Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 652
Filter
2.
Front Immunol ; 12: 791206, 2021.
Article in English | MEDLINE | ID: mdl-34804076

ABSTRACT

Osteosarcoma, Ewing sarcoma (EWS), and rhabdomyosarcoma (RMS) are the most common pediatric sarcomas. Conventional therapy for these sarcomas comprises neoadjuvant and adjuvant chemotherapy, surgical resection of the primary tumor and/or radiation therapy. Patients with metastatic, relapsed, or refractory tumors have a dismal prognosis due to resistance to these conventional therapies. Therefore, innovative therapeutic interventions, such as immunotherapy, are urgently needed. Recently, cancer research has focused attention on natural killer (NK) cells due their innate ability to recognize and kill tumor cells. Osteosarcoma, EWS and RMS, are known to be sensitive to NK cell cytotoxicity in vitro. In the clinical setting however, NK cell cytotoxicity against sarcoma cells has been mainly studied in the context of allogeneic stem cell transplantation, where a rapid immune reconstitution of NK cells plays a key role in the control of the disease, known as graft-versus-tumor effect. In this review, we discuss the evidence for the current and future strategies to enhance the NK cell-versus-pediatric sarcoma effect, with a clinical focus. The different approaches encompass enhancing antibody-dependent NK cell cytotoxicity, counteracting the NK cell mechanisms of self-tolerance, and developing adoptive NK cell therapy including chimeric antigen receptor-expressing NK cells.


Subject(s)
Bone Neoplasms/immunology , Immunotherapy, Adoptive/methods , Immunotherapy/trends , Killer Cells, Natural/immunology , Osteosarcoma/immunology , Rhabdomyosarcoma/immunology , Sarcoma, Ewing/immunology , Animals , Antibodies/metabolism , Bone Neoplasms/therapy , Child , Cytotoxicity, Immunologic , Graft vs Tumor Effect , Humans , Lymphocyte Activation , Osteosarcoma/therapy , Rhabdomyosarcoma/therapy , Sarcoma, Ewing/therapy
3.
Front Immunol ; 12: 754316, 2021.
Article in English | MEDLINE | ID: mdl-34721430

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of myeloid progenitor cells that dampen overwhelming adaptive immune responses through multiple mechanisms and are recognized as an attractive novel immune intervention therapy for counteracting the destructive effects of graft- versus -host disease (GVHD) developing after allogeneic bone marrow transplantation (BMT). MDSCs can be produced in great numbers for cellular therapy, but they present a mixture of subsets whose functions in GVHD prevention are undefined. Here, we generated MDSCs in vitro from murine BM cells in the presence of GM-CSF and defined the integrin CD11c as a marker to subdivide MDSCs into two functional subgroups: CD11b+CD11c+ and CD11b+CD11c- MDSCs. Isolated CD11b+CD11c+ and CD11b+CD11c- MDSCs both inhibited alloantigen-stimulated T-cell proliferation in vitro, although CD11b+CD11c+ MDSCs were more efficient and expressed higher levels of different immunosuppressive molecules. Likewise, expression of surface markers such as MHC class II, CD80, CD86, or PD-L1 further delineated both subsets. Most importantly, only the adoptive transfer of CD11b+CD11c+ MDSCs into a single MHC class I-disparate allogeneic BMT model prevented GVHD development and strongly decreased disease-induced mortality, while CD11b+CD11c- MDSCs were totally ineffective. Surprisingly, allogeneic T-cell homing and expansion in lymphatic and GVHD target organs were not affected by cotransplanted CD11b+CD11c+ MDSCs indicating a clear contradiction between in vitro and in vivo functions of MDSCs. However, CD11b+CD11c+ MDSCs shifted immune responses towards type 2 immunity reflected by increased Th2-specific cytokine expression of allogeneic T cells. Induction of type 2 immunity was mandatory for GVHD prevention, since CD11b+CD11c+ MDSCs were ineffective if recipients were reconstituted with STAT6-deficient T cells unable to differentiate into Th2 cells. Most importantly, the beneficial graft- versus -tumor (GVT) effect was maintained in the presence of CD11b+CD11c+ MDSCs since syngeneic tumor cells were efficiently eradicated. Strong differences in the transcriptomic landscape of both subpopulations underlined their functional differences. Defining CD11b+CD11c+ MDSCs as the subset of in vitro-generated MDSCs able to inhibit GVHD development might help to increase efficiency of MDSC therapy and to further delineate relevant target molecules and signaling pathways responsible for GVHD prevention.


Subject(s)
CD11 Antigens/analysis , CD11b Antigen/analysis , Graft vs Host Disease/prevention & control , Myeloid-Derived Suppressor Cells/immunology , Allografts , Animals , Bone Marrow Transplantation/adverse effects , Cell Differentiation/drug effects , Cells, Cultured , Gene Ontology , Graft vs Tumor Effect , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunity, Cellular , Immunomagnetic Separation , Mice , Myeloid-Derived Suppressor Cells/chemistry , Myeloid-Derived Suppressor Cells/classification , Myeloid-Derived Suppressor Cells/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radiation Chimera , T-Lymphocyte Subsets/immunology , Transcriptome
4.
Front Immunol ; 12: 757836, 2021.
Article in English | MEDLINE | ID: mdl-34712243

ABSTRACT

The therapeutic efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT) is limited by the development of graft-versus-host disease (GVHD). In GVHD, rigorous pre-conditioning regimen resets the immune landscape and inflammatory milieu causing immune dysregulation, characterized by an expansion of alloreactive cells and a reduction in immune regulatory cells. In acute GVHD (aGVHD), the release of damage- and pathogen- associated molecular patterns from damaged tissue caused by the conditioning regimen sets the stage for T cell priming, activation and expansion further exacerbating tissue injury and organ damage, particularly in the gastrointestinal tract. Studies have shown that donor T cells utilize multiple energetic and biosynthetic pathways to mediate GVHD that can be distinct from the pathways used by regulatory T cells for their suppressive function. In chronic GVHD (cGVHD), donor T cells may differentiate into IL-21 producing T follicular helper cells or tissue resident T helper cells that cooperate with germinal center B cells or memory B cells, respectively, to produce allo- and auto-reactive antibodies with subsequent tissue fibrosis. Alternatively, donor T cells can become IFN- γ/IL-17 cytokine expressing T cells that mediate sclerodermatous skin injury. Patients refractory to the first line standard regimens for GVHD treatment have a poor prognosis indicating an urgent need for new therapies to restore the balance between effector and regulatory immune cells while preserving the beneficial graft-versus-tumor effect. Emerging data points toward a role for metabolism in regulating these allo- and auto-immune responses. Here, we will discuss the preclinical and clinical data available on the distinct metabolic demands of acute and chronic GVHD and recent efforts in identifying therapeutic targets using metabolomics. Another dimension of this review will examine the changing microbiome after allo-HSCT and the role of microbial metabolites such as short chain fatty acids and long chain fatty acids on regulating immune responses. Lastly, we will examine the metabolic implications of coinhibitory pathway blockade and cellular therapies in allo-HSCT. In conclusion, greater understanding of metabolic pathways involved in immune cell dysregulation during allo-HSCT may pave the way to provide novel therapies to prevent and treat GVHD.


Subject(s)
Graft vs Host Disease/therapy , Metabolomics/trends , Acute Disease , Amino Acids/metabolism , Chronic Disease , Dysbiosis/complications , Dysbiosis/immunology , Energy Metabolism , Fatty Acids/physiology , Gastrointestinal Microbiome/immunology , Glutamine/metabolism , Glycolysis , Graft vs Host Disease/metabolism , Graft vs Tumor Effect , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immune Checkpoint Proteins/physiology , Immunomodulation , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Metabolomics/methods , Reactive Oxygen Species , T-Lymphocyte Subsets/immunology , Transplantation Conditioning/adverse effects , Transplantation, Homologous/adverse effects , Vitamins/metabolism
5.
Front Immunol ; 12: 668307, 2021.
Article in English | MEDLINE | ID: mdl-34489927

ABSTRACT

Management for high-risk neuroblastoma (NBL) has included autologous hematopoietic stem cell transplant (HSCT) and anti-GD2 immunotherapy, but survival remains around 50%. The aim of this study was to determine if allogeneic HSCT could serve as a platform for inducing a graft-versus-tumor (GVT) effect against NBL with combination immunocytokine and NK cells in a murine model. Lethally irradiated C57BL/6 (B6) x A/J recipients were transplanted with B6 bone marrow on Day +0. On day +10, allogeneic HSCT recipients were challenged with NXS2, a GD2+ NBL. On days +14-16, mice were treated with the anti-GD2 immunocytokine hu14.18-IL2. In select groups, hu14.18-IL2 was combined with infusions of B6 NK cells activated with IL-15/IL-15Rα and CD137L ex vivo. Allogeneic HSCT alone was insufficient to control NXS2 tumor growth, but the addition of hu14.18-IL2 controlled tumor growth and improved survival. Adoptive transfer of ex vivo CD137L/IL-15/IL-15Rα activated NK cells with or without hu14.18-IL2 exacerbated lethality. CD137L/IL-15/IL-15Rα activated NK cells showed enhanced cytotoxicity and produced high levels of TNF-α in vitro, but induced cytokine release syndrome (CRS) in vivo. Infusing Perforin-/- CD137L/IL-15/IL-15Rα activated NK cells had no impact on GVT, whereas TNF-α-/- CD137L/IL-15/IL-15Rα activated NK cells improved GVT by decreasing peripheral effector cell subsets while preserving tumor-infiltrating lymphocytes. Depletion of Ly49H+ NK cells also improved GVT. Using allogeneic HSCT for NBL is a viable platform for immunocytokines and ex vivo activated NK cell infusions, but must be balanced with induction of CRS. Regulation of TNFα or activating NK subsets may be needed to improve GVT effects.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Cytokines/pharmacology , Gangliosides/antagonists & inhibitors , Graft vs Tumor Effect , Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Killer Cells, Natural/drug effects , Killer Cells, Natural/transplantation , Lymphocyte Activation/drug effects , Neuroblastoma/therapy , Animals , Cell Line, Tumor , Combined Modality Therapy , Female , Gangliosides/immunology , Gangliosides/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neuroblastoma/immunology , Neuroblastoma/metabolism , Neuroblastoma/pathology
6.
Blood ; 138(24): 2583-2588, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34424962

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains a potential curative option for treating a variety of hematologic diseases, but acute and chronic graft-versus-host disease (GVHD) remain major barriers limiting efficacy. Acute gut GVHD occurs with marked increases in proinflammatory cytokines (including TNF and IL-6), which we recently demonstrated was exacerbated in obesity resulting in severe gastrointestinal pathology. Given the pleiotropic and overlapping effects of these 2 cytokines, we assessed the impact of dual TNF and IL-6R blockade on GVHD as well as graft-versus tumor (GVT) effects in different mouse GVHD models. Early administration of combined blockade resulted in greater protection and survival from acute gut GVHD compared with single blockade regimens and even development of later chronic skin GVHD. Importantly, double cytokine blockade preserved GVT effects reinforcing that GVT and GVHD can be delineated and may result in greater efficacy in allo-HSCT.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Receptors, Interleukin-6/antagonists & inhibitors , Tumor Necrosis Factor Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Disease Models, Animal , Etanercept/therapeutic use , Female , Graft vs Tumor Effect/drug effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous/methods
7.
Front Immunol ; 12: 647894, 2021.
Article in English | MEDLINE | ID: mdl-34262560

ABSTRACT

Acute graft-versus-host disease (aGVHD) is a lethal complication after allogeneic hematopoietic stem cell transplantation. The mechanism involves the recognition of host antigens by donor-derived T cells which induces augmented response of alloreactive T cells. In this study, we characterized the role of a previously identified novel classical secretory protein with antitumor function-LYG1 (Lysozyme G-like 1), in aGVHD. LYG1 deficiency reduced the activation of CD4+ T cells and Th1 ratio, but increased Treg ratio in vitro by MLR assay. By using major MHC mismatched aGVHD model, LYG1 deficiency in donor T cells or CD4+ T cells attenuated aGVHD severity, inhibited CD4+ T cells activation and IFN-γ expression, promoted FoxP3 expression, suppressed CXCL9 and CXCL10 expression, restrained allogeneic CD4+ T cells infiltrating in target organs. The function of LYG1 in aGVHD was also confirmed using haploidentical transplant model. Furthermore, administration of recombinant human LYG1 protein intraperitoneally aggravated aGVHD by promoting IFN-γ production and inhibiting FoxP3 expression. The effect of rhLYG1 could partially be abrogated with the absence of IFN-γ. Furthermore, LYG1 deficiency in donor T cells preserved graft-versus-tumor response. In summary, our results indicate LYG1 regulates aGVHD by the alloreactivity of CD4+ T cells and the balance of Th1 and Treg differentiation of allogeneic CD4+ T cells, targeting LYG1 maybe a novel therapeutic strategy for preventing aGVHD.


Subject(s)
Allografts/immunology , Graft vs Host Disease/immunology , Graft vs Tumor Effect/immunology , Muramidase/deficiency , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Cell Polarity/genetics , Cell Polarity/immunology , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Tumor Effect/genetics , Hematopoietic Stem Cell Transplantation , Humans , Interferon-gamma/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Muramidase/genetics , Recombinant Proteins/administration & dosage , Signal Transduction/genetics , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous/methods
8.
Blood ; 138(10): 858-870, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34036317

ABSTRACT

Invariant natural killer T (iNKT) cells are a T-cell subset with potent immunomodulatory properties. Experimental evidence in mice and observational studies in humans indicate that iNKT cells have antitumor potential as well as the ability to suppress acute and chronic graft-versus-host-disease (GVHD). Murine iNKT cells differentiate during thymic development into iNKT1, iNKT2, and iNKT17 sublineages, which differ transcriptomically and epigenomically and have subset-specific developmental requirements. Whether distinct iNKT sublineages also differ in their antitumor effect and their ability to suppress GVHD is currently unknown. In this work, we generated highly purified murine iNKT sublineages, characterized their transcriptomic and epigenomic landscape, and assessed specific functions. We show that iNKT2 and iNKT17, but not iNKT1, cells efficiently suppress T-cell activation in vitro and mitigate murine acute GVHD in vivo. Conversely, we show that iNKT1 cells display the highest antitumor activity against murine B-cell lymphoma cells both in vitro and in vivo. Thus, we report for the first time that iNKT sublineages have distinct and different functions, with iNKT1 cells having the highest antitumor activity and iNKT2 and iNKT17 cells having immune-regulatory properties. These results have important implications for the translation of iNKT cell therapies to the clinic for cancer immunotherapy as well as for the prevention and treatment of GVHD.


Subject(s)
Graft vs Host Disease , Graft vs Tumor Effect/immunology , Lymphocyte Activation , Lymphoma, B-Cell , Natural Killer T-Cells/immunology , Neoplasms, Experimental , Animals , Epigenomics , Female , Gene Expression Profiling , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Male , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy
9.
Front Immunol ; 12: 651288, 2021.
Article in English | MEDLINE | ID: mdl-33777050

ABSTRACT

The incidence of multiple myeloma (MM), a bone marrow (BM) resident hematological malignancy, is increasing globally. The disease has substantial morbidity and mortality and remains largely incurable. Clinical studies show that autologous stem cell transplantation (ASCT) remains efficacious in eligible patients, providing a progression free survival (PFS) benefit beyond novel therapies alone. Conventionally, improved PFS after ASCT is attributed to cytoreduction from myeloablative chemotherapy. However, ASCT results in immune effects beyond cytoreduction, including inflammation, lymphodepletion, T cell priming via immunogenic cell death, and disruption of the tumor BM microenvironment. In fact, a small subset of patients achieve very long-term control of disease post-ASCT, akin to that seen in the context of immune-mediated graft-vs.-myeloma effects after allogeneic SCT. These clinical observations coupled with recent definitive studies in mice demonstrating that progression after ASCT represents immune escape as a consequence of T cell exhaustion, highlight the potential for new immunotherapy maintenance strategies to prevent myeloma progression following consolidation with ASCT.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive/methods , Multiple Myeloma/therapy , Myeloablative Agonists/therapeutic use , Animals , Combined Modality Therapy/methods , Disease Models, Animal , Disease Progression , Graft vs Tumor Effect/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunogenic Cell Death , Melphalan/therapeutic use , Mice , Multiple Myeloma/immunology , Multiple Myeloma/mortality , Myeloablative Agonists/pharmacology , Progression-Free Survival , Receptors, Chimeric Antigen/immunology , Transplantation, Autologous , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
10.
J Immunol ; 206(4): 892-903, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33408257

ABSTRACT

Donor-derived lymphocytes from allogeneic hematopoietic cell transplantation (allo-HCT) or donor lymphocyte infusion can mediate eradication of host tumor cells in a process labeled the graft-versus-tumor (GVT) effect. Unfortunately, these treatments have produced limited results in various types of leukemia because of an insufficient GVT effect. In this context, molecular engineering of donor lymphocytes to increase the GVT effect may benefit cancer patients. Activating MyD88 signaling in CD8+ T cells via TLR enhances T cell activation and cytotoxicity. However, systemic administration of TLR ligands to stimulate MyD88 could induce hyperinflammation or elicit protumor effects. To circumvent this problem, we devised a synthetic molecule consisting of MyD88 linked to the ectopic domain of CD8a (CD8α:MyD88). We used this construct to test the hypothesis that MyD88 costimulation in donor CD8+ T cells increases tumor control following allo-HCT in mice by increasing T cell activation, function, and direct tumor cytotoxicity. Indeed, an increase in both in vitro and in vivo tumor control was observed with CD8α:MyD88 T cells. This increase in the GVT response was associated with increased T cell expansion, increased functional capacity, and an increase in direct cytotoxic killing of the tumor cells. However, MyD88 costimulation in donor CD8+ T cells was linked to increased yet nonlethal graft-versus-host disease in mice treated with these engineered CD8+ T cells. Given these observations, synthetic CD8α:MyD88 donor T cells may represent a unique and versatile approach to enhance the GVT response that merits further refinement to improve the effectiveness of allo-HCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia , Animals , CD8-Positive T-Lymphocytes , Graft vs Tumor Effect , Humans , Mice , Myeloid Differentiation Factor 88 , Transplantation, Homologous
11.
Front Immunol ; 11: 1517, 2020.
Article in English | MEDLINE | ID: mdl-32793207

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (aHSCT) is a curative therapy for a range of hematologic illnesses including aplastic anemia, sickle cell disease, immunodeficiency, and high-risk leukemia, but the efficacy of aHSCT is often undermined by graft-versus-host disease (GVHD), where T cells from the donor attack and destroy recipient tissues. Given the strong interconnection between T cell metabolism and cellular function, determining the metabolic pathways utilized by alloreactive T cells is fundamental to deepening our understanding of GVHD biology, including its initiation, propagation, and potential mitigation. This review summarizes the metabolic pathways available to alloreactive T cells and highlights key metabolic proteins and pathways linking T cell metabolism to effector function. Our current knowledge of alloreactive T cell metabolism is then explored, showing support for glycolysis, fat oxidation, and glutamine metabolism but also offering a potential explanation for how these presumably contradictory metabolic findings might be reconciled. Examples of additional ways in which metabolism impacts aHSCT are addressed, including the influence of butyrate metabolism on GVHD resolution. Finally, the caveats and challenges of assigning causality using our current metabolic toolbox is discussed, as well as likely future directions in immunometabolism, both to highlight the strengths of the current evidence as well as recognize some of its limitations.


Subject(s)
Isoantigens/immunology , Metabolic Networks and Pathways , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Energy Metabolism , Graft vs Host Disease/etiology , Graft vs Tumor Effect/immunology , Humans , Immunity, Cellular , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
12.
Blood ; 136(22): 2491-2497, 2020 11 26.
Article in English | MEDLINE | ID: mdl-32735639

ABSTRACT

The treatment of multiple myeloma (MM) is currently being redefined by humoral and cellular immunotherapies. For decades, there was limited belief in immune-based anti-MM therapy as a result of the moderate graft-versus-myeloma effect of allogeneic stem cell transplantation. Today, monoclonal antibodies comprise the new backbone of anti-MM therapy, and T-cell therapies targeting BCMA are emerging as the most potent single agents for MM treatment. Herein, we present our assessment of and vision for MM immunotherapy in the short and midterm.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Graft vs Tumor Effect/immunology , Immunotherapy/trends , Multiple Myeloma/therapy , Stem Cell Transplantation/trends , Allografts , Humans , Multiple Myeloma/immunology
13.
Front Immunol ; 11: 1068, 2020.
Article in English | MEDLINE | ID: mdl-32547559

ABSTRACT

Mature immunocompetent cells from the stem cell graft as well as early robust immune reconstitution are essential for the graft-vs. -tumor (GVT) effect to eliminate residual malignant cells after allogeneic hematopoietic stem cell transplantation (HSCT). In this prospective study we characterized graft composition of T- and NK cell subsets in 88 recipients of peripheral blood stem cell grafts with multicolor flowcytometry. Our primary aim was to analyze the impact of graft composition on immune reconstitution and clinical outcomes after transplantation. Patients transplanted with graft NK cell doses above the median value of 27 × 106/kg had significantly increased relapse-free-survival compared to patients transplanted with lower doses, HR 2.12 (95% CI 1.01-4.45, p = 0.04) Peripheral blood concentrations of NK cells obtained from donors before G-CSF mobilization were significantly correlated to graft NK cell doses (Spearman's ρ 0.53, p = 0.03). The dose of transplanted NK cells/kg correlated significantly with NK cell concentrations in patients early after transplantation (Spearman's ρ 0.26, p = 0.02, and ρ = 0.35, p = 0.001 for days 28 and 56, respectively). Early immune reconstitution above median values of NK cells was significantly associated with improved relapse-free survival (HR 2.84 [95% CI 1.29-6.28], p = 0.01, and HR 4.19 [95% CI 1.68-10.4], p = 0.002, for day 28 and 56, respectively). Early concentrations above the median value of the mature effector CD56dim NK cell subset were significantly associated with decreased relapse incidences at 1 year, 7% (95% CI 1.8-17) vs. 28% (95% CI 15-42), p = 0.04, and 7% (95% CI 1.8-18) vs. 26% (95% CI 14-40) %, p = 0.03, for days 28 and 56, respectively. The results suggest a protective effect of high doses of NK cells in grafts and during early immune reconstitution and support the perception of NK cells as innate effector cells with anti-tumor effects in the setting of allogeneic stem cell transplantation.


Subject(s)
Allografts/immunology , Graft vs Tumor Effect/immunology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Adult , Aged , CD56 Antigen/metabolism , Female , Hematologic Neoplasms/mortality , Hematopoietic Stem Cell Mobilization , Humans , Male , Middle Aged , Prospective Studies , Survival Analysis , Transplantation, Homologous , Young Adult
14.
Front Immunol ; 11: 989, 2020.
Article in English | MEDLINE | ID: mdl-32528476

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are innate immune cells that acquire the capacity to suppress adaptive immune responses. In the context of allogeneic hematopoietic stem cell transplantation (allo-HSCT), MDSCs (in the donor graft and in the recipient, after allo-HSCT) might mediate immune suppression through multiple mechanisms. However, it remains unclear how MDSCs can be distinguished from their normal myeloid counterparts in the hematopoietic stem cell donor graft and during immune reconstitution after allo-HSCT in the recipient. Our ability to understand their exact role in allo-HSCT is limited by the absence of a specific gene signature or surface markers for identifying MDSCs among myeloid cells and by their plasticity in different microenvironments. According to various studies, MDSCs might induce transplant tolerance and control graft vs. host disease (GVHD), but their impact on the graft vs. tumor effect (GVT) is not fully understood. In fact, we know that MDSCs commonly expand in patients with cancer, and they are thought to promote hematological malignancy progression. However, little is known about whether depleting them might be an effective strategy for enhancing GVT effects. Here, we review data published over the past 40 years on allo-HSCT to delineate the different MDSC subsets, and their abilities to induce transplant tolerance and preserve the GVT effect. This review will provide a basis for determining whether one MDSC subset might be proposed as the most appropriate candidate for cellular therapies, due to its ability to modulate GVHD.


Subject(s)
Graft Rejection/prevention & control , Graft Survival , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Myeloid-Derived Suppressor Cells/immunology , Transplantation Tolerance , Animals , Graft Rejection/immunology , Graft Rejection/metabolism , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Graft vs Tumor Effect , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Myeloid-Derived Suppressor Cells/metabolism , Phenotype , Risk Factors , Transplantation, Homologous , Treatment Outcome
15.
Front Immunol ; 11: 722, 2020.
Article in English | MEDLINE | ID: mdl-32411139

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-HCT) holds curative potential for many hematological disorders. However, the pathophysiology of the desired graft-versus-tumor effect is linked to life-threatening complications of acute graft-versus-host disease (GVHD). Allogeneic donor T lymphocytes are essential for causing GVHD, and their activation relies on the coordination of TCR engagement and co-stimulation, also known as Signal 1 and Signal 2. In addition to these signals, a network of secreted cytokines by immune cells provides a third signal, Signal 3, that is critical for the initiation and maintenance of GVHD. Strategies to target Signal 3 in human diseases have shown therapeutic benefit for inflammatory disorders such as Rheumatoid Arthritis and Inflammatory Bowel Disease. However, despite our growing understanding of their role in GVHD, the success of targeting individual cytokines has been modest with some notable exceptions. This review aims to describe current approaches toward targeting Signal 3 in clinical GVHD, and to highlight emerging studies in immune cell biology that may be harnessed for better clinical translation.


Subject(s)
Cytokines/metabolism , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Cytokines/antagonists & inhibitors , Graft vs Tumor Effect/immunology , Humans , Molecular Targeted Therapy/methods , Receptors, Antigen, T-Cell/immunology , Transplantation, Homologous/adverse effects
16.
Front Immunol ; 11: 673, 2020.
Article in English | MEDLINE | ID: mdl-32373125

ABSTRACT

Allogeneic hematopoietic cell transplantation (HCT) remains the only curative therapy for many hematological malignant and non-malignant disorders. However, key obstacles to the success of HCT include graft-versus-host disease (GVHD) and disease relapse due to absence of graft-versus-tumor (GVT) effect. Over the last decade, advances in "omics" technologies and systems biology analysis, have allowed for the discovery and validation of blood biomarkers that can be used as diagnostic test and prognostic test (that risk-stratify patients before disease occurrence) for acute and chronic GVHD and recently GVT. There are also predictive biomarkers that categorize patients based on their likely to respond to therapy. Newer mathematical analysis such as machine learning is able to identify different predictors of GVHD using clinical characteristics pre-transplant and possibly in the future combined with other biomarkers. Biomarkers are not only useful to identify patients with higher risk of disease progression, but also help guide treatment decisions and/or provide a basis for specific therapeutic interventions. This review summarizes biomarkers definition, omics technologies, acute, chronic GVHD and GVT biomarkers currently used in clinic or with potential as targets for existing or new drugs focusing on novel published work.


Subject(s)
Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Animals , Biomarkers/blood , Graft vs Host Disease/blood , Graft vs Tumor Effect , Humans , Prognosis , Transplantation, Homologous/methods , Treatment Outcome
17.
Nat Rev Clin Oncol ; 17(8): 475-492, 2020 08.
Article in English | MEDLINE | ID: mdl-32313224

ABSTRACT

Allogeneic haematopoietic stem cell transplantation (allo-HSCT) was the first successful therapy for patients with haematological malignancies, predominantly owing to graft-versus-tumour (GvT) effects. Dramatic methodological changes, designed to expand eligibility for allo-HSCT to older patients and/or those with comorbidities, have led to the use of reduced-intensity conditioning regimens, in parallel with more aggressive immunosuppression to better control graft-versus-host disease (GvHD). Consequently, disease relapse has become the major cause of death following allo-HSCT. Hence, the prevention and treatment of relapse has come to the forefront and remains an unmet medical need. Despite >60 years of preclinical and clinical studies, the immunological requirements necessary to achieve GvT effects without promoting GvHD have not been fully established. Herein, we review learnings from preclinical modelling and clinical studies relating to the GvT effect, focusing on mechanisms of relapse and on immunomodulatory strategies that are being developed to overcome disease recurrence after both allo-HSCT and autologous HSCT. Emphasis is placed on discussing current knowledge and approaches predicated on the use of cell therapies, cytokines to augment immune responses and dual-purpose antibody therapies or other pharmacological agents that can control GvHD whilst simultaneously targeting cancer cells.


Subject(s)
Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/immunology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Hematologic Neoplasms/complications , Hematologic Neoplasms/immunology , Hematopoietic Stem Cell Transplantation/trends , Humans , Immunosuppression Therapy/methods , Transplantation, Homologous/adverse effects
18.
Hematol Oncol ; 38(1): 89-102, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31465552

ABSTRACT

Graft-versus-host disease (GVHD) remains a major complication following allogeneic haematopoietic stem cell transplantation (allo-HSCT) leading to high transplant-related mortality. Natural killer (NK) cells have been found to mitigate GVHD without attenuating the graft-versus-tumour (GVT) activity in the murine model of haematopoietic stem cell transplantation. Sphingosine-1-phosphate receptor 5 (S1PR5) is a very important chemokine receptor on NK cells that governs NK cell distribution in vivo and trafficking at lesion sites. Our preliminary studies showed that the incidence of GVHD was negatively correlated with S1PR5 expression in the NK cells of patients after allo-HSCT. In the present study, we found that S1PR5 deficiency in murine NK cells blocked the migration of NK cells from the bone marrow to the GVHD target organs and attenuated the inhibitory effects on the alloreactive T cells, especially CD3+ CD8+ T cells, which may be the reason why the loss of S1PR5 in NK cells could aggravate GVHD in recipient mice. Furthermore, we also demonstrated that the absence of S1PR5 expression in NK cells did not interfere with the antitumour effects of NK cells and T cells in vivo. Taken together, our data indicate that S1PR5 plays an essential role in balancing GVHD and GVT activity.


Subject(s)
Disease Models, Animal , Graft vs Host Disease/prevention & control , Graft vs Tumor Effect , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/immunology , Receptors, Lysosphingolipid/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Lysosphingolipid/genetics , Transplantation, Homologous
19.
Crit Rev Immunol ; 39(2): 93-103, 2019.
Article in English | MEDLINE | ID: mdl-31679250

ABSTRACT

Communication between the nervous and immune systems is required for the body to regulate physiological homeostasis. Beta-adrenergic receptors expressed on immune cells mediate the modulation of immune response by neural activity. Activation of beta-adrenergic signaling results in suppression of antitumor immune response and limits the efficacy of cancer immunotherapy. Beta-adrenergic signaling is also involved in regulation of hematopoietic reconstitution, which is critical to the graft-versus-tumor (GVT) effect and to graft-versus-host disease (GVHD) following allogeneic hematopoietic cell transplantation (HCT). In this review, the function of beta-adrenergic signaling in mediating tumor immunosuppression will be highlighted. We will also discuss the implication of targeting beta-adrenergic signaling to improve the efficacy of cancer immunotherapy including the GVT effect, and to diminish the adverse effects including GVHD.


Subject(s)
Graft vs Tumor Effect/physiology , Hematopoietic Stem Cell Transplantation/methods , Immunotherapy/methods , Neoplasms/immunology , Receptors, Adrenergic, beta/metabolism , Animals , Humans , Immune Tolerance , Neoplasms/therapy , Neuroimmunomodulation , Signal Transduction , Tumor Escape , Tumor Microenvironment
20.
Liver Transpl ; 25(8): 1251-1264, 2019 08.
Article in English | MEDLINE | ID: mdl-31152624

ABSTRACT

Liver transplantation (LT) is currently considered an important method in treating hepatocellular carcinoma (HCC) and an alternative treatment for other liver malignancies. Here, we demonstrated that the graft-versus-tumor (GVT) effect exists in allogeneic liver transplantation (allo LT). Recipient-derived T cells played a critical role in the GVT process of allo LT, as demonstrated by extensive infiltration and significant activation of recipient T cells in the tumor after surgery. Moreover, this process was related to donor-derived T/B cells by improving the immune microenvironment in the tumor, as demonstrated by elevated levels of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), interleukin-2 (IL-2), IL-6, IL-16, chemokine (C-X-C motif) ligand 10 (CXCL10), and CXCL11 and decreased levels of IL-10 and IL-4 at tumor sites. Additionally, tacrolimus (FK506) treatment inhibited the GVT effect on allo LT. Donor liver-derived T/B cells infiltrate extrahepatic tumors to trigger a strong T-cell-mediated immune response and thus improve the tumor immune microenvironment.


Subject(s)
Carcinoma, Hepatocellular/surgery , Graft vs Tumor Effect/immunology , Liver Neoplasms/surgery , Liver Transplantation , Tumor Microenvironment/immunology , Allografts/immunology , Animals , Carcinoma, Hepatocellular/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Graft vs Tumor Effect/drug effects , Humans , Immunosuppressive Agents/administration & dosage , Isoantigens/immunology , Liver/immunology , Liver Neoplasms/immunology , Male , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tacrolimus/administration & dosage , Transplantation, Homologous , Tumor Microenvironment/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...