Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
2.
Rev Neurol (Paris) ; 177(1-2): 100-106, 2021.
Article in English | MEDLINE | ID: mdl-32718469

ABSTRACT

Intravenous immunoglobulins (IVIg) are commonly used for treatment of dysimmune diseases, but they are known to promote thrombotic events. The medical records of patients who received IVIg infusions to treat neuromuscular disorders were retrospectively studied during two periods: the on-demand period (May 2013-January 2015), when patients received anticoagulant prophylaxis based on personal thrombotic risk factors, and the systematic period (May 2015-January 2017), when patients received systematic anticoagulant prophylaxis. Of the 334 total patients included, 19/153 received anticoagulant prophylaxis in the on-demand period, and 181 were treated in the systematic period. In the on-demand period, thrombosis occurred in three patients (1.96%) as one central retinal artery occlusion, one pulmonary embolism, and one brachiocephalic vein thrombosis. In the systematic period, thrombosis occurred in two patients (1.1%), both as pulmonary embolisms. There was no statistical difference in thrombosis incidence between the periods (P=0.66). The only factor associated with thrombosis was splenectomy (20% versus 0.3% in patients without thrombosis, P=0.03). There were no adverse events due to thromboprophylaxis by low-molecular-weight heparin in either period. Systematic thromboprophylaxis did not significantly reduce the incidence of thrombosis versus thromboprophylaxis based on personal thrombotic risk.


Subject(s)
Pulmonary Embolism , Venous Thromboembolism , Anticoagulants , Humans , Immunoglobulins, Intravenous , Pulmonary Embolism/prevention & control , Retrospective Studies , Venous Thromboembolism/prevention & control
3.
Neth J Med ; 78(1): 34-36, 2020 02.
Article in English | MEDLINE | ID: mdl-32043476

ABSTRACT

Iron deficiency, without anaemia, is common in the general population and induces various symptoms. Its management consists of oral and intravenous supplementation for cases of inefficacy of or intolerance to oral iron. We assessed the efficacy of intravenous iron therapy in non-anaemic iron-deficient patients with fatigue. We prospectively evaluated the level of fatigue, using the Fatigue Severity Scale (FSS), in patients suffering from iron deficiency without anaemia, treated by intravenous iron at the moment of the perfusion (W0), after 4 weeks (W4), and 12 weeks (W12). Of 25 patients, at W0, the mean FFS was 49.3+/-13.7. There was a significant improvement in FSS at W4 (44+/-15; p = 0.01) and a sustained response at W12 with an FFS of 35.8+/-17.1 (p < 0,0001). There was no correlation between FSS and serum ferritin level at W12 (p=0.54) or between serum ferritin at W12 and difference between FSS at W0 and W12 (p=0.58). There were six mild adverse events (24%): asthenia (8%), nausea (8%), headache (4%), local pain (4%); and no serious adverse events. Our results suggest the rapid efficacy of intravenous iron in improving fatigue in iron deficiency without anaemia with a good profile of tolerance.


Subject(s)
Fatigue/drug therapy , Iron Deficiencies , Iron/pharmacology , Administration, Intravenous , Adolescent , Adult , Aged , Aged, 80 and over , Child , Fatigue/complications , Female , Humans , Iron/administration & dosage , Male , Middle Aged , Treatment Outcome , Young Adult
6.
Arch Dermatol Res ; 309(8): 611-623, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28695331

ABSTRACT

SAHA (vorinostat) is a histone deacetylase inhibitor approved by the USA Food and Drug Administration (FDA) for treating advanced refractory cutaneous T cell lymphomas. As SAHA alters the expression of many genes under control of the Sp1 transcription factor, we examined the effect of its association with the FDA-approved anticancer antibiotic Mithramycin A (MTR, plicamycin), a competitive inhibitor of Sp1 binding to DNA. Sézary syndrome (SS) cells, expanded ex vivo from peripheral blood mononuclear cells of 4 patients, were tested for their sensitivity to the drugs regarding cytotoxicity and differential responsive gene expression. Multivariate statistical methods were used to identify genes whose expression is altered by SAHA, MTR, and the synergist effect of the two drugs. MTR, like SAHA, induced the apoptosis of SS cells, while the two drugs in combination showed clear synergy or potentiation. Expression data stressed a likely important role of additive or synergistic epigenetic modifications in the combined effect of the two drugs, while direct inhibition of Sp1-dependent transcription seemed to have only limited impact. Ontological analysis of modified gene expression suggested that the two drugs, either independently or synergistically, counteracted many intertwined pro-survival pathways deregulated in SS cells, resistance of these tumors to intrinsic and extrinsic apoptosis, abnormal adhesion migration, and invasive properties, as well as immunosuppressive behavior. Our findings provide preliminary clues on the individual and combined effects of SAHA and MTR in SS cells and highlight a potential therapeutic interest of this novel pair of drugs for treatment of SS patients.


Subject(s)
Hydroxamic Acids/therapeutic use , Plicamycin/therapeutic use , Sezary Syndrome/drug therapy , Skin Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxamic Acids/administration & dosage , Plicamycin/administration & dosage , Transcriptome , Vorinostat
7.
Clin Genet ; 91(1): 131-136, 2017 01.
Article in English | MEDLINE | ID: mdl-27414984

ABSTRACT

Identifying women at risk of venous thrombosis (VT) under combined oral contraceptives (COC) is a major public health issue. The aim of this study was to investigate in COC users the impact on disease of genetic polymorphisms recently identified to associate with VT risk in the general population. Nine polymorphisms located on KNG1, F11, F5, F2, PROCR, FGG, TSPAN and SLC44A2 genes were genotyped in a sample of 766 patients and 464 controls as part of the PILGRIM (PILl Genetic Risk Monitoring) study. Cases were women who experienced an episode of documented VT during COC use, while controls were women with no history of VT using COC at the time of inclusion. Among the studied polymorphisms, only F11 rs2289252 was significantly associated with VT. The F11 rs2289252-A allele was associated with a 1.6-fold increased risk of VT (p < 0.0001). Besides, the combination of the rs2289252-A allele with non-O blood group, present in 52% of the cohort, was associated with an odds ratio of 4.00 (2.49-6.47; p < 10-4 ). The consideration of this genetic risk factor could help to better assess the risk of VT in COC users.


Subject(s)
Contraceptives, Oral, Combined/administration & dosage , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide , Venous Thrombosis/genetics , Adult , Alleles , Contraceptives, Oral, Combined/adverse effects , Drug Monitoring/methods , Factor XI/genetics , Female , Gene Frequency , Genotype , Humans , Odds Ratio , Risk Factors , Venous Thrombosis/etiology , Young Adult
8.
Blood Cancer J ; 6(9): e469, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27611922

ABSTRACT

The vast majority of patients with acute myeloid leukemia (AML) achieve complete remission (CR) after standard induction chemotherapy. However, the majority subsequently relapse and die of the disease. A leukemia stem cell (LSC) paradigm has been invoked to explain this failure of CR to reliably translate into cure. Indeed, LSCs are highly enriched in CD34+CD38- leukemic cells that exhibit positive aldehyde dehydrogenase activity (ALDH+) on flow cytometry, these LSCs are resistant to currently existing treatments in AML such as cytarabine and anthracycline that, at the cost of great toxicity on normal cells, are highly active against the leukemic bulk, but spare the LSCs responsible for relapse. To try to combat the LSC population selectively, a well-characterized ALDH inhibitor by the trivial name of dimethyl ampal thiolester (DIMATE) was assessed on sorted CD34+CD38- subpopulations from AML patients and healthy patients. ALDH activity and cell viability were monitored by flow cytometry. From enzyme kinetic studies DIMATE is an active enzyme-dependent, competitive, irreversible inhibitor of ALDH1. On cells in culture, DIMATE is a powerful inhibitor of ALDHs 1 and 3, has a major cytotoxic activity on human AML cell lines. Moreover, DIMATE is highly active against leukemic populations enriched in LSCs, but, unlike conventional chemotherapy, DIMATE is not toxic for healthy hematopoietic stem cells which retained, after treatment, their self-renewing and multi-lineage differentiation capacity in immunodeficient mice, xenografted with human leukemic cells. DIMATE eradicates specifically human AML cells and spares healthy mouse hematologic cells.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase/metabolism , Alkynes/pharmacology , Animals , Apoptosis/drug effects , Biomarkers , Caspases/metabolism , Cell Line, Tumor , Cell Survival , Child , Disease Models, Animal , Female , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Male , Mice , Middle Aged , Neoplastic Stem Cells/drug effects , Phenotype , Sulfhydryl Compounds/pharmacology , Xenograft Model Antitumor Assays , Young Adult
9.
Immunol Res ; 64(5-6): 1225-1236, 2016 12.
Article in English | MEDLINE | ID: mdl-27481509

ABSTRACT

Chemotherapies allow complete remission in more than 50 % of patients with acute myeloid leukemia (AML), however, with frequent relapse. This suggests that residual leukemic cells may escape to chemotherapy and immune system. Natural killer (NK) cells from AML patients (AML-NK) have a weaker natural cytotoxicity-activating receptors (NCRs) expression than NK cells from healthy donors (HD-NK). Coding genes for NCR1/NKp46, NCR2/NKp44 and NCR3/NKp30 are located at different loci on two different chromosomes; however, their expression is tightly coordinated. Most NK cells express either high (NCRbright) or low levels (NCRdull) of all three NCRs. This suggests the existence of negative/positive regulation factor(s) common to the three receptors. In order to find transcription factor(s) or pathway(s) involved in NCRs co-regulation, this study compared the transcriptomic signature of HD-NK and AML-NK cells, before and after in vitro NK cells culture. Microarrays analysis revealed a specific NK cells transcriptomic signature in patients with AML. However, in vitro NK cells expansion erased this signature and up-regulated expression of central molecules of NK functions, such as NCR, NKG2D and also ETS-1, regardless of their origin, i.e., AML-NK vs HD-NK. ETS-1 transcription factor was shown to bind to a specific and common region in the NCRs promoters, thus appearing as a good candidate to explain the coordinated regulation of three NCRs. Such results are encouraging regarding in vitro AML-NK cytotoxicity restoration and provide a new conceptual support for innovative cellular therapy based on in vitro NK cells expansion before their reinfusion in AML patients.


Subject(s)
Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , Receptors, Natural Cytotoxicity Triggering/metabolism , Adult , Aged , Aged, 80 and over , Cells, Cultured , Cytotoxicity, Immunologic , Female , Gene Expression Regulation , Humans , Immunophenotyping , Male , Middle Aged , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Receptors, Natural Cytotoxicity Triggering/genetics , Tissue Array Analysis , Transcriptome , Young Adult
10.
Med Mal Infect ; 46(4): 226-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27126350

ABSTRACT

OBJECTIVE: To study the efficacy and safety of micafungin for prophylaxis of invasive fungal infections in patients undergoing induction chemotherapy for acute myeloid leukemia. PATIENTS AND METHODS: A prospective observational single-center study of 41 patients from the hematology department between May 2012 and April 2015. Micafungin was administered once daily from the first day of induction chemotherapy to the end of the neutropenic phase. RESULTS: Neither Candida nor Aspergillus infection was documented in our 41 patients from the first day of micafungin infusion to the end of the neutropenic phase. Patients were followed for three months after discontinuation of micafungin and none of them contracted an invasive fungal infection. Only one patient presented with grade III-IV hepatic and ionic toxicities. CONCLUSION: Micafungin is associated with a good safety profile and is an interesting option for preventing invasive fungal infections in the high-risk population of patients presenting with hematological disorders.


Subject(s)
Antifungal Agents/therapeutic use , Echinocandins/therapeutic use , Leukemia, Myeloid, Acute/complications , Lipopeptides/therapeutic use , Mycoses/prevention & control , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bacterial Infections/etiology , Cytarabine/administration & dosage , Cytarabine/adverse effects , Daunorubicin/administration & dosage , Daunorubicin/adverse effects , Echinocandins/administration & dosage , Febrile Neutropenia/chemically induced , Febrile Neutropenia/complications , Female , Humans , Idarubicin/administration & dosage , Idarubicin/adverse effects , Lipopeptides/administration & dosage , Male , Micafungin , Middle Aged , Prospective Studies , Young Adult
11.
Clin Microbiol Infect ; 20(2): 160-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23607363

ABSTRACT

Cytomegalovirus (CMV) infection is a serious complication that may occur in the weeks or months following bone marrow transplantation. However, both Ganciclovir and the CMV infection itself can cause marrow toxicity, notably neutropenia, that may consequently expose these immunosuppressed patients to life-threatening bacterial and/or fungal infections. The aim of this retrospective study was to identify factors associated with the occurrence of grade III-IV neutropenia among patients receiving pre-emptive Ganciclovir therapy after allogeneic stem cell transplantation at our Institution. We identified 547 consecutive patients transplanted from January 2005 to June 2011 at our Institution. In all, 190 patients (35%) presented with CMV reactivation of whom 30 patients (5%) were excluded from the analysis because they already had neutropenia at the time of reactivation. Finally, 160 (29%) patients were analysed. According to multivariate analysis, at the time of treatment initiation, the risk factors significantly associated with a grade III-IV Ganciclovir-related neutropenia included a high viral load (hazard ratio (HR) = 2.68, 95% CI 1.25-5.737, p 0.01); an absolute neutrophil count >3000 was a protective factor (HR = 0.26, 95% CI 0.125-0.545, p <0001) whereas serum creatinine >2 mg/dL was associated with higher Ganciclovir-related neutropenia (HR = 2.4, 95% CI 1.11-5.17, p 0.002). This large analysis revealed three risk factors for Ganciclovir-related neutropenia among patients with CMV reactivation after allogeneic stem cell transplantation; prompt identification of patients at risk when antiviral therapy is started may allow clinicians to adopt adequate preventive measures, so reducing the morbidity and mortality associated with CMV reactivation.


Subject(s)
Antiviral Agents/adverse effects , Cytomegalovirus Infections/drug therapy , Ganciclovir/adverse effects , Neutropenia/chemically induced , Neutropenia/epidemiology , Adolescent , Adult , Aged , Antiviral Agents/administration & dosage , Female , Ganciclovir/administration & dosage , Humans , Male , Middle Aged , Retrospective Studies , Risk Factors , Stem Cell Transplantation/adverse effects , Transplantation, Homologous/adverse effects , Young Adult
12.
Bone Marrow Transplant ; 48(11): 1472-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23749104

ABSTRACT

Invasive fungal infections (IFIs) such as candidiasis and mold infections have caused significant morbidity and mortality among immunocompromised patients in recent years. Micafungin, a new echinocandin, inhibits fungal cell wall ß-glucan synthesis, with potent activity against most species of Candida and Aspergillus. The aim of this observational study was to investigate the efficacy and safety of micafungin in prophylaxis of IFIs in 26 high-risk adult patients with various hematological diseases receiving haplo-identical Allo-SCT. Only two patients had a history of possible aspergillosis before transplant treated by voriconazole. The patients received a median of four lines (2-7) of treatment before Allo-SCT. Thirteen patients (50%) received at least one prior Auto-SCT; and eight patients (31%) received a previous Allo-SCT. Patients received a median of 29 infusions (range, 15-85) of micafungin (50 mg/day i.v. as a 1-h infusion). The treatment was initiated at the beginning of the transplant conditioning regimen until the hospital discharge. None of our patients discontinued the treatment for drug-related adverse events. Micafungin was not associated with any hepatotoxicity. Only one patient (4%) discontinued the treatment because of early disease progression. In all patients no Candida and/or Aspergillus species was documented after 3 and 6 months from transplant. None of our patients presented a positive galactomannan antigenemia >0.5. Nine patients (35%) presented a CMV reactivation. Four patients presented an acute GVHD grade II and two patients presented a chronic GVHD. The median follow-up was 11 months (3-23). At the last follow-up, there were 20 patients (77%) who were alive; four patients (12%) died because of disease progression and two patients because of graft failure. Micafungin has a good safety and tolerability profile, with an efficacy in preventing IFI in this high-risk population. Our data provide support for an efficacy study in a prophylaxis setting, but prospective and comparative clinical trials using micafungin are urgently needed to define the role of this drug in prophylaxis after haplo-identical Allo-SCT.


Subject(s)
Antifungal Agents/adverse effects , Antifungal Agents/therapeutic use , Echinocandins/adverse effects , Echinocandins/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Lipopeptides/adverse effects , Lipopeptides/therapeutic use , Mycoses/prevention & control , Adult , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Micafungin , Middle Aged , Mycoses/etiology , Transplantation Conditioning/adverse effects , Transplantation Conditioning/methods , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL