graft Reaction/genetics
- Host vs Graft Reaction/immunology
- Interferon-gamma/antagonists & inhibitors
- Interferon-gamma/biosynthesis
- Interferon-gamma/deficiency
- Leukemia, B-Cell/genetics
- Leukemia, B-Cell/immunology
- Leukemia, B-Cell/therapy
- Leukocyte Transfusion/methods
- Lymphocyte Activation/genetics
- Lymphocyte Depletion
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Radiation Chimera/immunology
- Transplantation Conditioning/methods
Collapse
Affiliation(s)
- Marie-Therese Rubio
- Transplantation Biology Research Center, Bone Marrow Transplantation Section, Massachusetts General Hospital, Harvard Medical School, 13th Street, Boston, MA 02129, USA
| | | | | | | | | | | |
Collapse
|
36
|
Kim S, Poursine-Laurent J, Truscott SM, Lybarger L, Song YJ, Yang L, French AR, Sunwoo JB, Lemieux S, Hansen TH, Yokoyama WM. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature 2005; 436:709-13. [PMID: 16079848 DOI: 10.1038/nature03847] [Citation(s) in RCA: 984] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2005] [Accepted: 05/19/2005] [Indexed: 11/08/2022]
Abstract
Self versus non-self discrimination is a central theme in biology from plants to vertebrates, and is particularly relevant for lymphocytes that express receptors capable of recognizing self-tissues and foreign invaders. Comprising the third largest lymphocyte population, natural killer (NK) cells recognize and kill cellular targets and produce pro-inflammatory cytokines. These potentially self-destructive effector functions can be controlled by inhibitory receptors for the polymorphic major histocompatibility complex (MHC) class I molecules that are ubiquitously expressed on target cells. However, inhibitory receptors are not uniformly expressed on NK cells, and are germline-encoded by a set of polymorphic genes that segregate independently from MHC genes. Therefore, how NK-cell self-tolerance arises in vivo is poorly understood. Here we demonstrate that NK cells acquire functional competence through 'licensing' by self-MHC molecules. Licensing involves a positive role for MHC-specific inhibitory receptors and requires the cytoplasmic inhibitory motif originally identified in effector responses. This process results in two types of self-tolerant NK cells--licensed or unlicensed--and may provide new insights for exploiting NK cells in immunotherapy. This self-tolerance mechanism may be more broadly applicable within the vertebrate immune system because related germline-encoded inhibitory receptors are widely expressed on other immune cells.
Collapse
Affiliation(s)
- Sungjin Kim
- Howard Hughes Medical Institute, Rheumatology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Haraguchi K, Takahashi T, Matsumoto A, Asai T, Kanda Y, Kurokawa M, Ogawa S, Oda H, Taniguchi M, Hirai H, Chiba S. Host-Residual Invariant NK T Cells Attenuate Graft-versus-Host Immunity. THE JOURNAL OF IMMUNOLOGY 2005; 175:1320-8. [PMID: 16002737 DOI: 10.4049/jimmunol.175.2.1320] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Invariant NK T (iNKT) cells have an invariant TCR-alpha chain and are activated in a CD1d-restricted manner. They are thought to regulate immune responses and play important roles in autoimmunity, allergy, infection, and tumor immunity. They also appear to influence immunity after hemopoietic stem cell transplantation. In this study, we examined the role of iNKT cells in graft-vs-host disease (GVHD) and graft rejection in a mouse model of MHC-mismatched bone marrow transplantation, using materials including alpha-galactosylceramide, NKT cells expanded in vitro, and Jalpha18 knockout mice that lack iNKT cells. We found that host-residual iNKT cells constitute effector cells which play a crucial role in reducing the severity of GVHD, and that this reduction is associated with a delayed increase in serum Th2 cytokine levels. Interestingly, we also found that host-residual iNKT cause a delay in engraftment and, under certain conditions, graft rejection. These results indicate that host-residual iNKT cells attenuate graft-vs-host immunity rather than host-vs-graft immunity.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Bone Marrow Transplantation/immunology
- Bone Marrow Transplantation/pathology
- Disease Models, Animal
- Galactosylceramides/pharmacology
- Graft Enhancement, Immunologic/methods
- Graft Rejection/genetics
- Graft Rejection/immunology
- Graft vs Host Disease/genetics
- Graft vs Host Disease/immunology
- Graft vs Host Disease/mortality
- Graft vs Host Disease/therapy
- Hot Temperature
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/transplantation
- Lymphocyte Depletion
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Antigen, T-Cell, alpha-beta/deficiency
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/physiology
- Survival Analysis
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
Collapse
Affiliation(s)
- Kyoko Haraguchi
- Departments of Hematology/Oncology, Cell Therapy/Transplantation Medicine, University of Tokyo Graduate School of Medicine and Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Dupont B, Hsu KC. Inhibitory killer Ig-like receptor genes and human leukocyte antigen class I ligands in haematopoietic stem cell transplantation. Curr Opin Immunol 2005; 16:634-43. [PMID: 15342011 DOI: 10.1016/j.coi.2004.07.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Identification of inhibitory killer Ig-like receptor (KIR) genes and their cognate human leukocyte antigen (HLA) class I ligands in donor-recipient pairs for patients undergoing haematopoietic stem cell transplantation (HCT) as treatment for haematopoietic malignancies has recently gained considerable interest. One incentive for these studies is to identify potential donors who are not HLA identical with the recipient but who still could serve as acceptable and potentially preferred donors based on their KIR genotype. It is demonstrated that a majority of individuals have at least one inhibitory KIR gene for which they lack the cognate HLA class I ligand. Therefore, the clinical benefits conferred by 'missing KIR ligand' might not be limited only to HLA mismatched donor-recipient combinations but may be applicable also to HLA identical transplants and even autologous HCT.
Collapse
Affiliation(s)
- Bo Dupont
- Immunology Program, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA.
| | | |
Collapse
|
39
|
Passweg JR, Stern M, Koehl U, Uharek L, Tichelli A. Use of natural killer cells in hematopoetic stem cell transplantation. Bone Marrow Transplant 2005; 35:637-43. [PMID: 15654351 DOI: 10.1038/sj.bmt.1704810] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Adoptive immunotherapy using natural killer (NK) cells may prove useful, especially in situations where infusion of T cells is impractical such as in recipients of haploidentical stem cell transplantation (HSCT) from haploidentical donors. NK cells may induce potent antileukemic and possibly antirejection activity and may even mitigate graft versus host disease (GvHD). Whether such effects are clinically important and whether they are mediated mainly or exclusively by KIR-HLA class I interactions remains to be determined. Recent advances in graft engineering provide for methods to isolate large numbers of purified NK cells. Several groups have shown that clinical grade NK cells up to a dose of 10(7)/kg may be collected and purified for the purpose of infusion to patients. Early results, in a limited number of patients, show that these cell doses may be administered without adverse events and without inducing GvHD. Whether such infusions will be useful in preventing graft rejection, or exerting graft versus leukemia effects and hastening immune recovery requires further study.
Collapse
Affiliation(s)
- J R Passweg
- The Basel Stem Cell Transplant Team, Basel University Hospitals, Switzerland.
| | | | | | | | | |
Collapse
|
40
|
Hanash AM, Levy RB. Donor CD4+CD25+ T cells promote engraftment and tolerance following MHC-mismatched hematopoietic cell transplantation. Blood 2004; 105:1828-36. [PMID: 15494429 DOI: 10.1182/blood-2004-08-3213] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Allogeneic bone marrow transplantation (BMT) is a potentially curative treatment for both inherited and acquired diseases of the hematopoietic compartment; however, its wider use is limited by the frequent and severe outcome of graft-versus-host disease (GVHD). Unfortunately, efforts to reduce GVHD by removing donor T cells have resulted in poor engraftment and elevated disease recurrence. Alternative cell populations capable of supporting allogeneic hematopoietic stem/progenitor cell engraftment without inducing GVHD could increase numbers of potential recipients while broadening the pool of acceptable donors. Although unfractionated CD4(+) T cells have not been shown to be an efficient facilitating population, CD4(+)CD25(+) regulatory cells (T-reg's) were examined for their capacity to support allogeneic hematopoietic engraftment. In a murine fully major histocompatibility complex (MHC)-mismatched BMT model, cotransplantation of donor B6 T-reg's into sublethally conditioned BALB/c recipients supported significantly greater lineage-committed and multipotential donor progenitors in recipient spleens 1 week after transplantation and significantly increased long-term multilineage donor chimerism. Donor engraftment occurred without GVHD-related weight loss or lethality and was associated with tolerance to donor and host antigens by in vitro and in vivo analyses. Donor CD4(+)CD25(+) T cells may therefore represent a potential alternative to unfractionated T cells for promotion of allogeneic engraftment in clinical hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Alan M Hanash
- University of Miami Miller School of Medicine, Department of Microbiology and Immunology, PO Box 016960, Miami, FL 33101, USA
| | | |
Collapse
|
41
|
Drukker M. Immunogenicity of human embryonic stem cells: can we achieve tolerance? ACTA ACUST UNITED AC 2004; 26:201-13. [PMID: 15549307 DOI: 10.1007/s00281-004-0163-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2004] [Accepted: 04/18/2004] [Indexed: 12/21/2022]
Abstract
Human embryonic stem cells are unique in their capacity to propagate without losing pluripotency, and at the same time may readily differentiate to various cell types of the three embryonic germ layers. It is widely accepted today that differentiated human embryonic stem cells may in the future enable repair of vital tissues of the body. Detailed differentiation protocols need to be developed and safety issues associated with cellular therapeutics must be examined. One of the greatest hurdles facing transplantation is the development of immune rejection processes towards non-autologous cells. Profiling of histocompatibility antigens expressed on the cells reveals that they might be subjected to immune response. Here we describe the routes of immune recognition that can identify these antigens and the proposed ways for overcoming the rejection of human embryonic stem cell derivatives.
Collapse
Affiliation(s)
- Micha Drukker
- Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, 91904 Jerusalem, Israel.
| |
Collapse
|