Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Publication year range
1.
Best Pract Res Clin Haematol ; 37(2): 101555, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39098803

ABSTRACT

Allogeneic hematopoietic cell transplantation (alloHCT) provides a potential curative treatment for haematological malignancies. The therapeutic Graft-versus-Leukaemia (GvL) effect is induced by donor T cells attacking patient hematopoietic (malignant) cells. However, if healthy non-hematopoietic tissues are targeted, Graft-versus-Disease (GvHD) may develop. After HLA-matched alloHCT, GvL and GvHD are induced by donor T cells recognizing polymorphic peptides presented by HLA on patient cells, so-called minor histocompatibility antigens (MiHAs). The balance between GvL and GvHD depends on the tissue distribution of MiHAs and T-cell frequencies targeting these MiHAs. T cells against broadly expressed MiHAs induce GvL and GvHD, whereas those targeting MiHAs with hematopoietic-restricted expression induce GvL without GvHD. Recently, the MiHA repertoire identified in natural immune responses after alloHCT was expanded to 159 total HLA-I-restricted MiHAs, including 14 hematopoietic-restricted MiHAs. This review explores their potential relevance to predict, monitor, and manipulate GvL and GvHD for improving clinical outcome after HLA-matched alloHCT.


Subject(s)
Graft vs Host Disease , Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation , Minor Histocompatibility Antigens , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Graft vs Host Disease/immunology , Minor Histocompatibility Antigens/immunology , Minor Histocompatibility Antigens/genetics , Graft vs Leukemia Effect/immunology , Transplantation, Homologous , Hematologic Neoplasms/therapy , Hematologic Neoplasms/immunology , T-Lymphocytes/immunology , Allografts
2.
J Immunol ; 213(3): 384-393, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38864663

ABSTRACT

Allogeneic hematopoietic cell transplantation is an effective treatment for hematologic malignancies, but the complications such as graft-versus-host disease (GVHD) can limit its benefit. The conditioning regimens before transplant, including chemotherapy or irradiation, can trigger endoplasmic reticulum stress. IRE-1α is a major endoplasmic reticulum stress mediator that can further activate both spliced XBP-1 (XBP-1s) and regulated IRE-1-dependent decay (RIDD). IRE-1α-XBP-1s signaling controls dendritic cell (DC) differentiation and Ag presentation, crucial in GVHD progression. In this study, we used DC-specific XBP-1-deficient mice as donors or recipients and observed that XBP-1s was crucial for host DCs in the induction of GVHD but dispensable for the graft-versus-leukemia response. To specifically target IRE-1α in the host, we treated recipient mice with the IRE-1α inhibitor B-I09 for 3 d prior to bone marrow transplantation, which significantly suppressed GVHD development while maintaining the graft-versus-leukemia effect. XBP-1-deficient or BI09-treated recipients showed reduced DC survival after irradiation and bone marrow transplantation. Inhibition of IRE-1α also led to a reduction in DC alloreactivity, subsequently decreasing the proliferation and activation of allogeneic T cells. With further study using RIDD-deficient DCs, we observed that RIDD was also required for optimal DC activation. Taken together, XBP-1s and RIDD both promote host DC survival and alloreactivity that contribute to GVHD development.


Subject(s)
Dendritic Cells , Endoplasmic Reticulum Stress , Endoribonucleases , Graft vs Host Disease , Protein Serine-Threonine Kinases , X-Box Binding Protein 1 , Animals , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Mice , Endoplasmic Reticulum Stress/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Endoribonucleases/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , Mice, Knockout , Mice, Inbred C57BL , Hematopoietic Stem Cell Transplantation , Bone Marrow Transplantation , Signal Transduction , Cell Differentiation/immunology , Graft vs Leukemia Effect/immunology
3.
J Clin Invest ; 134(16)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38916965

ABSTRACT

Leukemia relapse is a major cause of death after allogeneic hematopoietic cell transplantation (allo-HCT). We tested the potential of targeting T cell (Tc) immunoglobulin and mucin-containing molecule 3 (TIM-3) for improving graft-versus-leukemia (GVL) effects. We observed differential expression of TIM-3 ligands when hematopoietic stem cells overexpressed certain oncogenic-driver mutations. Anti-TIM-3 Ab treatment improved survival of mice bearing leukemia with oncogene-induced TIM-3 ligand expression. Conversely, leukemia cells with low ligand expression were anti-TIM-3 treatment resistant. In vitro, TIM-3 blockade or genetic deletion in CD8+ Tc enhanced Tc activation, proliferation, and IFN-γ production while enhancing GVL effects, preventing Tc exhaustion, and improving Tc cytotoxicity and glycolysis in vivo. Conversely, TIM-3 deletion in myeloid cells did not affect allogeneic Tc proliferation and activation in vitro, suggesting that anti-TIM-3 treatment-mediated GVL effects are Tc induced. In contrast to anti-programmed cell death protein 1 (anti-PD-1) and anti-cytotoxic T lymphocyte-associated protein 4 (anti-CTLA-4) treatment, anti-TIM-3-treatment did not enhance acute graft-versus-host disease (aGVHD). TIM-3 and its ligands were frequently expressed in acute myeloid leukemia (AML) cells of patients with post-allo-HCT relapse. We decipher the connections between oncogenic mutations found in AML and TIM-3 ligand expression and identify anti-TIM-3 treatment as a strategy for enhancing GVL effects via metabolic and transcriptional Tc reprogramming without exacerbation of aGVHD. Our findings support clinical testing of anti-TIM-3 Ab in patients with AML relapse after allo-HCT.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Animals , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Mice , Hematopoietic Stem Cell Transplantation , Graft vs Leukemia Effect/immunology , Graft vs Leukemia Effect/genetics , Humans , Allografts , Ligands , Oncogenes , CD8-Positive T-Lymphocytes/immunology , Mice, Knockout , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/pathology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , CTLA-4 Antigen/antagonists & inhibitors , Gene Expression Regulation, Leukemic
4.
Front Immunol ; 15: 1339318, 2024.
Article in English | MEDLINE | ID: mdl-38711496

ABSTRACT

Allogeneic Hematopoietic Stem Cell Transplantation (allo-HSCT) is the only curative therapy for many hematologic malignancies, whereby the Graft-versus-Leukemia (GVL) effect plays a pivotal role in controlling relapse. However, the success of GVL is hindered by Graft-versus-Host Disease (GVHD), where donor T cells attack healthy tissues in the recipient. The ability of natural regulatory T cells (Treg) to suppress immune responses has been exploited as a therapeutical option against GVHD. Still, it is crucial to evaluate if the ability of Treg to suppress GVHD does not compromise the benefits of GVL. Initial studies in animal models suggest that Treg can attenuate GVHD while preserving GVL, but results vary according to tumor type. Human trials using Treg as GVHD prophylaxis or treatment show promising results, emphasizing the importance of infusion timing and Treg/Tcon ratios. In this review, we discuss strategies that can be used aiming to enhance GVL post-Treg infusion and the proposed mechanisms for the maintenance of the GVL effect upon the adoptive Treg transfer. In order to optimize the therapeutic outcomes of Treg administration in allo-HSCT, future efforts should focus on refining Treg sources for infusion and evaluating their specificity for antigens mediating GVHD while preserving GVL responses.


Subject(s)
Graft vs Host Disease , Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , Humans , Graft vs Leukemia Effect/immunology , Animals , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Transplantation, Homologous , Adoptive Transfer/methods , Hematologic Neoplasms/therapy , Hematologic Neoplasms/immunology
5.
Front Immunol ; 15: 1358668, 2024.
Article in English | MEDLINE | ID: mdl-38817602

ABSTRACT

Allogeneic hematopoietic cell transplantation (HCT) has transformed over the past several decades through enhanced supportive care, reduced intensity conditioning (RIC), improved human leukocyte antigen (HLA) typing, and novel graft-versus-host disease (GVHD)-prevention and treatment strategies. Most notably, the implementation of post-transplantation cyclophosphamide (PTCy) has dramatically increased the safety and availability of this life-saving therapy. Given reductions in nonrelapse mortality (NRM) with these advances, the HCT community has placed even greater emphasis on developing ways to reduce relapse - the leading cause of death after HCT. When using RIC HCT, protection from relapse relies predominantly on graft-versus-leukemia (GVL) reactions. Donor lymphocyte infusion (DLI), adoptive cellular therapy, checkpoint inhibition, and post-HCT maintenance strategies represent approaches under study that aim to augment or synergize with the GVL effects of HCT. Optimizing donor selection algorithms to leverage GVL represents another active area of research. Many of these strategies seek to harness the effects of T cells, which for decades were felt to be the primary mediators of GVL and the focus of investigation in relapse reduction. However, there is growing interest in capitalizing on the ability of natural killer (NK) cells to yield potent anti-tumor effects. A potential advantage of NK cell-based approaches over T cell-mediated is the potential to reduce NRM in addition to relapse. By decreasing infection, without increasing the risk of GVHD, NK cells may mitigate NRM, while still yielding relapse reduction through identification and clearance of cancer cells. Most T cell-focused relapse-prevention strategies must weigh the benefits of relapse reduction against the increased risk of NRM from GVHD. In contrast, NK cells have the potential to reduce both, potentially tipping the scales significantly in favor of survival. Here, we will review the role of NK cells in GVL, optimization of NK cell match or mismatch, and burgeoning areas of research in NK cell therapy such as adoptive transfer and chimeric antigen receptor (CAR) NK cells.


Subject(s)
Graft vs Host Disease , Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural , Transplantation, Homologous , Humans , Killer Cells, Natural/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Graft vs Leukemia Effect/immunology , Graft vs Host Disease/prevention & control , Graft vs Host Disease/immunology , Graft vs Host Disease/etiology , Transplantation Conditioning/methods , Animals
6.
Rinsho Ketsueki ; 65(4): 265-271, 2024.
Article in Japanese | MEDLINE | ID: mdl-38684437

ABSTRACT

Hematopoietic cell transplantation (HCT) is considered a curative treatment for hematological malignancies. However, HCT recipients often face complications such as graft-versus-host disease (GVHD) and disease relapse. Clinical factors like age and HLA disparity are recognized as risks for GVHD. Notably, sex-mismatched HCT, particularly with female donors and male recipients (F→M), is reported to increase the risk of chronic GVHD. This adverse effect of F→M HCT is thought to result from allogeneic immune response against minor histocompatibility antigens encoded on the Y-chromosome of a male recipient (HY-antigens). Indeed, antibodies against HY-antigens (HY-Abs) were detected three months after F→M HCT, and the cumulative number of HY-Abs was significantly associated with increased risks of chronic GVHD and non-relapse mortality. This review focuses on F→M HCT, shedding light on its impact in several clinical settings and presenting clinical evidence of its allogeneic response, encompassing GVHD and graft-versus-leukemia (GVL) effects. Additionally, potential clinical options to mitigate adverse effects in F→M HCT will be discussed. Further investigation is required to improve clinical outcomes and understand allogenic immunological reconstitution after F→M HCT.


Subject(s)
Graft vs Host Disease , Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation , Tissue Donors , Transplantation, Homologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Graft vs Host Disease/immunology , Graft vs Host Disease/etiology , Graft vs Leukemia Effect/immunology , Female , Male
SELECTION OF CITATIONS
SEARCH DETAIL