Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Leukemia ; 37(9): 1842-1849, 2023 09.
Article in English | MEDLINE | ID: mdl-37507426

ABSTRACT

Intensive induction chemotherapy achieves complete remissions (CR) in >60% of patients with acute myeloid leukemia (AML) but overall survival (OS) is poor for relapsing patients not eligible for allogeneic hematopoietic stem cell transplantation (allo-HSCT). Oral azacytidine may be used as maintenance treatment in AML in first remission, but can be associated with substantial side effects, and less toxic strategies should be explored. Twenty AML patients in first CR (CR1) ineligible for allo-HSCT were treated with FDC101, an autologous RNA-loaded mature dendritic cell (mDC) vaccine expressing two leukemia-associated antigens (LAAs). Each dose consisted of 2.5-5 × 106 mDCs per antigen, given weekly until week 4, at week 6, and then monthly, during the 2-year study period. Patients were followed for safety and long-term survival. Treatment was well tolerated, with mild and transient injection site reactions. Eleven of 20 patients (55%) remained in CR, while 4 of 6 relapsing patients achieved CR2 after salvage therapy and underwent allo-HSCT. OS at five years was 75% (95% CI: 50-89), with 70% of patients ≥60 years of age being long-term survivors. Maintenance therapy with this DC vaccine was well tolerated in AML patients in CR1 and was accompanied by encouraging 5-year long-term survival.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Humans , Induction Chemotherapy , Transplantation, Homologous , Leukemia, Myeloid, Acute/therapy , Remission Induction , Recurrence , Dendritic Cells , Retrospective Studies , Antigens, Neoplasm , WT1 Proteins/genetics
2.
Clin Transl Immunology ; 9(3): e1117, 2020.
Article in English | MEDLINE | ID: mdl-32153780

ABSTRACT

OBJECTIVES: Innovative post-remission therapies are needed to eliminate residual AML cells. DC vaccination is a promising strategy to induce anti-leukaemic immune responses. METHODS: We conducted a first-in-human phase I study using TLR7/8-matured DCs transfected with RNA encoding the two AML-associated antigens WT1 and PRAME as well as CMVpp65. AML patients in CR at high risk of relapse were vaccinated 10× over 26 weeks. RESULTS: Despite heavy pretreatment, DCs of sufficient number and quality were generated from a single leukapheresis in 11/12 cases, and 10 patients were vaccinated. Administration was safe and resulted in local inflammatory responses with dense T-cell infiltration. In peripheral blood, increased antigen-specific CD8+ T cells were seen for WT1 (2/10), PRAME (4/10) and CMVpp65 (9/10). For CMVpp65, increased CD4+ T cells were detected in 4/7 patients, and an antibody response was induced in 3/7 initially seronegative patients. Median OS was not reached after 1057 days; median RFS was 1084 days. A positive correlation was observed between clinical benefit and younger age as well as mounting of antigen-specific immune responses. CONCLUSIONS: Administration of TLR7/8-matured DCs to AML patients in CR at high risk of relapse was feasible and safe and resulted in induction of antigen-specific immune responses. Clinical benefit appeared to occur more likely in patients <65 and in patients mounting an immune response. Our observations need to be validated in a larger patient cohort. We hypothesise that TLR7/8 DC vaccination strategies should be combined with hypomethylating agents or checkpoint inhibition to augment immune responses. TRIAL REGISTRATION: The study was registered at https://clinicaltrials.gov on 17 October 2012 (NCT01734304) and at https://www.clinicaltrialsregister.eu (EudraCT-Number 2010-022446-24) on 10 October 2013.

3.
Front Immunol ; 9: 385, 2018.
Article in English | MEDLINE | ID: mdl-29535740

ABSTRACT

Immune checkpoint inhibition has been shown to successfully reactivate endogenous T cell responses directed against tumor-associated antigens, resulting in significantly prolonged overall survival in patients with various tumor entities. For malignancies with low endogenous immune responses, this approach has not shown a clear clinical benefit so far. Therapeutic vaccination, particularly dendritic cell (DC) vaccination, is a strategy to induce T cell responses. Interaction of DCs and T cells is dependent on receptor-ligand interactions of various immune checkpoints. In this study, we analyzed the influence of blocking antibodies targeting programmed cell death protein 1 (PD-1), HVEM, CD244, TIM-3, and lymphocyte activation gene 3 (LAG-3) on the proliferation and cytokine secretion of T cells after stimulation with autologous TLR-matured DCs. In this context, we found that LAG-3 blockade resulted in superior T cell activation compared to inhibition of other pathways, including PD-1/PD-L1. This result was consistent across different methods to measure T cell stimulation (proliferation, IFN-γ secretion), various stimulatory antigens (viral and bacterial peptide pool, specific viral antigen, specific tumor antigen), and seen for both CD4+ and CD8+ T cells. Only under conditions with a weak antigenic stimulus, particularly when combining antigen presentation by peripheral blood mononuclear cells with low concentrations of peptides, we observed the highest T cell stimulation with dual blockade of LAG-3 and PD-1 blockade. We conclude that priming of novel immune responses can be strongly enhanced by blockade of LAG-3 or dual blockade of LAG-3 and PD-1, depending on the strength of the antigenic stimulus.


Subject(s)
Antibodies, Blocking/pharmacology , Antigen-Presenting Cells/immunology , Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Programmed Cell Death 1 Receptor/immunology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Toll-Like Receptors/metabolism , Lymphocyte Activation Gene 3 Protein
4.
J Hematol Oncol ; 8: 93, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26219463

ABSTRACT

BACKGROUND: T cell function is crucial for the success of several novel immunotherapeutic strategies for the treatment of acute myeloid leukemia (AML). However, changes in phenotype and function of T cells have been described in various hematologic malignancies, mimicking T cell exhaustion known from chronic viral infections. Detailed knowledge about phenotype and function of T cells in AML patients at different stages of the disease is indispensable for optimal development and application of immunotherapeutic strategies for this disease. METHODS: We used flow cytometry-based assays to characterize T cell phenotype and function in peripheral blood and bone marrow of AML patients at diagnosis, at relapse after intensive chemotherapy, and at relapse after allogeneic stem cell transplantation (SCT). Surface expression of CD244, PD-1, CD160, and TIM-3 was determined, and proliferation and production of IFN-γ, TNF-α, and IL-2 were measured. RESULTS: We detected similar expression of inhibitory molecules on T cells from patients at diagnosis and from age-matched healthy controls. At relapse after SCT, however, PD-1 expression was significantly increased compared to diagnosis, both on CD4(+) and CD8(+) T cells. This pattern was not associated with age and cytomegalovirus (CMV) status but with a shift towards effector memory cells in relapsed AML patients. Proliferation and cytokine production assays did not reveal functional defects in T cells of AML patients, neither at diagnosis nor at relapse. CONCLUSION: We thus conclude that T cell exhaustion does not play a major role in AML. Immunotherapeutic strategies targeting autologous T cells thus have particularly good prospects in the setting of AML.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Leukemia, Myeloid, Acute/immunology , Programmed Cell Death 1 Receptor/metabolism , Female , Humans , Leukemia, Myeloid, Acute/genetics , Male , Programmed Cell Death 1 Receptor/immunology , Recurrence
5.
Immunotherapy ; 5(1): 63-78, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23256799

ABSTRACT

The prognosis of acute myeloid leukemia, particularly when associated with adverse chromosomal or molecular aberrations, is poor due to a high relapse rate after induction chemotherapy. Postremission therapy for elimination of minimal residual disease remains a major challenge. Allogeneic hematopoietic stem cell transplantation has proven to provide a potent antileukemic effect. Novel strategies are needed for patients ineligible for this treatment. Here current immunotherapeutic concepts in acute myeloid leukemia in a nonallogeneic hematopoietic stem cell transplantation setting are reviewed. Data gathered with different monoclonal antibodies are discussed. Adoptive transfer of NK and T cells is reviewed, including evolving data on T-cell engineering. Results of systemic cytokine administration and of therapeutic vaccinations with peptides, modified leukemic cells and dendritic cells are presented. One particular focus of this review is the integration of currently running clinical trials. Recent immunotherapeutic studies have been encouraging and further interesting results are to be expected.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Immunotherapy/methods , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Antibodies, Monoclonal, Murine-Derived , Cytokines/immunology , Cytokines/therapeutic use , Dendritic Cells/immunology , Humans , Immunotherapy, Adoptive , T-Lymphocytes/immunology
6.
J Immunol ; 187(8): 3911-7, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21969315

ABSTRACT

MicroRNAs (miRNAs) are an important class of cellular regulators that modulate gene expression and thereby influence cell fate and function. In the immune system, miRNAs act at checkpoints during hematopoietic development and cell subset differentiation, they modulate effector cell function, and they are implicated in the maintenance of homeostasis. Dendritic cells (DCs), the professional APCs involved in the coordination of adaptive immune responses, are also regulated by miRNAs. Some DC-relevant miRNAs, including miR-155 and miR-146a, are shared with other immune cells, whereas others have been newly identified. In this review, we summarize the current understanding of where miRNAs are active during DC development from myeloid precursors and differentiation into specialized subsets, and which miRNAs play roles in DC function.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , MicroRNAs/immunology , Animals , Humans
7.
Mol Immunol ; 48(1-3): 333-40, 2010.
Article in English | MEDLINE | ID: mdl-20822813

ABSTRACT

microRNAs have emerged as a novel layer of regulation of cellular development and function, including cells of the immune system. microRNA expression profiles and function of several microRNAs have been elucidated in granulocyte macrophage colony-stimulating factor derived dendritic cells (GM-CSF DC). In this study we determined the microRNA expression profile from plasmacytoid DC (pDC) and conventional DC (cDC) generated in murine FMS-related tyrosine kinase 3 ligand (Flt3L) bone marrow culture. We observed distinct miRNA expression signatures in these two different DC subsets and found that pDC were closer related to CD4(+) T cells than to cDC. Expression of a selected subset of microRNAs was also compared between cDC and GM-CSF DC. Furthermore, we show that inhibition of two differentially expressed microRNAs, miR-221 and miR-222, during differentiation resulted in skewed pDC/cDC ratios. Among the confirmed or potential targets for miR-221 and miR-222 are c-Kit, p27(kip1) and E2-2. While c-Kit is expressed by DC progenitors and p27(kip1) is a cell cycle regulator, E2-2 does transcriptionally regulate pDC development. Our data demonstrate that microRNAs can influence Flt3-driven DC differentiation.


Subject(s)
Cell Differentiation/genetics , Dendritic Cells/cytology , MicroRNAs , Animals , Cell Differentiation/immunology , Cell Line , Cell Separation , Flow Cytometry , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transfection
8.
J Immunol ; 185(1): 400-9, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20530258

ABSTRACT

Dendritic cells (DCs) are central for the induction of T cell immunity and tolerance. Fundamental for DCs to control the immune system is their differentiation from precursors into various DC subsets with distinct functions and locations in lymphoid organs and tissues. In contrast to the differentiation of epidermal Langerhans cells (LCs) and their seeding into the epidermis, LC maturation, turnover, and MHC class II Ag presentation capacities are strictly dependent on the presence of Dicer, which generates mature microRNAs (miRNAs). Absence of miRNAs caused a strongly disturbed steady-state homeostasis of LCs by increasing their turnover and apoptosis rate, leading to progressive ablation of LCs with age. The failure to maintain LCs populating the epidermis was accompanied by a proapoptotic gene expression signature. Dicer-deficient LCs showed largely increased cell sizes and reduced expression levels of the C-type lectin receptor Langerin, resulting in the lack of Birbeck granules. In addition, LCs failed to properly upregulate MHC class II, CD40, and CD86 surface molecules upon stimulation, which are critical hallmarks of functional DC maturation. This resulted in inefficient induction of CD4 T cell proliferation, whereas Dicer-deficient LCs could properly stimulate CD8 T cells. Taken together, Dicer-dependent generation of miRNAs affects homeostasis and function of epidermal LCs.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/immunology , DEAD-box RNA Helicases/genetics , Endoribonucleases/genetics , Langerhans Cells/cytology , Langerhans Cells/immunology , MicroRNAs/physiology , Animals , Apoptosis/genetics , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/genetics , Cell Movement/immunology , Cell Survival/genetics , Cell Survival/immunology , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Endoribonucleases/deficiency , Endoribonucleases/physiology , Gene Expression Regulation/immunology , Homeostasis/genetics , Homeostasis/immunology , Langerhans Cells/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Ribonuclease III
SELECTION OF CITATIONS
SEARCH DETAIL
...