Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 21(1)2019 12 25.
Article in English | MEDLINE | ID: mdl-31881723

ABSTRACT

Myelodysplastic syndrome (MDS) defines a group of heterogeneous hematologic malignancies that often progresses to acute myeloid leukemia (AML). The leading treatment for high-risk MDS patients is azacitidine (Aza, Vidaza®), but a significant proportion of patients are refractory and all patients eventually relapse after an undefined time period. Therefore, new therapies for MDS are urgently needed. We present here evidence that acadesine (Aca, Acadra®), a nucleoside analog exerts potent anti-leukemic effects in both Aza-sensitive (OCI-M2S) and resistant (OCI-M2R) MDS/AML cell lines in vitro. Aca also exerts potent anti-leukemic effect on bone marrow cells from MDS/AML patients ex-vivo. The effect of Aca on MDS/AML cell line proliferation does not rely on apoptosis induction. It is also noteworthy that Aca is efficient to kill MDS cells in a co-culture model with human medullary stromal cell lines, that mimics better the interaction occurring in the bone marrow. These initial findings led us to initiate a phase I/II clinical trial using Acadra® in 12 Aza refractory MDS/AML patients. Despite a very good response in one out 4 patients, we stopped this trial because the highest Aca dose (210 mg/kg) caused serious renal side effects in several patients. In conclusion, the side effects of high Aca doses preclude its use in patients with strong comorbidities.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Ribonucleosides/therapeutic use , Aged , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Apoptosis/drug effects , Azacitidine/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Clinical Trials as Topic , Drug Resistance, Neoplasm/drug effects , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/pathology , Recurrence , Ribonucleosides/pharmacology , Treatment Failure
2.
Oncotarget ; 8(29): 47103-47109, 2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28514758

ABSTRACT

Azacitidine (AZA), the reference treatment for most higher-risk myelodysplastic (MDS) patients can also improve overall survival (OS) in elderly acute myeloid leukemia (AML) patients ineligible for intensive chemotherapy, but reliable biological markers predicting response and OS in patients treated with AZA are lacking. In a preliminary study, we found that an increase of the percentage of BCL2L10, an anti-apoptotic member of the bcl-2 family, was correlated with AZA resistance. In this study, we assessed prospectively by flow cytometry the prognostic value of BCL2L10 positive bone marrow mononuclear cells in 70 patients (42 MDS and 28 AML), prior to AZA treatment.In patients with baseline marrow blasts below 30%, the baseline percentage of bone marrow BCL2L10 positive cells inversely correlated with response to AZA and OS independently of the International Prognostic Scoring System (IPSS) and IPSS-revised (IPSS-R). Specifically, OS was significantly lower in patients with more than 10% BCL2L10 positive cells (median 8.3 vs 22.9 months in patients with less than 10% positivity, p = 0,001). In summary, marrow BCL2L10 positive cells may be a biomarker for azacitidine response and OS, with a potential impact in clinical practice.


Subject(s)
Bone Marrow Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/mortality , Proto-Oncogene Proteins c-bcl-2/metabolism , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , Biomarkers , Bone Marrow Cells/pathology , Female , Gene Expression , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/drug therapy , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins c-bcl-2/genetics , Treatment Outcome
3.
Diabetes ; 65(10): 3062-74, 2016 10.
Article in English | MEDLINE | ID: mdl-27388216

ABSTRACT

Activation of the p53 pathway in adipose tissue contributes to insulin resistance associated with obesity. However, the mechanisms of p53 activation and the effect on adipocyte functions are still elusive. Here we found a higher level of DNA oxidation and a reduction in telomere length in adipose tissue of mice fed a high-fat diet and an increase in DNA damage and activation of the p53 pathway in adipocytes. Interestingly, hallmarks of chronic DNA damage are visible at the onset of obesity. Furthermore, injection of lean mice with doxorubicin, a DNA damage-inducing drug, increased the expression of chemokines in adipose tissue and promoted its infiltration by proinflammatory macrophages and neutrophils together with adipocyte insulin resistance. In vitro, DNA damage in adipocytes increased the expression of chemokines and triggered the production of chemotactic factors for macrophages and neutrophils. Insulin signaling and effect on glucose uptake and Glut4 translocation were decreased, and lipolysis was increased. These events were prevented by p53 inhibition, whereas its activation by nutlin-3 reproduced the DNA damage-induced adverse effects. This study reveals that DNA damage in obese adipocytes could trigger p53-dependent signals involved in alteration of adipocyte metabolism and secretory function leading to adipose tissue inflammation, adipocyte dysfunction, and insulin resistance.


Subject(s)
Adipocytes/metabolism , DNA Damage/genetics , Tumor Suppressor Protein p53/metabolism , 3T3-L1 Cells , Animals , Blotting, Western , Chemotaxis/genetics , Chemotaxis/physiology , DNA Damage/physiology , Flow Cytometry , Fluorescent Antibody Technique , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Mice , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Telomere/genetics , Tumor Suppressor Protein p53/genetics
4.
Oncotarget ; 7(18): 26120-36, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27027430

ABSTRACT

Differentiation-inducing factor (DIF) defines a group of chlorinated hexaphenones that orchestrate stalk-cell differentiation in the slime mold Dictyostelium discoideum (DD). DIF-1 and 3 have also been reported to have tumor inhibiting properties; however, the mechanisms that underlie the effects of these compounds remain poorly defined. Herein, we show that DIF-3 rapidly triggers Ca2+ release and a loss of mitochondrial membrane potential (MMP) in the absence of cytochrome c and Smac release and without caspase activation. Consistently with these findings, we also detected no evidence of apoptosis in cells treated with DIF-3 but instead found that this compound induced autophagy. In addition, DIF-3 promoted mitochondrial fission in K562 and HeLa cells, as assessed by electron and confocal microscopy analysis. Importantly, DIF-3 mediated the phosphorylation and redistribution of dynamin-related protein 1 (DRP1) from the cytoplasmic to the microsomal fraction of K562 cells. Pharmacological inhibition or siRNA silencing of DRP1 not only inhibited mitochondrial fission but also protected K562 cells from DIF-3-mediated cell death. Furthermore, DIF-3 potently inhibited the growth of imatinib-sensitive and imatinib-resistant K562 cells. It also inhibited tumor formation in athymic mice engrafted with an imatinib-resistant CML cell line. Finally, DIF-3 exhibited a clear selectivity toward CD34+ leukemic cells from CML patients, compared with CD34- cells. In conclusion, we show that the potent anti-leukemic effect of DIF-3 is mediated through the induction of mitochondrial fission and caspase-independent cell death. Our findings may have important therapeutic implications, especially in the treatment of tumors that exhibit defects in apoptosis regulation.


Subject(s)
Apoptosis/drug effects , GTP Phosphohydrolases/metabolism , Gene Expression Regulation, Leukemic/drug effects , Hexanones/pharmacology , Leukemia/pathology , Microtubule-Associated Proteins/metabolism , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/metabolism , Reactive Oxygen Species/metabolism , Animals , Biomarkers, Tumor/metabolism , Caspases/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dynamins , Female , Humans , K562 Cells , Leukemia/drug therapy , Leukemia/metabolism , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Phosphorylation/drug effects , Xenograft Model Antitumor Assays
5.
Invest Ophthalmol Vis Sci ; 56(11): 6456-66, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26447979

ABSTRACT

PURPOSE: To evaluate the therapeutic potential of Col-Treg, a collagen II-specific type 1 regulatory T-cell immunotherapy for the treatment of noninfectious uveitis (NIU). METHODS: Col-Treg cells were produced from collagen II-specific T cell receptor (TCR) transgenic mice or peripheral blood of healthy donors. Phenotypic characterization was performed by flow cytometry, and cytokine secretion was evaluated with Flowcytomix or ELISA. In vitro functional characterization included ATP hydrolysis, cytotoxicity, and contact-independent T-cell suppression and plasticity assays. Col-Treg migration was assessed by quantitative PCR specific to Col-Treg TCR. Col-Treg cells were administered intravenously in mice displaying experimental autoimmune uveitis (EAU) induced by interphotoreceptor retinoid-binding protein (IRBP) immunizations. Efficacy of Col-Treg was assessed by ophthalmology, histology, and immunohistochemistry. RESULTS: Mice Col-Treg cells displayed identity features of type 1 Treg cells with expression of CD25, FoxP3, low surface expression of CD127, and cytokine secretion profile (IL-10(high), IL-4(low), IFN-γ(int)). In vitro functional assays demonstrated Col-Treg suppressive capacity via soluble factor-dependent immunosuppression, cytotoxicity, and ATP hydrolysis. Col-Treg cells expressed granzyme B, CD39, and glucocorticoid-induced TNF-related protein (GITR). Administration of Col-Treg in EAU mice inhibited clinical and morphologic signs of uveitis and decreased ocular leukocyte infiltration. Col-Treg cells homed in the ocular tissues 24 hours after intravenous injection. Human Col-Treg cells were comparable to mice Col-Treg cells in identity and function and did not show the capacity to differentiate into Th17 cells in vitro. CONCLUSIONS: These results demonstrate the therapeutic potential of Col-Treg cells as a targeted approach for the treatment of NIU and the feasibility of translating this approach to the human clinical setting.


Subject(s)
Collagen Type II/administration & dosage , Immunity, Cellular , Immunotherapy/methods , T-Lymphocytes, Regulatory/immunology , Uveitis/therapy , Animals , Collagen Type II/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Injections, Intravenous , Mice , Mice, Inbred DBA , Mice, Transgenic , Uveitis/immunology , Uveitis/pathology
6.
Oncoscience ; 2(1): 44-9, 2015.
Article in English | MEDLINE | ID: mdl-25815361

ABSTRACT

Hairy cell leukemia (HCL) is a chronic B-cell lymphoproliferative disorder that accounts for 2% of all leukemia. Recent identification of the recurrent V600E BRAF mutation in a majority of HCL patients has led some teams to evaluate the clinical potential of vemurafenib, a BRAF V600 specific inhibitor in a limited number of refractory HCL patients. Recently, we published the case of an HCL patient successfully treated with a low dose of vemurafenib. Eight months after the ending of treatment this patient relapsed. We present here the successful retreatment of this patient with a second line of vemurafenib. Our data suggest for the first time that vemurafenib at the dose of 240 mg once a day could be sufficient to maintain a complete hematological remission after an initial induction treatment with low-dose vemurafenib (2 × 240 mg) daily without inducing major toxicity.

8.
Oncotarget ; 3(12): 1557-65, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23238683

ABSTRACT

The advent of tyrosine kinase inhibitor (TKI) therapy has considerably improved the survival of patients suffering chronic myelogenous leukemia (CML). Indeed, inhibition of BCR-ABL by imatinib, dasatinib or nilotinib triggers durable responses in most patients suffering from this disease. Moreover, resistance to imatinib due to kinase domain mutations can be generally circumvented using dasatinib or nilotinib, but the multi-resistant T315I mutation that is insensitive to these TKIs, remains to date a major clinical problem. In this line, ponatinib (AP24534) has emerged as a promising therapeutic option in patients with all kinds of BCR-ABL mutations, especially the T315I one. However and surprisingly, the effect of ponatinib has not been extensively studied on imatinib-resistant CML cell lines. Therefore, in the present study, we used several CML cell lines with different mechanisms of resistance to TKI to evaluate the effect of ponatinib on cell viability, apoptosis and signaling. Our results show that ponatinib is highly effective on both sensitive and resistant CML cell lines, whatever the mode of resistance and also on BaF3 murine B cells carrying native BCR-ABL or T315I mutation. We conclude that ponatinib could be effectively used for all types of TKI-resistant patients.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/antagonists & inhibitors , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridazines/pharmacology , Animals , Apoptosis/drug effects , Benzamides/pharmacology , Cell Survival/drug effects , Dasatinib , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Inhibitory Concentration 50 , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mutation , Piperazines/pharmacology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Thiazoles/pharmacology , Tumor Cells, Cultured
9.
Oncotarget ; 3(12): 1688-99, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23518796

ABSTRACT

Simalikalactone E (SkE) is a quassinoid extracted from a widely used Amazonian antimalarial remedy. Although SkE has previously been shown to have cytostatic and/or cytotoxic activities in some tumor cell lines, its mechanism of action has not yet been characterized. We show here that SkE in the high nanomolar range inhibited the growth of various leukemic and solid tumor cell lines. Importantly, SkE was highly efficient at inhibiting chronic myelogenous leukemia (CML) cells that exhibit constitutive activation of the MAPK pathway and, accordingly, it impaired the phosphorylation of ERK1/2. SkE also abrogated MEK1/2 and B-Raf phosphorylation but had no effect on Ras activity. Moreover, SkE was particularly effective against melanoma cell lines carrying the B-Raf-V600E mutation. Importantly, SkE resensitized the PLX-4032-resistant 451Lu melanoma cell line (451Lu-R) and was more efficient than U0126, a MEK inhibitor, and PLX-4032 (PLX) at inducing the apoptosis of two hairy cell leukemia (HCL) patient samples carrying the B-Raf-V600E mutation. Finally, SkE was as efficient as imatinib at inhibiting tumor formation in a xenograft model of CML cells in athymic mice. In conclusion, we show that SkE, a very potent inhibitor of B-Raf-V600E, is highly effective against cancer cell lines that exhibit constitutive activation of the ERK1/2 pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Quassins/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HEK293 Cells , Humans , K562 Cells , Leukemia, Hairy Cell/enzymology , Leukemia, Hairy Cell/genetics , Leukemia, Hairy Cell/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Nude , Mutation , Phosphorylation , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction/drug effects , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Time Factors , Transfection , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...