Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
2.
J Immunol ; 198(6): 2500-2512, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28159900

ABSTRACT

Acute graft-versus-host disease (aGVHD) continues to be a frequent and devastating complication of allogeneic hematopoietic stem cell transplantation (HSCT), posing as a significant barrier against the widespread use of HSCTs as a curative modality. Recent studies suggested serum/plasma microRNAs (miRs) may predict aGVHD onset. However, little is known about the functional role of circulating miRs in aGVHD. In this article, we show in two independent cohorts that miR-29a expression is significantly upregulated in the serum of allogeneic HSCT patients at aGVHD onset compared with non-aGVHD patients. Serum miR-29a is also elevated as early as 2 wk before time of diagnosis of aGVHD compared with time-matched control subjects. We demonstrate novel functional significance of serum miR-29a by showing that miR-29a binds and activates dendritic cells via TLR7 and TLR8, resulting in the activation of the NF-κB pathway and secretion of proinflammatory cytokines TNF-α and IL-6. Treatment with locked nucleic acid anti-miR-29a significantly improved survival in a mouse model of aGVHD while retaining graft-versus-leukemia effects, unveiling a novel therapeutic target in aGVHD treatment or prevention.


Subject(s)
Dendritic Cells/physiology , Graft vs Host Disease/diagnosis , Graft vs Leukemia Effect/genetics , Hematopoietic Stem Cell Transplantation , MicroRNAs/biosynthesis , Acute Disease , Cohort Studies , Graft vs Host Disease/genetics , Humans , Inflammation/genetics , Interleukin-6/metabolism , MicroRNAs/blood , Middle Aged , NF-kappa B/metabolism , Prognosis , RNA, Small Interfering/genetics , Signal Transduction , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Transplantation, Homologous , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
3.
Biol Blood Marrow Transplant ; 22(1): 71-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26256940

ABSTRACT

Statins possess potent immunomodulatory effects that may play a role in preventing acute graft-versus-host disease (aGVHD) after allogeneic hematopoietic cell transplantation (allo-HCT). We performed a phase II study of atorvastatin for aGVHD prophylaxis when given to allo-HCT recipients and their HLA-matched sibling donors. Atorvastatin (40 mg/day) was administered to sibling donors, beginning 14 days before the anticipated start of stem cell collection. Allo-HCT recipients (n = 40) received atorvastatin (40 mg/day) in addition to standard aGVHD prophylaxis. The primary endpoint was cumulative incidence of grades II to IV aGVHD at day 100. Atorvastatin was well tolerated, with no attributable grades III to IV toxicities in donors or their recipients. Day 100 and 180 cumulative incidences of grades II to IV aGVHD were 30% (95% confidence interval [CI], 17% to 45%) and 40% (95% CI, 25% to 55%), respectively. One-year cumulative incidence of chronic GVHD was 43% (95% CI, 32% to 69%). One-year nonrelapse mortality and relapse incidences were 5.5% (95% CI, .9% to 16.5%) and 38% (95% CI, 18% to 47%), respectively. One-year progression-free and overall survival rates were 54% (95% CI, 38% to 71%) and 82% (95% CI, 69% to 94%). One-year GVHD-free, relapse-free survival was 27% (95% CI, 16% to 47%). These results did not differ from our historical control subjects (n = 96). Although safe and tolerable, the addition of atorvastatin did not appear to provide any benefit to standard GVHD prophylaxis alone.


Subject(s)
Atorvastatin/administration & dosage , Graft vs Host Disease/mortality , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Histocompatibility Testing , Tissue Donors , Acute Disease , Adult , Aged , Allografts , Disease-Free Survival , Female , Graft vs Host Disease/etiology , Humans , Male , Middle Aged , Survival Rate
4.
Cancer Immunol Res ; 3(7): 787-94, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25735952

ABSTRACT

The Epstein-Barr virus (EBV) is an oncogenic, γ-herpesvirus associated with a broad spectrum of disease. Although most immune-competent individuals can effectivley develop efficient adaptive immune responses to EBV, immunocompromised individuals are at serious risk for developing life-threatening diseases, such as Hodgkin lymphoma and posttransplant lymphoproliferative disorder (PTLD). Given the significant morbidity associated with EBV infection in high-risk populations, there is a need to develop vaccine strategies that restore or enhance EBV-specific immune responses. Here, we identify the EBV immediate-early protein BZLF1 as a potential target antigen for vaccine development. Primary tumors from patients with PTLD and a chimeric human-murine model of EBV-driven lymphoproliferative disorder (EBV-LPD) express BZLF1 protein. Pulsing human dendritic cells (DC) with recombinant BZLF1 followed by incubation with autologous mononuclear cells led to expansion of BZLF1-specific CD8(+) T cells in vitro and primed BZLF1-specific T-cell responses in vivo. In addition, vaccination of hu-PBL-SCID mice with BZLF1-transduced DCs induced specific cellular immunity and significantly prolonged survival from fatal EBV-LPD. These findings identify BZLF1 as a candidate target protein in the immunosurveillance of EBV and provide a rationale for considering BZLF1 in vaccine strategies to enhance primary and recall immune responses and potentially prevent EBV-associated diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpesvirus 4, Human/immunology , Immunity, Cellular/immunology , Trans-Activators/immunology , Vaccines, DNA/immunology , Animals , Humans , Mice , Mice, SCID
5.
Oncotarget ; 6(5): 2693-708, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25393910

ABSTRACT

Treatment options for patients with Epstein-Barr Virus-driven lymphoproliferative diseases (EBV-LPD) are limited. Chemo-immunotherapeutic approaches often lead to immune suppression, risk of lethal infection and EBV reactivation, thus it is essential to identify agents that can deliver direct anti-tumor activity while preserving innate and adaptive host immune surveillance. Silvestrol possesses direct anti-tumor activity in multiple hematologic malignancies while causing minimal toxicity to normal mononuclear cells. However, the effects of silvestrol on immune function have not been described. We utilized in vitro and in vivo models of EBV-LPD to simultaneously examine the impact of silvestrol on both tumor and normal immune function. We show that silvestrol induces direct anti-tumor activity against EBV-transformed lymphoblastoid cell lines (LCL), with growth inhibition, decreased expression of the EBV oncogene latent membrane protein-1, and inhibition of the downstream AKT, STAT1 and STAT3 signaling pathways. Silvestrol promoted potent indirect anti-tumor effects by preserving expansion of innate and EBV antigen-specific adaptive immune effector subsets capable of effective clearance of LCL tumor targets in autologous co-cultures. In an animal model of spontaneous EBV-LPD, silvestrol demonstrated significant therapeutic activity dependent on the presence of CD8-positive T-cells. These findings establish a novel immune-sparing activity of silvestrol, justifying further exploration in patients with EBV-positive malignancies.


Subject(s)
Adaptive Immunity/drug effects , Antineoplastic Agents/pharmacology , Epstein-Barr Virus Infections/drug therapy , Herpesvirus 4, Human/drug effects , Immunity, Innate/drug effects , Lymphoproliferative Disorders/drug therapy , Protein Biosynthesis/drug effects , Triterpenes/pharmacology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic/drug effects , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Lymphoproliferative Disorders/virology , Mice, SCID , Proto-Oncogene Proteins c-akt/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Time Factors , Viral Matrix Proteins/metabolism
6.
Blood ; 122(23): 3778-83, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24085765

ABSTRACT

The coexpression of the MLL partial tandem duplication (PTD) and the FLT3 internal tandem duplication (ITD) mutations associate with a poor outcome in cytogenetically normal acute myeloid leukemia (AML). In mice, a double knock-in (dKI) of Mll(PTD/wt) and Flt3(ITD/wt) mutations induces spontaneous AML with an increase in DNA methyltransferases (Dnmt1, 3a, and 3b) and global DNA methylation index, thereby recapitulating its human AML counterpart. We determined that a regulator of Dnmts, miR-29b, is downregulated in bone marrow of dKI AML mice. Bortezomib exerted a dose-dependent increase in miR-29b expression in AML blasts ex vivo, followed by decreased Dnmts, reduced proliferation, and increased apoptosis. In vivo, bortezomib was not active against dKI AML, yet liposomal-encapsulated bortezomib, as a single agent, reversed downregulation of miR-29b in vivo and induced a long-term (90-day) disease-free remission in 80% of dKI AML mice that exhibited high leukemic burden at the start of therapy, yet showed no signs of relapse at autopsy. Taken together, these data support that liposomal bortezomib, as a single agent, eradicates Mll(PTD/wt):Flt3(ITD/wt) AML in mouse and may represent a powerful and potentially curative approach to high-risk human disease.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Myeloid-Lymphoid Leukemia Protein/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Antineoplastic Agents/administration & dosage , Boronic Acids/administration & dosage , Bortezomib , DNA Methylation , Drug Carriers , Humans , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Leukemia, Experimental/therapy , Leukemia, Myeloid, Acute/metabolism , Liposomes , Mice , Mice, Mutant Strains , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Proteasome Inhibitors/administration & dosage , Pyrazines/administration & dosage , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Tandem Repeat Sequences
SELECTION OF CITATIONS
SEARCH DETAIL
...