Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 479
1.
Front Immunol ; 15: 1339318, 2024.
Article En | MEDLINE | ID: mdl-38711496

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.


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
2.
Rinsho Ketsueki ; 65(4): 265-271, 2024.
Article Ja | MEDLINE | ID: mdl-38684437

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.


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
4.
Int J Hematol ; 115(3): 371-381, 2022 Mar.
Article En | MEDLINE | ID: mdl-35037229

Genomic deletion of donor-patient-mismatched HLA alleles in leukemic cells is a major cause of relapse after allogeneic hematopoietic stem cell transplantation (HSCT). Mismatched HLA is frequently lost as an individual allele or a whole region in HLA-class I, however, it is downregulated in HLA-class II. We hypothesized that there might be a difference in T cell recognition capacity against epitopes associated with HLA-class I and HLA-class II and consequently such allogeneic immune pressure induced HLA alterations in leukemic cells. To investigate this, we conducted in vitro experiments with T cell receptor-transduced T (TCR-T) cells. The cytotoxic activity of NY-ESO-1-specific TCR-T cells exhibited similarly against K562 cells with low HLA-A*02:01 expression. However, we demonstrated that the cytokine production against low HLA-DPB1*05:01 expression line decreased gradually from the HLA expression level approximately 2-log lower than normal expressors. Using sort-purified leukemia cells before and after HSCT, we applied the next-generation sequencing, and revealed that there were several marked downregulations of HLA-class II alleles which demonstrated consistently low expression from pre-transplantation. The marked downregulation of HLA-class II may lead to decreased antigen recognition ability of antigen-specific T cells and may be one of immune evasion mechanism associated with HLA-class II downregulation.


Down-Regulation , Epitopes/immunology , Hematopoietic Stem Cell Transplantation , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Leukemia/genetics , Leukemia/immunology , T-Lymphocytes/immunology , Transplantation, Homologous , Alleles , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Humans , K562 Cells , Leukemia/therapy , Recurrence
5.
J Leukoc Biol ; 111(2): 427-437, 2022 02.
Article En | MEDLINE | ID: mdl-34057741

The prognosis for acute myeloid leukemia (AML) relapse post allogeneic hematopoietic stem cell transplantation (alloSCT) is dismal. Novel effective treatment is urgently needed. Clinical benefit of alloSCT greatly relies on the graft-versus-leukemia (GVL) effect. The mechanisms that mediate immune escape of leukemia (thus causing GVL failure) remain poorly understood. Studies of human GVL have been hindered by the lack of optimal clinically relevant models. Here, using our large, longitudinal clinical tissue bank that include AML cells and G-CSF mobilized donor hematopoietic stem cells (HSCs), we successfully established a novel GVL model in humanized mice. Donor HSCs were injected into immune-deficient NOD-Cg-Prkdcscid IL2rgtm1Wjl /SzJ (NSG) mice to build humanized mice. Immune reconstitution in these mice recapitulated some clinical scenario in the patient who received the corresponding HSCs. Allogeneic but HLA partially matched patient-derived AML cells were successfully engrafted in these humanized mice. Importantly, we observed a significantly reduced (yet incomplete elimination of) leukemia growth in humanized mice compared with that in control NSG mice, demonstrating a functional (but defective) GVL effect. Thus, for the first time, we established a novel humanized mouse model that can be used for studying human GVL responses against human AML cells in vivo. This novel clinically relevant model provides a valuable platform for investigating the mechanisms of human GVL and development of effective leukemia treatments.


Disease Models, Animal , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Leukemia, Myeloid, Acute/therapy , Animals , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Leukemia, Myeloid, Acute/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Transplantation, Homologous
6.
J Clin Invest ; 131(24)2021 12 15.
Article En | MEDLINE | ID: mdl-34730109

Despite the curative potential of hematopoietic stem cell transplantation (HSCT), conditioning-associated toxicities preclude broader clinical application. Antibody-drug conjugates (ADCs) provide an attractive approach to HSCT conditioning that minimizes toxicity while retaining efficacy. Initial studies of ADC conditioning have largely focused on syngeneic HSCT. However, to treat acute leukemias or induce tolerance for solid organ transplantation, this approach must be expanded to allogeneic HSCT (allo-HSCT). Using murine allo-HSCT models, we show that pharmacologic Janus kinase 1/2 (JAK1/2) inhibition combined with CD45- or cKit-targeted ADCs enables robust multilineage alloengraftment. Strikingly, myeloid lineage donor chimerism exceeding 99% was achievable in fully MHC-mismatched HSCT using this approach. Mechanistic studies using the JAK1/2 inhibitor baricitinib revealed marked impairment of T and NK cell survival, proliferation, and effector function. NK cells were exquisitely sensitive to JAK1/2 inhibition due to interference with IL-15 signaling. Unlike irradiated mice, ADC-conditioned mice did not develop pathogenic graft-versus-host alloreactivity when challenged with mismatched T cells. Finally, the combination of ADCs and baricitinib balanced graft-versus-host disease and graft-versus-leukemia responses in delayed donor lymphocyte infusion models. Our allo-HSCT conditioning strategy exemplifies the promise of immunotherapy to improve the safety of HSCT for treating hematologic diseases.


Azetidines/pharmacology , Hematopoietic Stem Cell Transplantation , Immunoconjugates/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyrazoles/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Allografts , Animals , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/drug effects , Graft vs Leukemia Effect/genetics , Graft vs Leukemia Effect/immunology , Interleukin-15/genetics , Interleukin-15/immunology , Janus Kinase 1/genetics , Janus Kinase 1/immunology , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology
7.
Front Immunol ; 12: 715893, 2021.
Article En | MEDLINE | ID: mdl-34594330

Allogeneic stem cell transplantation (alloSCT) is a curative therapy for hematopoietic malignancies. The therapeutic effect relies on donor T cells and NK cells to recognize and eliminate malignant cells, known as the graft-versus-leukemia (GVL) effect. However, off target immune pathology, known as graft-versus-host disease (GVHD) remains a major complication of alloSCT that limits the broad application of this therapy. The presentation of recipient-origin alloantigen to donor T cells is the primary process initiating GVHD and GVL. Therefore, the understanding of spatial and temporal characteristics of alloantigen presentation is pivotal to attempts to separate beneficial GVL effects from detrimental GVHD. In this review, we discuss mouse models and the tools therein, that permit the quantification of alloantigen presentation after alloSCT.


Antigen Presentation/immunology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Transplantation Immunology , Animals , Graft vs Host Disease/etiology , Graft vs Leukemia Effect/immunology , H-2 Antigens/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Isoantigens/immunology , Mice , Minor Histocompatibility Antigens/immunology , Molecular Mimicry/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
8.
J Interferon Cytokine Res ; 41(9): 310-318, 2021 09.
Article En | MEDLINE | ID: mdl-34543129

Graft-versus-host disease (GVHD) is a physiological response of the graft to allogeneic hosts. However, the effector cells, affected organ(s), and cytokines in the GVHD remain controversially discussed, without having determined a particular cytotoxic activity of the graft against the host. After i.v. injection of C57BL/6 (H-2b) spleen cells into irradiated BDF1 (H-2b/d) mice, the hosts developed interferon-gamma (IFN-γ)-dependent bone marrow (BM) GVHD on days 5-17. When H-2DdKd transgenic H-2b lymphoma cells were i.p. inoculated into irradiated, H-2b splenocyte-transplanted H-2b/d mice, the infiltration of macrophages cytotoxic against H-2DdKd transgenic H-2b mouse skin epithelia (a GVHD activity) into the peritoneal cavity preceded several days the infiltration of interleukin (IL)-2-dependent cytotoxic T lymphocytes (CTLs) to achieve a graft-versus-leukemia (GVL) effect. In contrast, allogeneic BM transplanted alone into the irradiated mice did not induce GVHD for 44 days, whereas i.v. injection of graft anti-host macrophages or graft anti-host CTLs along with allogeneic BM, respectively, induced GVHD or promoted the GVL effect in the absence of GVHD. These results revealed that macrophage-induced GVHD and the CTL-mediated GVL effect were a set (Th1: IFN-γ/IL-2) response of the graft to allogeneic hosts and leukemia cells, respectively, and that graft T cell activation rather than inhibition skipped GVHD after BM transplantation.


Bone Marrow/immunology , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Macrophages/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Bone Marrow Transplantation/methods , Cell Line, Tumor , Hematopoietic Stem Cell Transplantation/methods , Interferon-gamma/immunology , Interleukin-2/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA
9.
Front Immunol ; 12: 697854, 2021.
Article En | MEDLINE | ID: mdl-34220860

Graft versus host disease (GVHD) is a common complication and the leading cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Pharmacological immunosuppression used in GVHD prophylaxis and treatment lacks specificity and can increase the likelihood of infection and relapse. Regulatory T lymphocytes (Tregs) play a vital role in restraining excessive immune responses and inducing peripheral immune tolerance. In particular, clinical trials have demonstrated that Tregs can prevent and treat GVHD, without increasing the risk of relapse and infection. Hence, adoptive transfer of Tregs to control GVHD using their immunosuppressive properties represents a promising therapeutic approach. To optimally apply Tregs for control of GVHD, a thorough understanding of their biology is necessary. In this review, we describe the biological characteristics of Tregs, including how the stability of FOXP3 expression can be maintained. We will also discuss the mechanisms underlying Tregs-mediated modulation of GVHD and approaches to effectively increase Tregs' numbers. Finally, we will examine the developing trends in the use of Tregs for clinical therapy.


Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Cell Proliferation , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/immunology , Humans , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Immunotherapy, Adoptive , Models, Immunological , Peripheral Tolerance , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Tissue Donors
10.
Front Immunol ; 12: 652105, 2021.
Article En | MEDLINE | ID: mdl-33968052

Transplantation of allogeneic hematopoietic cells faces two barriers: failure of engraftment due to a host versus graft reaction, and the attack of donor cells against the patient, the graft versus host (GVH) reaction. This reaction may lead to GVH disease (GVHD), but in patients transplanted due to leukemia or other malignant disorders, this may also convey the benefit of a graft versus leukemia (GVL) effect. The interplay of transplant conditioning with donor and host cells and the environment in the patient is complex. The microbiome, particularly in the intestinal tract, profoundly affects these interactions, directly and via soluble mediators, which also reach other host organs. The microenvironment is further altered by the modifying effect of malignant cells on marrow niches, favoring the propagation of the malignant cells. The development of stable mixed donor/host chimerism has the potential of GVHD prevention without necessarily increasing the risk of relapse. There has been remarkable progress with novel conditioning regimens and selective T-cell manipulation aimed at securing engraftment while preventing GVHD without ablating the GVL effect. Interventions to alter the microenvironment and change the composition of the microbiome and its metabolic products may modify graft/host interactions, thereby further reducing GVHD, while enhancing the GVL effect. The result should be improved transplant outcome.


Chimerism , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Leukemia/therapy , Neoplasm Recurrence, Local/prevention & control , Disease-Free Survival , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Reaction/drug effects , Graft vs Host Reaction/genetics , Graft vs Host Reaction/immunology , Graft vs Leukemia Effect/genetics , Graft vs Leukemia Effect/immunology , Humans , Leukemia/genetics , Leukemia/immunology , Leukemia/mortality , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/immunology , Transplantation Conditioning/methods , Transplantation, Homologous/adverse effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
11.
Br J Haematol ; 195(3): 433-446, 2021 11.
Article En | MEDLINE | ID: mdl-34046897

Allogeneic immune responses underlie the graft-versus-leukaemia effect of stem cell transplantation, but disease relapse occurs in many patients. Minor histocompatibility antigen (mHAg) peptides mediate alloreactive T cell responses and induce graft-versus-leukaemia responses when expressed on patient haematopoietic tissue. We vaccinated nine HA-1-negative donors against HA-1 with a 'prime-boost' protocol of either two or three DNA 'priming' vaccinations prior to 'boost' with modified vaccinia Ankara (MVA). HA-1-specific CD8+ T cell responses were observed in seven donors with magnitude up to 1·5% of total CD8+ T cell repertoire. HA-1-specific responses peaked two weeks post-MVA challenge and were measurable in most donors after 12 months. HA-1-specific T cells demonstrated strong cytotoxic activity and lysed target cells with endogenous HA-1 protein expression. The pattern of T cell receptor (TCR) usage by HA-1-specific T cells revealed strong conservation of T cell receptor beta variable 7-9 (TRBV7-9) usage between donors. These findings describe one of the strongest primary peptide-specific CD8+ T cell responses yet recorded to a DNA-MVA prime-boost regimen and this may reflect the strong immunogenicity of mHAg peptides. Prime-boost vaccination in donors or patients may prove of substantial benefit in boosting graft-versus-leukaemia responses.


Antigens, Neoplasm/immunology , Graft vs Leukemia Effect/immunology , Minor Histocompatibility Antigens/immunology , Oligopeptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccination , Vaccines, DNA/therapeutic use , Vaccinia virus/immunology , Viral Vaccines/therapeutic use , Adult , Aged , Allografts , Cytotoxicity, Immunologic , Epitopes/immunology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , HLA-A2 Antigen/immunology , Hematopoietic Stem Cell Transplantation , Humans , Immunogenicity, Vaccine , Immunologic Memory , Male , Middle Aged , Peptides/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Vaccines, Attenuated , Vaccines, DNA/immunology , Viral Vaccines/immunology
12.
Front Immunol ; 12: 641910, 2021.
Article En | MEDLINE | ID: mdl-33732262

Histone deacetylase inhibitors are currently the most studied drugs because of their beneficial effects on inflammatory response. Emerging data from numerous basic studies and clinical trials have shown that histone deacetylase inhibitors can suppress immune-mediated diseases, such as graft-vs.-host disease (GVHD), while retaining beneficial graft-vs.-leukemia (GVL) effects. These drugs prevent and/or treat GVHD by modifying gene expression and inhibiting the production of proinflammatory cytokines, regulating the function of alloreactive T cells, and upregulating the function and number of regulatory T cells. Some of these drugs may become new immunotherapies for GVHD and other immune diseases.


Graft vs Host Disease , Graft vs Leukemia Effect , Histone Deacetylase Inhibitors/therapeutic use , Immunologic Factors/therapeutic use , Stem Cell Transplantation , T-Lymphocytes, Regulatory/immunology , Allografts , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/drug effects , Graft vs Leukemia Effect/immunology , Humans
13.
JCI Insight ; 6(3)2021 02 08.
Article En | MEDLINE | ID: mdl-33554953

Graft-versus-host disease (GVHD) is a pathological process caused by an exaggerated donor lymphocyte response to host antigens after allogeneic hematopoietic cell transplantation (allo-HCT). Donor T cells undergo extensive clonal expansion and differentiation, which culminate in damage to recipient target organs. Damage to the gastrointestinal tract is a main contributor to morbidity and mortality. The loss of diversity among intestinal bacteria caused by pretransplant conditioning regimens leads to an outgrowth of opportunistic pathogens and exacerbated GVHD after allo-HCT. Using murine models of allo-HCT, we found that an increase of Bacteroides in the intestinal microbiota of the recipients was associated with reduced GVHD in mice given fecal microbial transplantation. Administration of Bacteroides fragilis through oral gavage increased gut microbiota diversity and beneficial commensal bacteria and significantly ameliorated acute and chronic GVHD development. Preservation of gut integrity following B. fragilis exposure was likely attributed to increased short chain fatty acids, IL-22, and regulatory T cells, which in turn improved gut tight junction integrity and reduced inflammatory cytokine production of pathogenic T cells. The current study provides a proof of concept that a single strain of commensal bacteria can be a safe and effective means to protect gut integrity and ameliorate GVHD after allo-HCT.


Bacteroides fragilis/immunology , Gastrointestinal Microbiome/immunology , Graft vs Host Disease/prevention & control , Allografts , Animals , Disease Models, Animal , Fecal Microbiota Transplantation , Graft vs Host Disease/immunology , Graft vs Host Disease/microbiology , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Isoantigens/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , T-Lymphocytes/immunology , Tumor Cells, Cultured
16.
Cancer Res ; 81(4): 1063-1075, 2021 02 15.
Article En | MEDLINE | ID: mdl-33277367

Autophagy is a vital cellular process whose role in T immune cells is poorly understood, specifically, in its regulation of allo-immunity. Stimulation of wild-type T cells in vitro and in vivo with allo-antigens enhances autophagy. To assess the relevance of autophagy to T-cell allo-immunity, we generated T-cell-specific Atg5 knock-out mice. Deficiency of ATG5-dependent autophagy reduced T-cell proliferation and increased apoptosis following in vitro and in vivo allo-stimulation. The absence of ATG5 in allo-stimulated T cells enhanced their ability to release effector cytokines and cytotoxic functions, uncoupling their proliferation and effector functions. Absence of autophagy reduced intracellular degradation of cytotoxic enzymes such as granzyme B, thus enhancing the cytotoxicity of T cells. In several in vivo models of allo-HSCT, ATG5-dependent dissociation of T-cell functions contributed to significant reduction in graft-versus-host disease (GVHD) but retained sufficient graft versus tumor (GVT) response. Our findings demonstrate that ATG5-dependent autophagy uncouples T-cell proliferation from its effector functions and offers a potential new strategy to enhance outcomes after allo-HSCT. SIGNIFICANCE: These findings demonstrate that induction of autophagy in donor T-cell promotes GVHD, while inhibition of T-cell autophagy mitigates GVHD without substantial loss of GVL responses.


Autophagy-Related Protein 5/physiology , Graft vs Host Disease/genetics , Graft vs Leukemia Effect/genetics , T-Lymphocytes/physiology , Animals , Autophagy-Related Protein 5/genetics , Cell Proliferation/genetics , Cells, Cultured , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Leukemia Effect/immunology , Humans , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
17.
Front Immunol ; 11: 593863, 2020.
Article En | MEDLINE | ID: mdl-33324410

Allogeneic hematopoietic stem cell transplantation is a potentially curative procedure for many malignant diseases. Donor T cells prevent disease recurrence via graft-versus-leukemia (GVL) effect. Donor T cells also contribute to graft-versus-host disease (GVHD), a debilitating and potentially fatal complication. Novel treatment strategies are needed which allow preservation of GVL effects without causing GVHD. Using murine models, we show that targeting IL-2-inducible T cell kinase (ITK) in donor T cells reduces GVHD while preserving GVL effects. Both CD8+ and CD4+ donor T cells from Itk-/- mice produce less inflammatory cytokines and show decrease migration to GVHD target organs such as the liver and small intestine, while maintaining GVL efficacy against primary B-cell acute lymphoblastic leukemia (B-ALL). Itk-/- T cells exhibit reduced expression of IRF4 and decreased JAK/STAT signaling activity but upregulating expression of Eomesodermin (Eomes) and preserve cytotoxicity, necessary for GVL effect. Transcriptome analysis indicates that ITK signaling controls chemokine receptor expression during alloactivation, which in turn affects the ability of donor T cells to migrate to GVHD target organs. Our data suggest that inhibiting ITK could be a therapeutic strategy to reduce GVHD while preserving the beneficial GVL effects following allo-HSCT treatment.


Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Graft vs Leukemia Effect/genetics , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation , Protein-Tyrosine Kinases/genetics , Animals , Cell Movement/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic , Diagnosis, Differential , Disease Models, Animal , Gene Expression Regulation , Graft vs Host Disease/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Immunity, Innate , Immunologic Memory , Immunomodulation , Interleukin-2/metabolism , Mice , Mice, Knockout , Protein-Tyrosine Kinases/metabolism , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
18.
Front Immunol ; 11: 558143, 2020.
Article En | MEDLINE | ID: mdl-33178187

Disease relapse and graft-versus-host disease (GVHD) are the major complications affecting the outcomes of allogeneic hematopoietic stem cell transplantation (allo-HSCT). While the functions of αßT cells are extensively studied, the role of donor γδT cells in allo-HSCT is less well defined. Using TCRδ-/- donors lacking γδT cells, we demonstrated that donor γδT cells were critical in mediating graft-versus-leukemia (GVL) effect during allo-HSCT. In the absence of donor γδT cells, IFN-γ production by CD8+ T cells was severely impaired. Vγ4 subset was the major γδT cell subset mediating the GVL effect in vivo, which was partially dependent on IL-17A. Meanwhile, donor γδT cells could mitigate acute GVHD in a murine allo-HSCT model by suppressing CD4+ T cell activation and the major γδT cell subset that exerted this protective function was also Vγ4 γδT cells. Therefore, our findings provide evidence that donor γδT cells, especially Vγ4 subset, can enhance GVL effect and mitigate aGVHD during allo-HSCT.


Graft vs Host Disease/etiology , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adoptive Transfer , Animals , Biomarkers , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Cytotoxicity, Immunologic , Graft vs Leukemia Effect/genetics , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Immunohistochemistry , Immunophenotyping , Luminescent Measurements , Mice , Mice, Transgenic
19.
J Immunol ; 205(12): 3480-3490, 2020 12 15.
Article En | MEDLINE | ID: mdl-33158956

Acute graft-versus-host disease (aGVHD) is one major serious complication that is induced by alloreactive donor T cells recognizing host Ags and limits the success of allogeneic hematopoietic stem cell transplantation. In the current studies, we identified a critical role of Kras in regulating alloreactive T cell function during aGVHD. Kras deletion in donor T cells dramatically reduced aGVHD mortality and severity in an MHC-mismatched allogeneic hematopoietic stem cell transplantation mouse model but largely maintained the antitumor capacity. Kras-deficient CD4 and CD8 T cells exhibited impaired TCR-induced activation of the ERK pathway. Kras deficiency altered TCR-induced gene expression profiles, including the reduced expression of various inflammatory cytokines and chemokines. Moreover, Kras deficiency inhibited IL-6-mediated Th17 cell differentiation and impaired IL-6-induced ERK activation and gene expression in CD4 T cells. These findings support Kras as a novel and effective therapeutic target for aGVHD.


CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation , Proto-Oncogene Proteins p21(ras)/deficiency , Th17 Cells/immunology , Allografts , Animals , Cell Line, Tumor , Graft vs Host Disease/genetics , Graft vs Leukemia Effect/genetics , Interleukin-6/genetics , Interleukin-6/immunology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Transgenic , Proto-Oncogene Proteins p21(ras)/immunology
20.
Am J Hematol ; 95(12): 1590-1598, 2020 12.
Article En | MEDLINE | ID: mdl-32857869

Allogeneic hematopoietic stem cell transplantation (alloSCT) for myeloid leukemia remains one of the most effective anti-tumor treatments available, capable of curing an increasingly higher proportion of patients. Alloreactivity generated by T cells has limited efficacy in the early post-transplant period while most patients will relapse within 6 months after transplantation. Prior studies in T cell depleted grafts showed that, with the elimination of T cells, natural killer (NK) cells provide most of the anti-tumor effect in the early post-transplant period. Administration of unmodified T cells to mitigate infections and relapse will expose the patient to a high risk of graft-vs-host disease (GvHD). Post-transplant cyclophosphamide (PTCy), initially used for haploidentical (haplo) donor transplants, is now also increasingly utilized in HLA matched donor transplants to prevent GvHD. In most patients, PTCy eliminates, at least in part, alloreactive T and NK cells early post-transplant. Administration of modified NK cells in the early post-transplant period makes intuitive sense to enhance the anti-tumor effect of the graft and thereby prevent relapse. Effective application of cellular therapy early after transplant has opened a new direction and could revolutionize the field of hematopoietic stem cell transplantation.


Cyclophosphamide/therapeutic use , Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute , T-Lymphocytes/immunology , Allografts , Graft vs Leukemia Effect/drug effects , Graft vs Leukemia Effect/immunology , Humans , Killer Cells, Natural/pathology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Recurrence , T-Lymphocytes/pathology
...