Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 106
2.
JCI Insight ; 9(9)2024 Apr 11.
Article En | MEDLINE | ID: mdl-38602775

Allogeneic hematopoietic stem cell transplantation (aHSCT) can cure patients with otherwise fatal leukemias and lymphomas. However, the benefits of aHSCT are limited by graft-versus-host disease (GVHD). Minnelide, a water-soluble analog of triptolide, has demonstrated potent antiinflammatory and antitumor activity in several preclinical models and has proven both safe and efficacious in clinical trials for advanced gastrointestinal malignancies. Here, we tested the effectiveness of Minnelide in preventing acute GVHD as compared with posttransplant cyclophosphamide (PTCy). Strikingly, we found Minnelide improved survival, weight loss, and clinical scores in an MHC-mismatched model of aHSCT. These benefits were also apparent in minor MHC-matched aHSCT and xenogeneic HSCT models. Minnelide was comparable to PTCy in terms of survival, GVHD clinical score, and colonic length. Notably, in addition to decreased donor T cell infiltration early after aHSCT, several regulatory cell populations, including Tregs, ILC2s, and myeloid-derived stem cells in the colon were increased, which together may account for Minnelide's GVHD suppression after aHSCT. Importantly, Minnelide's GVHD prevention was accompanied by preservation of graft-versus-tumor activity. As Minnelide possesses anti-acute myeloid leukemia (anti-AML) activity and is being applied in clinical trials, together with the present findings, we conclude that this compound might provide a new approach for patients with AML undergoing aHSCT.


Diterpenes , Epoxy Compounds , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Phenanthrenes , Graft vs Host Disease/prevention & control , Graft vs Host Disease/drug therapy , Animals , Mice , Hematopoietic Stem Cell Transplantation/methods , Diterpenes/pharmacology , Diterpenes/therapeutic use , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Phenanthrenes/pharmacology , Phenanthrenes/therapeutic use , Humans , Transplantation, Homologous , Female , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Disease Models, Animal , Graft vs Leukemia Effect/drug effects , Mice, Inbred C57BL , Male
3.
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
4.
Sci Rep ; 11(1): 13125, 2021 06 23.
Article En | MEDLINE | ID: mdl-34162921

Posttransplantation cyclophosphamide (PTCy) has become a popular option for haploidentical hematopoietic stem cell transplantation (HSCT). However, personalized methods to adjust immune intensity after PTCy for each patient's condition have not been well studied. Here, we investigated the effects of reducing the dose of PTCy followed by α-galactosylceramide (α-GC), a ligand of iNKT cells, on the reciprocal balance between graft-versus-host disease (GVHD) and the graft-versus-leukemia (GVL) effect. In a murine haploidentical HSCT model, insufficient GVHD prevention after reduced-dose PTCy was efficiently compensated for by multiple administrations of α-GC. The ligand treatment maintained the enhanced GVL effect after reduced-dose PTCy. Phenotypic analyses revealed that donor-derived B cells presented the ligand and induced preferential skewing to the NKT2 phenotype rather than the NKT1 phenotype, which was followed by the early recovery of all T cell subsets, especially CD4+Foxp3+ regulatory T cells. These studies indicate that α-GC administration soon after reduced-dose PTCy restores GVHD-preventing activity and maintains the GVL effect, which is enhanced by reducing the dose of PTCy. Our results provide important information for the development of a novel strategy to optimize PTCy-based transplantation, particularly in patients with a potential relapse risk.


Cyclophosphamide/therapeutic use , Galactosylceramides/therapeutic use , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/drug effects , Adjuvants, Pharmaceutic/therapeutic use , Animals , B-Lymphocytes/drug effects , Bone Marrow Transplantation/adverse effects , Dendritic Cells/drug effects , Dose-Response Relationship, Drug , Female , Macrophages/drug effects , Mice , Mice, Inbred C57BL
5.
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
6.
Blood ; 137(16): 2243-2255, 2021 04 22.
Article En | MEDLINE | ID: mdl-33511398

Donor T cells mediate both graft-versus-leukemia (GVL) activity and graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (allo-HCT). Development of methods that preserve GVL activity while preventing GVHD remains a long-sought goal. Tolerogenic anti-interleukin-2 (IL-2) monoclonal antibody (JES6-1) forms anti-IL-2/IL-2 complexes that block IL-2 binding to IL-2Rß and IL-2Rγ on conventional T cells that have low expression of IL-2Rα. Here, we show that administration of JES6 early after allo-HCT in mice markedly attenuates acute GVHD while preserving GVL activity that is dramatically stronger than observed with tacrolimus (TAC) treatment. The anti-IL-2 treatment downregulated activation of the IL-2-Stat5 pathway and reduced production of granulocyte-macrophage colony-stimulating factor (GM-CSF). In GVHD target tissues, enhanced T-cell programmed cell death protein 1 (PD-1) interaction with tissue-programmed cell death-ligand 1 (PD-L1) led to reduced activation of protein kinase-mammalian target of rapamycin pathway and increased expression of eomesodermin and B-lymphocyte-induced maturation protein-1, increased T-cell anergy/exhaustion, expansion of Foxp3-IL-10-producing type 1 regulatory (Tr1) cells, and depletion of GM-CSF-producing T helper type 1 (Th1)/cytotoxic T cell type 1 (Tc1) cells. In recipient lymphoid tissues, lack of donor T-cell PD-1 interaction with tissue PD-L1 preserved donor PD-1+TCF-1+Ly108+CD8+ T memory progenitors and functional effectors that have strong GVL activity. Anti-IL-2 and TAC treatments have qualitatively distinct effects on donor T cells in the lymphoid tissues, and CD8+ T memory progenitor cells are enriched with anti-IL-2 treatment compared with TAC treatment. We conclude that administration of tolerogenic anti-IL-2 monoclonal antibody early after allo-HCT represents a novel approach for preventing acute GVHD while preserving GVL activity.


Antibodies, Monoclonal/therapeutic use , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/drug effects , Hematopoietic Stem Cell Transplantation , Interleukin-2/immunology , Animals , Antibodies, Monoclonal/immunology , Graft vs Host Disease/immunology , Immunosuppressive Agents/therapeutic use , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tacrolimus/therapeutic use , Transplantation, Homologous
7.
Front Immunol ; 12: 785774, 2021.
Article En | MEDLINE | ID: mdl-34987512

Graft-versus-host disease (GVHD) remains the major cause of mortality and morbidity in non-relapse patients after allogeneic hematopoietic cell transplantation (allo-HCT). As the number of patients undergoing allo-HCT increases, it will become imperative to determine safe and effective treatment options for patients with GVHD, especially those who become refractory to systemic steroid therapy. Daratumumab (Dara), a humanized IgG1 (ĸ subclass) monoclonal antibody targeting the CD38 epitope, is used for the treatment of multiple myeloma. CD38 is a multifunctional ectoenzyme that behaves either as an enzyme, a cell adhesion molecule or a cell surface receptor involved in cell signaling. CD38 is also expressed on various immune effector and suppressor cells. However, the role of CD38 in the immune response remains elusive. We questioned whether CD38 is a potential therapeutic target against alloreactive T cells in the GVHD pathological process. Here, we investigated the impact of Dara on xenogeneic GVHD (xeno-GVHD) and graft-versus-leukemia (GVL) effects in a humanized murine model of transplantation, where human peripheral blood mononuclear cells were adoptively transplanted into immunocompromised NOD.SCID.gc-null (NSG) mice. Mice receiving Dara treatment experienced less weight loss, longer survival and lower GVHD scores compared with those in the control group. Histological evaluations, flow cytometry, RNA-sequencing and RT-qPCR analysis revealed that Dara efficaciously mitigated GVHD through multiple mechanisms including inhibition of the proliferation, activation and differentiation of CD8+ cytotoxic T cells, reduced expression of cytotoxic effector molecules, pro-inflammatory cytokines, chemokines and chemoattractant receptors by T cells and promotion of immunosuppressive T cells. More importantly, Dara preserved the GVL effect in a humanized mouse model of leukemia by metabolic reprograming of T cells to promote the induction of Th17, Th1/17and Tc1/17 cells. Our findings indicate that Dara may be an attractive therapeutic option to separate GVHD from GVL effects in patients with hematopoietic malignancies receiving allo-HCT.


Antibodies, Monoclonal/administration & dosage , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/drug effects , Hematopoietic Stem Cell Transplantation/adverse effects , Leukemia/therapy , Animals , Cell Line, Tumor , Female , Graft vs Host Disease/immunology , Healthy Volunteers , Hematopoietic Stem Cell Transplantation/methods , Humans , Leukemia/immunology , Leukocytes, Mononuclear , Mice , Primary Cell Culture , Transplantation, Heterologous/adverse effects , Transplantation, Homologous/adverse effects , Transplantation, Homologous/methods , Xenograft Model Antitumor Assays
8.
Blood ; 137(8): 1090-1103, 2021 02 25.
Article En | MEDLINE | ID: mdl-32976550

The nuclear receptor (NR) subclass, retinoid X receptors (RXRs), exert immunomodulatory functions that control inflammation and metabolism via homodimers and heterodimers, with several other NRs, including retinoic acid receptors. IRX4204 is a novel, highly specific RXR agonist in clinical trials that potently and selectively activates RXR homodimers, but not heterodimers. In this study, in vivo IRX4204 compared favorably with FK506 in abrogating acute graft-versus-host disease (GVHD), which was associated with inhibiting allogeneic donor T-cell proliferation, reducing T-helper 1 differentiation, and promoting regulatory T-cell (Treg) generation. Recipient IRX4204 treatment reduced intestinal injury and decreased IFN-γ and TNF-α serum levels. Transcriptional analysis of donor T cells isolated from intestines of GVHD mice treated with IRX4204 revealed significant decreases in transcripts regulating proinflammatory pathways. In vitro, inducible Treg differentiation from naive CD4+ T cells was enhanced by IRX4204. In vivo, IRX4204 increased the conversion of donor Foxp3- T cells into peripheral Foxp3+ Tregs in GVHD mice. Using Foxp3 lineage-tracer mice in which both the origin and current FoxP3 expression of Tregs can be tracked, we demonstrated that IRX4204 supports Treg stability. Despite favoring Tregs and reducing Th1 differentiation, IRX4204-treated recipients maintained graft-versus-leukemia responses against both leukemia and lymphoma cells. Notably, IRX4204 reduced in vitro human T-cell proliferation and enhanced Treg generation in mixed lymphocyte reaction cultures. Collectively, these beneficial effects indicate that targeting RXRs with IRX4204 could be a novel approach to preventing acute GVHD in the clinic.


Bone Marrow Transplantation , Cyclopropanes/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Leukemia Effect/drug effects , Retinoid X Receptors/agonists , Animals , Bone Marrow Transplantation/adverse effects , Drug Repositioning , Female , Graft vs Host Disease/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology
9.
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
10.
Int J Mol Sci ; 21(12)2020 Jun 16.
Article En | MEDLINE | ID: mdl-32560120

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the main therapeutic strategy for patients with both malignant and nonmalignant disorders. The therapeutic benefits of allo-HSCT in malignant disorders are primarily derived from the graft-versus-leukemia (GvL) effect, in which T cells in the donor graft recognize and eradicate residual malignant cells. However, the same donor T cells can also recognize normal host tissues as foreign, leading to the development of graft-versus-host disease (GvHD), which is difficult to separate from GvL and is the most frequent and serious complication following allo-HSCT. Inhibition of donor T cell toxicity helps in reducing GvHD but also restricts GvL activity. Therefore, developing a novel therapeutic strategy that selectively suppresses GvHD without affecting GvL is essential. Recent studies have shown that inhibition of histone deacetylases (HDACs) not only inhibits the growth of tumor cells but also regulates the cytotoxic activity of T cells. Here, we compile the known therapeutic potential of HDAC inhibitors in preventing several stages of GvHD pathogenesis. Furthermore, we will also review the current clinical features of HDAC inhibitors in preventing and treating GvHD as well as maintaining GvL.


Graft vs Host Disease/drug therapy , Graft vs Leukemia Effect/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Neoplasms/therapy , Clinical Trials as Topic , Graft vs Host Disease/enzymology , Hematopoietic Stem Cell Transplantation/adverse effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Neoplasms/enzymology , Neoplasms/metabolism , T-Lymphocytes, Cytotoxic/drug effects , Transplantation, Homologous
11.
Int J Hematol ; 112(2): 249-253, 2020 Aug.
Article En | MEDLINE | ID: mdl-32185622

Patients with FMS-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) acute myeloid leukemia (AML) respond to conventional induction chemotherapy, with remission rates similar to those seen in other subtypes; however, they are much more likely to relapse and relapse is rapid. For this reason, eligible patients receive consolidation therapy with early allogenic transplantation, but the recurrence rate remains high, even after transplantation. Moreover, the optimal therapy for patients with FLT3-ITD AML who relapse after allogeneic hematopoietic stem cell transplantation remains unclear. Here, we report a case in which graft-versus-leukemia (GVL) effects were induced by gilteritinib administration after a second transplant from the same donor, resulting in sustained remission of early FLT3-ITD AML relapse after allogeneic transplantation. Several studies suggest that the benefits of FLT3 tyrosine kinase inhibitors (FLT3-TKI) after allogeneic transplantation are attributable to GVL induction, as well as direct effects on FLT3 mutation-positive leukemia cells. With this in mind, we induced lymphodepletion using L-PAM to further enhance GVL induction by donor lymphocytes and FLT3-TKI. We believe that enhancement of GVL induction by lymphodepletion should be considered before FLT3-TKI use, if the prognosis is very poor, such as in patients with recurrence following allogeneic transplantation.


Aniline Compounds/administration & dosage , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/surgery , Pyrazines/administration & dosage , Recurrence , Tandem Repeat Sequences/genetics , Tissue Donors , fms-Like Tyrosine Kinase 3/genetics , Aniline Compounds/pharmacology , Female , Graft vs Leukemia Effect/drug effects , Humans , Leukemia, Myeloid, Acute/immunology , Mutation , Pyrazines/pharmacology , Remission Induction , Reoperation , Treatment Outcome , fms-Like Tyrosine Kinase 3/antagonists & inhibitors
12.
Eur J Immunol ; 50(8): 1220-1233, 2020 08.
Article En | MEDLINE | ID: mdl-32133644

Glucocorticoids (GCs) are widely used to treat acute graft-versus-host disease (aGvHD) due to their immunosuppressive activity, but they also reduce the beneficial graft-versus-leukemia (GvL) effect of the allogeneic T cells contained in the graft. Here, we tested whether aGvHD therapy could be improved by delivering GCs with the help of inorganic-organic hybrid nanoparticles (IOH-NPs) that preferentially target myeloid cells. IOH-NPs containing the GC betamethasone (BMP-NPs) efficiently reduced morbidity, mortality, and tissue damage in a totally MHC mismatched mouse model of aGvHD. Therapeutic activity was lost in mice lacking the GC receptor (GR) in myeloid cells, confirming the cell type specificity of our approach. BMP-NPs had no relevant systemic activity but suppressed cytokine and chemokine gene expression locally in the small intestine, which presumably explains their mode of action. Most importantly, BMP-NPs delayed the development of an adoptively transferred B cell lymphoma better than the free drug, although the overall incidence was unaffected. Our findings thus suggest that employing IOH-NPs could diminish the risk of relapse associated with GC therapy of aGvHD patients while still allowing to efficiently ameliorate the disease.


Betamethasone/analogs & derivatives , Graft vs Host Disease/drug therapy , Graft vs Leukemia Effect/drug effects , Nanoparticles/administration & dosage , Acute Disease , Animals , Betamethasone/administration & dosage , Cytokines/blood , Disease Models, Animal , Intestine, Small/immunology , Intestine, Small/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/drug effects
13.
Cancer Med ; 8(2): 501-514, 2019 02.
Article En | MEDLINE | ID: mdl-30677244

Deferasirox is an oral iron-chelating agent having possible antileukemia and immune modulatory effects. Few reports have evaluated deferasirox in the setting of allogeneic hematopoietic stem cell transplantation (allo-HSCT). We investigated the impact of deferasirox after allo-HSCT in acute myeloid leukemia (AML). Of 326 consecutive patients undergoing allo-HSCT in remission, analysis of 198 patients not receiving deferasirox revealed the negative prognostic effect of hyperferritinemia (≥1000 ng/mL) before and after allo-HSCT on survival mainly due to increase in relapse. Of 276 patients with hyperferritinemia at 1 month after allo-HSCT, 128 patients (46%) received deferasirox. Deferasirox induced a faster decline in serum ferritin level with a manageable safety profile, which significantly reduced relapse rather than nonrelapse mortality, resulting in better survival compared to patients not receiving deferasirox. Of note, the deferasirox group had a significantly higher incidence of chronic graft-vs-host disease, indicating improved graft-vs-leukemia (GVL) effects evidenced by the presence of suppressed regulatory T cells and sustained higher proportion of NK cells in peripheral blood. This study firstly demonstrates the improved survival and restoration of GVL effects of patients with AML by deferasirox, which also clarifies the detrimental effect of hyperferritinemia through after allo-HSCT.


Deferasirox/therapeutic use , Graft vs Leukemia Effect/drug effects , Hematopoietic Stem Cell Transplantation , Iron Chelating Agents/therapeutic use , Leukemia, Myeloid, Acute/therapy , Adolescent , Adult , Aged , Female , Ferritins/blood , Humans , Leukemia, Myeloid, Acute/blood , Male , Middle Aged , Treatment Outcome , Young Adult
14.
Blood ; 133(3): 266-279, 2019 01 17.
Article En | MEDLINE | ID: mdl-30514750

Graft-versus-host disease (GVHD) remains one of the major complications after allogeneic bone marrow transplantation (allo-BMT). Sirtuin-1 (Sirt-1) plays a crucial role in various biological processes including cellular senescence, metabolism, and inflammatory responses. Sirt-1 deacetylation regulates different transcription factors that are important for modulating immune responses. In the current study, we addressed the role of Sirt-1 in GVHD induction by employing Sirt-1 conditional knockout mice as well as a pharmacological Sirt-1 inhibitor. Using major histocompatibility complex (MHC)-mismatched and MHC-matched murine BMT models, we found that Sirt-1-/- T cells had a reduced ability to induce acute GVHD (aGVHD) via enhanced p53 acetylation. Sirt-1-deficient T cells also promoted induced regulatory T cell (iTreg) differentiation and inhibited interferon-γ production after allo-BMT. Sirt-1 deletion in iTregs increased Foxp3 stability and restrained iTreg conversion into pathogenic T cells. Furthermore, we found that administration with a Sirt-1 inhibitor, Ex-527, significantly improved recipient survival and clinical scores, with no signs of tumor relapse. These results indicate that Sirt-1 inhibition can attenuate GVHD while preserving the graft-versus-leukemia effect. Consistently, Sirt-1-deficient T cells also displayed a remarkably reduced ability to induce chronic GVHD (cGVHD). Mechanistic studies revealed that Sirt-1 deficiency in T cells enhanced splenic B-cell reconstitution and reduced follicular T helper cell development. Sirt-1 deficiency in T cells modulated donor B-cell responses reducing both B-cell activation and plasma cell differentiation. In addition, therapeutic Sirt-1 inhibition could both prevent cGVHD and reduce established cGVHD. In conclusion, Sirt-1 is a promising therapeutic target for the control of aGVHD and cGVHD pathogenesis and possesses high potential for clinical application.


B-Lymphocytes/immunology , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Lymphocyte Activation/immunology , Sirtuin 1/physiology , T-Lymphocytes, Regulatory/immunology , Acetylation , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Bone Marrow Transplantation , Carbazoles/pharmacology , Cell Differentiation , Female , Graft vs Host Disease/drug therapy , Graft vs Host Disease/pathology , Graft vs Leukemia Effect/drug effects , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Sirtuin 1/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous , Tumor Suppressor Protein p53/metabolism
15.
Bone Marrow Transplant ; 53(12): 1518-1521, 2018 12.
Article En | MEDLINE | ID: mdl-30116019

Allogeneic stem cell transplantation (Allo-HSCT) is sine qua non to cure high-risk acute myeloid leukaemia (AML). In spite the advent of highly active antiretroviral treatment, HIV-infected patients display a remarkable risk for haematological neoplasms such as non-Hodgkin lymphomas, Hodgkin lymphoma and acute leukaemia. Several case series have confirmed the efficacy of the autologous stem cell transplantation for the treatment of non-Hodgkin lymphomas in the HIV setting. Nonetheless, there is a paucity of data for the role of the Allo-HSCT in HIV-infected individuals with haematological malignancies. Herein, we presented the successful long-term outcome of a HIV-infected patient who received reduced intensity conditioned, matched unrelated donor transplant with alemtuzumab as graft-versus-host disease prophylaxis for therapy-related acute myeloid leukaemia. We propose that Allo-HSCT in HIV patients is safe and that alemtuzumab-based conditioning could further work to eradicate HIV in those whose donor is not CCR5 homozygous.


Alemtuzumab/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Graft vs Leukemia Effect/drug effects , Hematopoietic Stem Cell Transplantation/adverse effects , Leukemia, Myeloid, Acute/drug therapy , Melphalan/therapeutic use , Transplantation Conditioning/adverse effects , Vidarabine/analogs & derivatives , Adult , Alemtuzumab/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Hematopoietic Stem Cell Transplantation/methods , Humans , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/pathology , Male , Melphalan/pharmacology , Transplantation Conditioning/methods , Vidarabine/pharmacology , Vidarabine/therapeutic use
16.
Biol Blood Marrow Transplant ; 24(10): 2034-2039, 2018 10.
Article En | MEDLINE | ID: mdl-29908231

More than half of patients undergoing hematopoietic cell transplantation at our institution are ethnic or racial minorities, making the search for matched unrelated donors more challenging. Since the introduction of haploidentical bone marrow transplant (haplo-BMT) into our pediatric BMT program in 2015, 69.2% of recipients have been minorities. Herein, we describe our experience with the first 13 pediatric and young adult patients with hematologic malignancies who have undergone T cell-replete haplo-BMT after myeloablative conditioning (MAC) at our institution. We have previously documented that in experimental haplo-BMT, post-transplant bendamustine (PT-BEN) is at least as effective as post-transplant cyclophosphamide (PT-CY) against graft-versus-host disease (GVHD) and elicits superior graft-versus-leukemia (GVL) effects. We report on, for the first time in humans, 4 patients treated with PT-CY and PT-BEN after haplo-BMT as part of our ongoing institutional phase I/II study (NCT02996773). The remaining 9 patients reviewed in this report received PT-CY. Our findings indicate that MAC haplo-BMT is well tolerated by children and young adults with advanced hematologic malignancies with no observed nonrelapse mortality or grades III to IV GVHD. All patients who underwent haplo-BMT remain alive and disease-free with a median follow-up of 15.6 months (range, 1.5 to 31.2). Preliminary findings from our ongoing clinical trial demonstrate that partial substitution of PT-BEN for PT-CY is feasible and safe after haplo-BMT as an immune modulatory strategy to alleviate GVHD and potentially more effectively preserve GVL.


Bendamustine Hydrochloride/administration & dosage , Bone Marrow Transplantation , Cyclophosphamide/administration & dosage , Graft vs Host Disease/prevention & control , Hematologic Neoplasms/therapy , Unrelated Donors , Adolescent , Adult , Allografts , Child , Child, Preschool , Female , Graft vs Leukemia Effect/drug effects , Hematopoietic Stem Cell Transplantation , Humans , Male , Transplantation Conditioning , Transplantation, Haploidentical , Young Adult
17.
Clin Cancer Res ; 24(7): 1604-1616, 2018 04 01.
Article En | MEDLINE | ID: mdl-29367429

Purpose: Allogeneic bone marrow transplantation (BMT) provides curative therapy for leukemia via immunologic graft-versus-leukemia (GVL) effects. In practice, this must be balanced against life threatening pathology induced by graft-versus-host disease (GVHD). Recipient dendritic cells (DC) are thought to be important in the induction of GVL and GVHD.Experimental Design: We have utilized preclinical models of allogeneic BMT to dissect the role and modulation of recipient DCs in controlling donor T-cell-mediated GVHD and GVL.Results: We demonstrate that recipient CD8α+ DCs promote activation-induced clonal deletion of allospecific donor T cells after BMT. We compared pretransplant fms-like tyrosine kinase-3 ligand (Flt-3L) treatment to the current clinical strategy of posttransplant cyclophosphamide (PT-Cy) therapy. Our results demonstrate superior protection from GVHD with the immunomodulatory Flt-3L approach, and similar attenuation of GVL responses with both strategies. Strikingly, Flt-3L treatment permitted maintenance of the donor polyclonal T-cell pool, where PT-Cy did not.Conclusions: These data highlight pre-transplant Flt-3L therapy as a potent new therapeutic strategy to delete alloreactive T cells and prevent GVHD, which appears particularly well suited to haploidentical BMT where the control of infection and the prevention of GVHD are paramount. Clin Cancer Res; 24(7); 1604-16. ©2018 AACR.


CD8 Antigens/immunology , Cyclophosphamide/pharmacology , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Membrane Proteins/immunology , T-Lymphocytes/immunology , Animals , Bone Marrow Transplantation/methods , Dendritic Cells/drug effects , Female , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/drug effects , Leukemia/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/drug effects , Tissue Donors , Transplantation, Homologous/methods
18.
Front Immunol ; 9: 3003, 2018.
Article En | MEDLINE | ID: mdl-30627129

Allogeneic hematopoietic cell transplantation (allo-HCT) is an effective immunotherapeutic approach for various hematologic and immunologic ailments. Despite the beneficial impact of allo-HCT, its adverse effects cause severe health concerns. After transplantation, recognition of host cells as foreign entities by donor T cells induces graft-vs.-host disease (GVHD). Activation, proliferation and trafficking of donor T cells to target organs and tissues are critical steps in the pathogenesis of GVHD. T cell activation is a synergistic process of T cell receptor (TCR) recognition of major histocompatibility complex (MHC)-anchored antigen and co-stimulatory/co-inhibitory signaling in the presence of cytokines. Most of the currently used therapeutic regimens for GVHD are based on inhibiting the allogeneic T cell response or T-cell depletion (TCD). However, the immunosuppressive drugs and TCD hamper the therapeutic potential of allo-HCT, resulting in attenuated graft-vs.-leukemia (GVL) effect as well as increased vulnerability to infection. In view of the drawback of overbroad immunosuppression, co-stimulatory, and co-inhibitory molecules are plausible targets for selective modulation of T cell activation and function that can improve the effectiveness of allo-HCT. Therefore, this review collates existing knowledge of T cell co-stimulation and co-inhibition with current research that may have the potential to provide novel approaches to cure GVHD without sacrificing the beneficial effects of allo-HCT.


Costimulatory and Inhibitory T-Cell Receptors/metabolism , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Signal Transduction/immunology , T-Lymphocytes/immunology , Clinical Trials as Topic , Costimulatory and Inhibitory T-Cell Receptors/agonists , Costimulatory and Inhibitory T-Cell Receptors/antagonists & inhibitors , Costimulatory and Inhibitory T-Cell Receptors/immunology , Graft vs Host Disease/therapy , Graft vs Leukemia Effect/drug effects , Graft vs Leukemia Effect/immunology , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunotherapy/methods , Major Histocompatibility Complex/immunology , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transplantation, Homologous/adverse effects , Treatment Outcome
19.
Dermatology ; 233(4): 303-313, 2017.
Article En | MEDLINE | ID: mdl-29232687

BACKGROUND: Cure of acute leukemia after transplantation is mediated by the grafted cells. We investigated the graft-versus-leukemia effect (GVL) in patients with cutaneous acute graft-versus-host disease (GVHD) treated with photochemotherapy (psoralen and ultraviolet light type A). METHOD: Forty-seven patients with acute leukemia were followed 5,000 days after transplantation to assess survival and GVL by multivariate analysis. The primary predictor was time to treatment of cutaneous acute GVHD by photochemotherapy separated into treatment start during the first week of acute GVHD versus after the first week of acute GVHD. RESULTS: Photochemotherapy started after the first week of acute GVHD predicted GVL with a hazard ratio (HR) of 3.94 (95% confidence interval, CI, 1.67-9.33, p = 0.0018) and survival with preserved GVL with an HR of 2.63 (95% CI 1.30-5.32, p = 0.007). The effects on GVL and survival with preserved GVL were present regardless of whether the patients were transplanted in remission or relapse (p < 0.05). Chronic GVHD came earlier in the group that started photochemotherapy after 1 week of acute GHVD, but chronic GVHD did not increase the GVL. CONCLUSION: The timing of photochemotherapy after cutaneous acute GVHD may direct the GVL and predict long-term leukemia-free survival.


Ficusin/therapeutic use , Graft vs Leukemia Effect/drug effects , Immunity, Innate , Leukemia/therapy , Photochemotherapy/methods , Acute Disease , Adolescent , Adult , Child , Child, Preschool , Disease-Free Survival , Female , Hematopoietic Stem Cell Transplantation , Humans , Infant , Leukemia/immunology , Leukemia/mortality , Male , Middle Aged , Photosensitizing Agents/therapeutic use , Survival Rate/trends , Sweden/epidemiology , Time Factors , Ultraviolet Rays , Young Adult
20.
J Clin Invest ; 127(7): 2765-2776, 2017 Jun 30.
Article En | MEDLINE | ID: mdl-28604385

The graft-versus-leukemia (GVL) effect in allogeneic hematopoietic stem cell transplantation (alloSCT) is potent against chronic phase chronic myelogenous leukemia (CP-CML), but blast crisis CML (BC-CML) and acute myeloid leukemias (AML) are GVL resistant. To understand GVL resistance, we studied GVL against mouse models of CP-CML, BC-CML, and AML generated by the transduction of mouse BM with fusion cDNAs derived from human leukemias. Prior work has shown that CD4+ T cell-mediated GVL against CP-CML and BC-CML required intact leukemia MHCII; however, stem cells from both leukemias were MHCII negative. Here, we show that CP-CML, BC-CML, and AML stem cells upregulate MHCII in alloSCT recipients. Using gene-deficient leukemias, we determined that BC-CML and AML MHC upregulation required IFN-γ stimulation, whereas CP-CML MHC upregulation was independent of both the IFN-γ receptor (IFN-γR) and the IFN-α/ß receptor IFNAR1. Importantly, IFN-γR-deficient BC-CML and AML were completely resistant to CD4- and CD8-mediated GVL, whereas IFN-γR/IFNAR1 double-deficient CP-CML was fully GVL sensitive. Mouse AML and BC-CML stem cells were MHCI+ without IFN-γ stimulation, suggesting that IFN-γ sensitizes these leukemias to T cell killing by mechanisms other than MHC upregulation. Our studies identify the requirement of IFN-γ stimulation as a mechanism for BC-CML and AML GVL resistance, whereas independence from IFN-γ renders CP-CML more GVL sensitive, even with a lower-level alloimmune response.


Graft vs Leukemia Effect/drug effects , Interferon-gamma/pharmacology , Leukemia, Myeloid/drug therapy , Neoplasms, Experimental/drug therapy , Neoplastic Stem Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Graft vs Leukemia Effect/genetics , Graft vs Leukemia Effect/immunology , Humans , Interferon-gamma/immunology , Leukemia, Myeloid/genetics , Leukemia, Myeloid/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplastic Stem Cells/pathology
...