Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Eur J Haematol ; 2024 May 07.
Article En | MEDLINE | ID: mdl-38712850

BACKGROUND: IMAGE is a retrospective cohort study of patients enrolled in early access programs (EAPs) in France with relapsed/refractory multiple myeloma (RRMM) receiving isatuximab with pomalidomide and dexamethasone (Isa-Pd). METHODS: Patients aged ≥18 years with RRMM who received ≥1 dose of Isa under the EAPs between July 29, 2019 and August 30, 2020 were included. Effectiveness endpoints included progression-free survival (PFS) and response rates. Verbatim terms for adverse events (AEs) were coded using the Medical Dictionary for Regulatory Activities and not graded for severity. RESULTS: A total of 294 and 299 patients were included in the effectiveness and safety populations, respectively. IMAGE included patients who received one prior line of treatment (10.2%) and were daratumumab-refractory (19.1%). At median follow-up of 14.2 months, median PFS in the effectiveness population was 12.4 months (95% CI 9.0-15.0). Overall response and very good partial response rates were 46.3% and 27.9%, respectively. Subgroup analyses reflected similar results. In the safety population, 26.4% of patients reported at least one AE; the most common any-grade AE was neutropenia (9.4%). CONCLUSION: IMAGE demonstrated Isa-Pd had meaningful effectiveness in median PFS and depth of response and no new safety signals in a real-world context, consistent with clinical trial results.

2.
Nat Commun ; 15(1): 3452, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658543

Mutations in chromatin regulators are widespread in cancer. Among them, the histone H3 lysine 27 methyltransferase Polycomb Repressive Complex 2 (PRC2) shows distinct alterations according to tumor type. This specificity is poorly understood. Here, we model several PRC2 alterations in one isogenic system to reveal their comparative effects. Focusing then on lymphoma-associated EZH2 mutations, we show that Ezh2Y641F induces aberrant H3K27 methylation patterns even without wild-type Ezh2, which are alleviated by partial PRC2 inhibition. Remarkably, Ezh2Y641F rewires the response to PRC2 inhibition, leading to induction of antigen presentation genes. Using a unique longitudinal follicular lymphoma cohort, we further link EZH2 status to abnormal H3K27 methylation. We also uncover unexpected variability in the mutational landscape of successive biopsies, pointing to frequent co-existence of different clones and cautioning against stratifying patients based on single sampling. Our results clarify how oncogenic PRC2 mutations disrupt chromatin and transcription, and the therapeutic vulnerabilities this creates.


Enhancer of Zeste Homolog 2 Protein , Histones , Lymphoma, Follicular , Mutation , Polycomb Repressive Complex 2 , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Lymphoma, Follicular/genetics , Lymphoma, Follicular/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Histones/metabolism , Histones/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Methylation , Chromatin/metabolism , Chromatin/genetics , Transcription, Genetic
3.
Br J Haematol ; 201(6): 1103-1115, 2023 06.
Article En | MEDLINE | ID: mdl-36974007

Lenalidomide maintenance in myeloma is well established. Nevertheless, pomalidomide could provide an alternative. Myeloma patients in first relapse, initially treated in the Intergroupe Francophone du Myélome (IFM) 2009 trial, and subsequently in the IFM 2013-01 phase 2 trial, received four cycles of salvage therapy with pomalidomide plus cyclophosphamide plus dexamethasone (PCD) with transplantation plus 2 PCD consolidation or without transplantation but with 5 PCD and for all patients pomalidomide plus dexamethasone maintenance therapy. This consisted of 28-day cycles of pomalidomide 4 mg daily on days 1-21 and dexamethasone 20 mg weekly until progression. The primary endpoint was an improved response to treatment. A total of 75/100 patients reached therapy. The median follow-up time was 73 months. The median duration of treatment was 23.7 months. One third of patients improved their response from the initiation of treatment: 11%, 19% and 4% to a very good partial response, complete response or stringent complete response respectively. The median progression-free survival time was 33.2 months and the median overall survival time was not reached. Among the 75 patients, the reasons for pomalidomide discontinuation were progressive disease (54%), adverse events (AEs) (30%), investigator discretion (11%) and consent withdrawal (5%). Grade (G) 3/4 haematological AEs included neutropenia (51%) and lymphopenia (35%); G3/4 drug-related non-haematological AEs (>5%) comprised 13% infections. Long-term administration of pomalidomide and dexamethasone is feasible and one third of the patients improved their response.


Multiple Myeloma , Humans , Salvage Therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dexamethasone
5.
Lancet Oncol ; 22(10): 1378-1390, 2021 10.
Article En | MEDLINE | ID: mdl-34529931

BACKGROUND: CASSIOPEIA part 1 showed superior depth of response and significantly improved progression-free survival with daratumumab, bortezomib, thalidomide, and dexamethasone (D-VTd) versus bortezomib, thalidomide, and dexamethasone (VTd) as induction and consolidation in patients with autologous stem-cell transplant (ASCT)-eligible newly diagnosed multiple myeloma. In part 2, we compared daratumumab maintenance versus observation only. METHODS: CASSIOPEIA is a two-part, open-label, randomised, phase 3 trial of patients aged 18-65 years with newly diagnosed multiple myeloma and Eastern Cooperative Oncology Group performance status 0-2, done in 111 European academic and community practice centres. In part 1, patients were randomly assigned (1:1) to induction and consolidation with D-VTd or VTd. Patients still on study who had a partial response or better were randomly assigned (1:1) by an interactive web-response system to daratumumab 16 mg/kg intravenously every 8 weeks (a reduced frequency compared with standard daratumumab long-term dosing) or observation only for up to 2 years. Stratification factors were induction treatment and depth of response in part 1. The part 2 primary endpoint was progression-free survival from second randomisation. This preplanned interim analysis of progression-free survival was done after 281 events and shall be considered the primary analysis of progression-free survival. Sponsor personnel and designees who were involved in the analysis were masked to treatment group until the independent data monitoring committee recommended that the preplanned interim analysis be considered the main analysis of progression-free survival in part 2. Otherwise, treatment assignments were unmasked. The interaction between induction and consolidation and maintenance was tested at a two-sided significance level of 0·05 by a stratified Cox regression model that included the interaction term between maintenance treatment and induction and consolidation treatment. Efficacy analyses were done in the maintenance-specific intention-to-treat population, which comprised all patients who underwent second randomisation. Safety was analysed in all patients in the daratumumab group who received at least one dose and all patients randomly assigned to observation only. This trial is registered with ClinicalTrials.gov, NCT02541383. Long-term follow-up is ongoing and the trial is closed to new participants. FINDINGS: Between May 30, 2016, and June 18, 2018, 886 patients (458 [84%] of 543 in the D-VTd group and 428 [79%] of 542 in the VTd group) were randomly assigned to daratumumab maintenance (n=442) or observation only (n=444). At a median follow-up of 35·4 months (IQR 30·2-39·9) from second randomisation, median progression-free survival was not reached (95% CI not evaluable [NE]-NE) with daratumumab versus 46·7 months (40·0-NE) with observation only (hazard ratio 0·53, 95% CI 0·42-0·68, p<0·0001). A prespecified analysis of progression-free survival results showed a significant interaction between maintenance and induction and consolidation therapy (p<0·0001). The most common grade 3 or 4 adverse events were lymphopenia (16 [4%] of 440 patients in the daratumumab group vs eight [2%] of 444 patients in the observation-only group), hypertension (13 [3%] vs seven [2%]), and neutropenia (nine [2%] vs ten [2%]). Serious adverse events occurred in 100 (23%) patients in the daratumumab group and 84 (19%) patients in the observation-only group. In the daratumumab group, two adverse events led to death (septic shock and natural killer-cell lymphoblastic lymphoma); both were related to treatment. INTERPRETATION: Daratumumab maintenance every 8 weeks for 2 years significantly reduced the risk of disease progression or death compared with observation only. Longer follow-up and other ongoing studies will shed further light on the optimal daratumumab-containing post-ASCT maintenance treatment strategy. FUNDING: Janssen Research & Development, the Intergroupe Francophone du Myélome, and the Dutch-Belgian Cooperative Trial Group for Hematology Oncology.


Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bortezomib/administration & dosage , Dexamethasone/administration & dosage , Multiple Myeloma/therapy , Stem Cell Transplantation , Thalidomide/administration & dosage , Adolescent , Adult , Aged , Antibodies, Monoclonal/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bortezomib/adverse effects , Dexamethasone/adverse effects , Drug Administration Schedule , Europe , Female , Humans , Maintenance Chemotherapy , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Progression-Free Survival , Stem Cell Transplantation/adverse effects , Thalidomide/adverse effects , Time Factors , Transplantation, Autologous , Young Adult
8.
Lancet ; 394(10192): 29-38, 2019 07 06.
Article En | MEDLINE | ID: mdl-31171419

BACKGROUND: Bortezomib, thalidomide, and dexamethasone (VTd) plus autologous stem-cell transplantation is standard treatment in Europe for transplant-eligible patients with newly diagnosed multiple myeloma. We evaluated whether the addition of daratumumab to VTd before and after autologous stem-cell transplantation would improve stringent complete response rate in patients with newly diagnosed multiple myeloma. METHODS: In this two-part, randomised, open-label, phase 3 CASSIOPEIA trial, we recruited transplant-eligible patients with newly diagnosed multiple myeloma at 111 European sites. Patients were randomly assigned (1:1) to receive four pre-transplant induction and two post-transplant consolidation cycles of VTd alone (VTd group) or in combination with daratumumab (D-VTd group). The primary endpoint of part 1 was stringent complete response assessed 100 days after transplantation. Part 2 (maintenance) is ongoing. The trial is registered with ClinicalTrials.gov, number NCT02541383. FINDINGS: Between Sept 22, 2015, and Aug 1, 2017, 1085 patients were enrolled at 111 European sites and were randomly assigned to the D-VTd group (n=543) or the VTd group (n=542). At day 100 after transplantation, 157 (29%) of 543 patients in the D-VTd group and 110 (20%) of 542 patients in the VTd group in the intention-to-treat population had achieved a stringent complete response (odds ratio 1·60, 95% CI 1·21-2·12, p=0·0010). 211 (39%) patients in the D-VTd group versus 141 (26%) in the VTd group achieved a complete response or better, and 346 (64%) of 543 versus 236 (44%) of 542 achieved minimal residual disease-negativity (10-5 sensitivity threshold, assessed by multiparametric flow cytometry; both p<0·0001). Median progression-free survival from first randomisation was not reached in either group (hazard ratio 0·47, 95% CI 0·33-0·67, p<0·0001). 46 deaths on study were observed (14 vs 32, 0·43, 95% CI 0·23-0·80). The most common grade 3 or 4 adverse events were neutropenia (28% vs 15%), lymphopenia (17% vs 10%), and stomatitis (13% vs 16%). INTERPRETATION: D-VTd before and after autologous stem-cell transplantation improved depth of response and progression-free survival with acceptable safety. CASSIOPEIA is the first study showing the clinical benefit of daratumumab plus standard of care in transplant-eligible patients with newly diagnosed multiple myeloma. FUNDING: The Intergroupe Francophone du Myélome and Dutch-Belgian Cooperative Trial Group for Hematology Oncology.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Hematopoietic Stem Cell Transplantation/methods , Multiple Myeloma/therapy , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bortezomib/administration & dosage , Bortezomib/therapeutic use , Chemotherapy, Adjuvant , Combined Modality Therapy , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Survival Analysis , Thalidomide/administration & dosage , Thalidomide/therapeutic use , Transplantation, Autologous , Treatment Outcome , Young Adult
9.
Clin Cancer Res ; 25(14): 4224-4230, 2019 07 15.
Article En | MEDLINE | ID: mdl-31053600

PURPOSE: Carfilzomib is a novel generation proteasome inhibitor. The Carmysap trial demonstrated that twice-weekly KMP (carfilzomib, melphalan, prednisone) might challenge the MPV (melphalan, prednisone, bortezomib) standard. We sought to study KMP weekly, allowing to increase carfilzomib's dose with maintained efficacy and improved safety profile. PATIENTS AND METHODS: IFM2012-03, a phase I multicenter study of KMP weekly in elderly patients with newly diagnosed multiple myeloma (eNDMM), aimed to determine the MTD of carfilzomib. Carfilzomib was given intravenously at 36, 45, 56, and 70 mg/m2/day on days 1, 8, 15, and 22 with melphalan and prednisone, for nine 35-day induction cycles, followed by carfilzomib maintenance for 1 year. Three dose-limiting toxicities (DLT) determined MTD at the lower dose. RESULTS: Thirty eNDMMs were treated, 6 per cohort at 36, 45, and 56 mg/m2 and 12 at 70 mg/m². There was one DLT at 36 mg/m2 (lymphopenia), one at 45 mg/m2 (lysis syndrome), two at 56 mg/m2 (cardiac insufficiency and febrile neutropenia), and two at 70 mg/m2 (vomiting and elevated liver enzymes). The safety profile was acceptable; however, specific attention must be paid to the risk of cardiovascular events, especially for elderly patients. The overall response rate was 93.3%, with 46.6% complete response. CONCLUSIONS: The MTD dose of carfilzomib was 70 mg/m2 in this KMP weekly study in eNDMM. Response rates, and especially CR rate, were remarkable in this population, and would benefit from being assessed in a larger-scale study. The IFM2012-03 study demonstrated that the MTD of carfilzomib weekly is 70 mg/m2 in eNDMM, and 56 mg/m2 for patients older than 75 years. Carfilzomib used weekly in combination has a good efficacy and safety profile in eNDMM.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Aged , Aged, 80 and over , Drug Administration Schedule , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Maximum Tolerated Dose , Melphalan/administration & dosage , Multiple Myeloma/pathology , Oligopeptides/administration & dosage , Patient Safety , Prednisone/administration & dosage , Response Evaluation Criteria in Solid Tumors , Survival Rate , Treatment Outcome
10.
Blood ; 132(24): 2555-2563, 2018 12 13.
Article En | MEDLINE | ID: mdl-30282798

It is important to have an effective therapy for patients with multiple myeloma (MM) at first relapse, particularly if an autologous stem cell transplant (ASCT) is considered at this stage. This multicenter, phase 2 trial evaluated the efficacy and safety of weekly oral pomalidomide-cyclophosphamide-dexamethasone (PCD) in patients with MM in first relapse after treatment with lenalidomide-bortezomib-dexamethasone (RVD). All patients had received RVD as induction and consolidation therapy, plus lenalidomide maintenance for 1 year (arm A). Half had also received an ASCT after induction (arm B). At MM relapse, all patients received 4 oral cycles of pomalidomide 4 mg (days 1-21), cyclophosphamide 300 mg (days 1, 8, 15, and 22), and dexamethasone 40 mg (days 1-4 and days 15-18 of a 28-day cycle; PCD). Responding patients in arm A underwent ASCT and received 2 additional cycles of PCD, whereas those in arm B received 5 cycles of PCD. All patients received pomalidomide-dexamethasone maintenance until disease progression. Primary end point was partial remission or better after the initial 4 cycles of PCD. Responses were obtained in 82/97 (85%) patients evaluated: complete remission (n = 1; 1%), very good partial remission (n = 32; 33%), and partial remission (n = 49; 51%). Three patients (3%) had stable disease, and 6 (6%) had disease progression (6 response failures). Forty-five (94%) of the 48 patients in arm A underwent planned ASCT. PCD was effective therapy after first relapse with RVD. After 4 cycles, the rate of partial remission or better was 85%, and 94% of planned ASCTs were performed. Toxicity was mostly hematologic and manageable. This trial was registered at www.clinicaltrials.gov as #NCT02244125.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Administration, Oral , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Disease-Free Survival , Female , Follow-Up Studies , Humans , Male , Middle Aged , Recurrence , Survival Rate , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/analogs & derivatives
11.
Clin Case Rep ; 6(1): 232-234, 2018 01.
Article En | MEDLINE | ID: mdl-29375875

Infectious lymph nodes mimicking lymphoma is challenging for accurate staging. Although 18F-FDG is a nonspecific tracer accumulating not only in tumor cells but also in inflammatory tissues, the metabolic features and uptake kinetics give valuable information: 18F-FDG PET/CT appears as a useful problem solver tool in ambiguous situation.

12.
Invest Radiol ; 46(5): 341-9, 2011 May.
Article En | MEDLINE | ID: mdl-21263330

OBJECTIVES: The aim of this pilot study was to assess post-treatment apparent diffusion coefficient (ADC) changes of diffuse large B-cell lymphoma lesions on respiratory-gated whole-body diffusion-weighted imaging (DWI), with integrated (18)F-fluoro-2-deoxyglucose positron emission tomography/computed tomography (PET/CT) as the reference standard. MATERIALS AND METHODS: A total of 15 patients underwent both whole-body DWI (b = 50, 400, 800 s/mm(2)) and PET/CT before initiation and after 4 cycles of chemotherapy. ADC of residual masses (lymph node and organ lesions) was assessed both visually and quantitatively, including measurement of mean ADC (ADC).(Figure is included in full-text article.) RESULTS: After chemotherapy, among 85 examined lymph node regions, residual nodes were present in 62 (73%) regions on DWI. Of these 62 regions, 26 had persistent lymph nodes with longest transverse diameter >10 mm, ie, positive based on DWI size criteria. The mean ADC of these 26 regions increased from 0.658 × 10(-3) ± 0.153 mm(2)/s (standard deviation) at baseline to 1.501 × 10(-3) ± 0.307 mm(2)/s (paired t test, P < 0.0001). Only 6 of these 26 regions were considered positive on PET/CT. Combining visual ADC analysis to size criteria reduced the number of false-positive results of DWI from 20 to 2 regions. For organ involvement, ADC values also increased compared with baseline (1.558 × 10(-3) ± 0.424 mm(2)/s vs. 0.675 × 10(-3) ± 0.135 mm(2)/s, respectively; P = 0.0009). CONCLUSIONS: Whole-body DWI with ADC mapping can show a significant increase in ADC values of residual masses persisting after treatment and may help to assess the treatment response in patients with diffuse large B-cell lymphoma.


Diffusion Magnetic Resonance Imaging/methods , Lymphoma, Large B-Cell, Diffuse/diagnosis , Adult , Aged , Antineoplastic Agents/therapeutic use , Diffusion Magnetic Resonance Imaging/instrumentation , Female , Fluorodeoxyglucose F18 , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Pilot Projects , Positron-Emission Tomography , Prospective Studies , Statistics, Nonparametric , Time Factors , Tomography, X-Ray Computed , Young Adult
13.
Radiology ; 254(2): 521-31, 2010 Feb.
Article En | MEDLINE | ID: mdl-20093523

PURPOSE: To compare posttreatment bone marrow changes at whole-body dynamic contrast material-enhanced magnetic resonance (MR) imaging with clinical response in patients with multiple myeloma (MM) and to determine if this technique can be used to assess treatment response in patients with MM. MATERIALS AND METHODS: This study was approved by an institutional review board; all patients gave informed written consent. Thirty patients (21 men, nine women; mean age, 58 years +/- 10 [standard deviation]) underwent whole-body dynamic contrast-enhanced MR imaging before treatment, after induction chemotherapy (n = 30), and after autologous stem cell transplantation (ASCT) (n = 20). Maximal percentages of bone marrow (BME(max)) and focal lesion (FLE(max)) enhancement were assessed at each MR imaging examination. Clinical responses were determined on the basis of international uniform response criteria. Posttreatment changes in BME(max)and FLE(max)were compared with clinical response to therapy by using the Mann-Whitney U test. Receiver operating characteristic (ROC) analysis of posttreatment BME(max)was used to identify poor responders. RESULTS: Eleven of 30 patients were good responders to induction chemotherapy; 16 of 20 patients were good responders to ASCT. After induction chemotherapy, mean BME(max)differed between good and poor responders (94.3% vs 138.4%, respectively; P = .02). With the exclusion of results from six examinations with focal lesions in which a poor clinical response was classified but BME(max)had normalized, a posttreatment BME(max)of more than 96.8% had 100% sensitivity for the identification of poor responders (specificity, 76.9%; area under the ROC curve, 0.90; P = .0001). Mean FLE(max)after induction chemotherapy did not differ between good and poor responders. Mean timing (ie, the number of postcontrast dynamic acquisitions where FLE(max)was observed) was significantly delayed in good responders compared with poor responders (4.7 vs 2.9, P < .0001). Post-ASCT MR imaging results correctly depicted all four clinically good responders whose disease subsequently progressed. CONCLUSION: With quantitative analysis of BME(max)and the timing of FLE(max), whole-body dynamic contrast-enhanced MR imaging can be used to assess treatment response in patients with MM.


Bone Marrow/pathology , Magnetic Resonance Imaging/methods , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Whole Body Imaging , Adult , Aged , Contrast Media , Disease Progression , Female , Humans , Male , Meglumine , Middle Aged , Organometallic Compounds , ROC Curve , Retrospective Studies , Statistics, Nonparametric , Stem Cell Transplantation , Transplantation, Autologous , Treatment Outcome
14.
Clin Infect Dis ; 48(8): 1042-51, 2009 Apr 15.
Article En | MEDLINE | ID: mdl-19281327

BACKGROUND: Empirical antifungal therapy is the standard of care for neutropenic patients with hematological malignancies who remain febrile despite broad-spectrum antibacterial treatment. Recent diagnostic improvements may ensure the early diagnosis of potentially invasive fungal disease. Reserving antifungals for this stage may achieve similar survival rates and reduce treatment toxicity and costs. METHODS: In this multicenter, open-label, randomized noninferiority trial, we compared an empirical antifungal strategy with a preemptive one. Empirical treatment was defined as antibacterial treatment of patients who have persistent or recurrent fever. Preemptive treatment was defined as treatment of patients who have clinical, imaging, or galactomannan-antigen-assay evidence suggesting fungal disease. First-line antifungal treatment was amphotericin B deoxycholate (1 mg/kg/day) or liposomal amphotericin (3 mg/kg/day), depending on daily renal function. The primary efficacy outcome was the proportion of patients alive at 14 days after recovery from neutropenia. RESULTS: The median duration of neutropenia (neutrophil count, <500 cells/mm3) for the 293 patients enrolled was 18 days (range, 5-69 days). By intention-to-treat analysis, survival was 97.3% with empirical treatment and 95.1% with preemptive treatment. The lower 95% confidence limit for the difference in mortality was -5.9%, which was within the noninferiority margin of -8%. Probable or proven invasive fungal infections were more common among patients who received preemptive treatment than among patients who received empirical treatment (13 of 143 vs. 4 of 150; P < .05), and most infections occurred during induction therapy (12 of 73 patients in the preemptive treatment group vs. 3 of 78 patients in the empirical treatment group were infected during induction therapy; P < .01). Preemptive treatment did not decrease nephrotoxicity but decreased costs of antifungal therapy by 35%. CONCLUSIONS: Preemptive treatment increased the incidence of invasive fungal disease, without increasing mortality, and decreased the costs of antifungal drugs. Empirical treatment may provide better survival rates for patients receiving induction chemotherapy.


Antifungal Agents/administration & dosage , Mycoses/drug therapy , Neutropenia/complications , Opportunistic Infections/drug therapy , Adult , Aged , Amphotericin B/administration & dosage , Amphotericin B/therapeutic use , Antibiotic Prophylaxis , Antifungal Agents/therapeutic use , Chi-Square Distribution , Deoxycholic Acid/administration & dosage , Deoxycholic Acid/therapeutic use , Drug Combinations , Female , Fever/physiopathology , Humans , Male , Middle Aged , Multicenter Studies as Topic , Mycoses/complications , Mycoses/diagnosis , Mycoses/prevention & control , Opportunistic Infections/prevention & control , Risk Factors , Statistics, Nonparametric
15.
Eur J Haematol ; 83(3): 246-50, 2009 Sep.
Article En | MEDLINE | ID: mdl-19302557

Although the survival of patients with hairy cell leukemia (HCL) has been improved by the therapeutic introduction of interferon alpha and purine analogs, it is still worsened by complications such as severe infections. In this long-term study, we identified factors influencing patient outcomes in 73 patients with HCL. Median age at diagnosis was 53 yr and the gender ratio (M/F) was 2.3. At the time of HCL diagnosis, 60 patients (82%) were symptomatic and 22 of these had an infection. After a median follow-up of 13 yr, eight patients had died of secondary cancer (n = 2), HCL progression (n = 1) and age-related complications (n = 5). The 10-yr overall survival (OS), progression-free survival and relapse rates were 91 +/- 3%, 14 +/- 5% and 87 +/- 5%, respectively. In multivariate analyses, age >53 yr was the only factor adversely influencing OS and secondary cancer incidence, with adjusted hazard ratio (HR) of 9.30 (95%CI, 1.15-76.6; P = 0.037) and 2.80 (95%CI, 1.05-7.71; P = 0.04), respectively. Eleven patients developed severe infections. Absolute lymphocyte count (<1 x 10(9)/L) at diagnosis was the only factor influencing the occurrence of severe infections, with an adjusted HR of 4.01 (P = 0.007). Strikingly, we did not observe any significant correlation between neutrophil or monocyte counts and the incidence of infection. We confirmed long-term survival in HCL but found a high incidence of infection - even late in the course of the disease. The absolute lymphocyte count at diagnosis is a risk factor for the occurrence of severe infections. In addition to careful monitoring of infections, prompt initiation of anti-HCL treatment should be considered in patients with low lymphocyte counts.


Infections/complications , Infections/physiopathology , Leukemia, Hairy Cell/complications , Leukemia, Hairy Cell/physiopathology , Adult , Aged , Female , Follow-Up Studies , Humans , Infections/diagnosis , Leukemia, Hairy Cell/diagnosis , Lymphocytes/cytology , Male , Middle Aged , Monocytes/cytology , Neutrophils/cytology , Nucleosides/chemistry , Retrospective Studies , Risk Factors , Treatment Outcome
16.
Bull Cancer ; 93(5): 453-62, 2006 May.
Article Fr | MEDLINE | ID: mdl-16777623

Granulocyte colony stimulating factors (G-CSF) are largely used in the treatment of hematologic disorders to improve both the myelosuppression which might directly result from the disease or indirectly induced by the numerous chemotherapy regimen. G-CSF reduces the depth and duration of neutropenia in lymphoma patients and thus allows the design of more dose intense chemotherapy regimen which were shown to improve outcome particularly in patients with diffuse large B-cell and Hodgkin's lymphoma. G-CSF has been studied in patients with acute leukemias (ALL and AML) both concomitantly to induction chemotherapy to sensitize leukemic cells and after chemotherapy to reduce the duration of neutropenia and incidence of severe infection but it's benefit in these settings is still controversial. Myelodysplastic syndromes (MDS) can benefit from G-CSF in association with erythropoietin, particularly for patients with relative good prognosis according to the IPSS score at diagnosis. Still, an improvement of Quality of life needs to be demonstrated in the vue of the cost of these strategies. In aplastic anemia (AA), G-CSF has been used as a support during infection or in association with immunosuppressive treatments but caution is needed regarding the risk of clonal evolution in AA. The benefit of low dose G-CSF in chronic severe neutropenia is well established but the long term consequences of continuous G-CSF support are not known. Finally, G-CSF given alone or after chemotherapy as become one of the key components of hematopoietic stem cell mobilization allowing the use of high dose therapies with autologous or allogeneic stem cell support.


Granulocyte Colony-Stimulating Factor/therapeutic use , Hematologic Diseases/drug therapy , Neutropenia/prevention & control , Hodgkin Disease/drug therapy , Humans , Leukemia, Myeloid, Acute/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Myelodysplastic Syndromes/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recombinant Proteins
...