Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Br J Haematol ; 202(2): 379-383, 2023 07.
Article in English | MEDLINE | ID: mdl-37192755

ABSTRACT

Autologous hematopoietic stem cell transplant (ASCT) is the standard curative treatment for patients with high-risk relapsed/refractory Hodgkin lymphoma (R/R HL). The AETHERA study showed survival gain with Brentuximab Vedotin (BV) maintenance after ASCT in BV-naive patients, which was recently confirmed in the retrospective AMAHRELIS cohort, including a majority of BV-exposed patients. However, this approach has not been compared to intensive tandem auto/auto or auto/allo transplant strategies, which were used before BV approval. Here, we matched BV maintenance (AMAHRELIS) and tandem SCT (HR2009) cohorts, and observed that BV maintenance was associated with better survival outcome in patients with HR R/R HL.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hodgkin Disease , Immunoconjugates , Humans , Brentuximab Vedotin , Hodgkin Disease/drug therapy , Retrospective Studies , Immunoconjugates/therapeutic use , Stem Cell Transplantation , Cohort Studies
3.
Eur J Nucl Med Mol Imaging ; 48(4): 1144-1153, 2021 04.
Article in English | MEDLINE | ID: mdl-32860075

ABSTRACT

INTRODUCTION: Patients with relapsed/refractory Hodgkin lymphoma (R/R HL) experience high response rates upon anti-PD1 therapy. In these patients, the optimal duration of treatment and the risk of relapse after anti-PD1 discontinuation are unknown. METHODS: We retrospectively analyzed patients with R/R HL who responded to anti-PD1 monotherapy and discontinued the treatment either because of unacceptable toxicity or prolonged remission. A machine learning algorithm based on 17 candidate variables was trained and validated to predict progression-free survival (PFS) landmarked at the time of discontinuation of anti-PD1 therapy. RESULTS: Forty patients from 14 centers were randomly assigned to training (n = 25) and validation (n = 15) sets. At the time of anti-PD1 discontinuation, patients had received treatment for a median duration of 11.2 (range, 0-time to best response was not statistically significant in discriminating patients with PFS lesser or greater than 12 months). Considering PFS status as a binary variable (alive or dead) at a specific time point (12 months) is convenient, intuitive and allows for comparing the value of potential predicting variables in these two groups of patients. Nonetheless, this approach has two drawbacks: first, it binarizes outcome; second, it excludes patients alive with a time to last follow up lesser 12 months. Therefore, it is less powerful to demonstrate statistically significant association with PFS even if it exists 5 months. Patients discontinued anti-PD1 treatment either because of prolonged remission (N = 27, 67.5%) or unacceptable toxicity (N = 13, 32.5%). Most patients were in CR (N = 35, 87.5%) at the time of anti-PD1 discontinuation. In the training set, the machine learning algorithm identified that the most important variables to predict PFS were patients' age, time to best response, and presence or absence of CR. The performance observed in the training set was validated in the validation set. CONCLUSION: In this pilot, proof of concept study using a machine learning algorithm, we identified biomarkers capable of predicting the risk of relapse after anti-PD1 discontinuation (age, time to best response, quality of response). Once confirmed, these simple biomarkers will represent useful tools to guide the management of these patients.


Subject(s)
Hodgkin Disease , Chronic Disease , Hodgkin Disease/drug therapy , Humans , Neoplasm Recurrence, Local/drug therapy , Progression-Free Survival , Retrospective Studies
4.
Curr Res Transl Med ; 66(3): 87-89, 2018 09.
Article in English | MEDLINE | ID: mdl-30191811

ABSTRACT

Patients with relapsed or refractory Hodgkin's lymphoma are likely incurable with standard treatment. Idelalisib, a delta-isoform specific Phosphatidyl-inositol-3-kinase (PI3K) inhibitor has shown its efficacy in other hematopoietic B malignancies. We report the case of a 51-years old patient with relapsed and refractory Hodgkin's Lymphoma receiving idelalisib after several regimens of chemotherapy. He achieved a good partial response for several months, unfortunately, idelalisib had to be stopped because of the onset of a severe polyradiculoneuritis attributed to this treatment. We assume here that the polyradiculoneuritis could be caused by T cell mediated autoimmunity to myelin proteins. To our knowledge, this adverse event has never been described so far with idelalisib.


Subject(s)
Hodgkin Disease/drug therapy , Polyradiculopathy/chemically induced , Purines/adverse effects , Quinazolinones/adverse effects , Acute Disease , Adult , Hodgkin Disease/pathology , Humans , Male , Polyradiculopathy/diagnosis , Purines/therapeutic use , Quinazolinones/therapeutic use , Recurrence
5.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1093-1094: 158-166, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30029202

ABSTRACT

Ibrutinib is an orally administered first-in-class irreversible Bruton's tyrosine kinase (BTK) covalent inhibitor for the treatment of patients with B-cell malignancies. Several isolated clinical observations reported its efficacy in central nervous system dissemination. Herein, we described the development and validation of an ultra-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) procedure for the quantification of ibrutinib and its active metabolite PCI-45227 in cerebrospinal fluid (CSF). This is the first complete validated method for quantification of ibrutinib and PCI-45227 in CSF. The compounds were eluted on a Waters BEH C18 column (50.0 × 2.1 mm; 1.7 µm) using a gradient elution with a mobile phase composed of ammonium formate buffer 5 mM pH 3.2 and acetonitrile +0.1% formic acid with a flow rate of 400 µL·min-1. Two deuterated internal standards were used to obtain the most accurate quantification. The CSF samples were prepared by a simple and rapid dilution. The method was validated by testing the selectivity, response function, intra-day and inter-day precisions, trueness, limits of detection (LOD) and lower limits of quantification (LLOQ). The validation results proved that the methods were suitable to quantify ibrutinib and PCI-45227 in real biological CSF samples from 0.50 (ibrutinib) or 1.00 (PCI-45227) to 30.00 ng·mL-1. Furthermore, the developed method was adapted to allow the quantification of both compounds in plasma and the results were compared to those reported in literature. The plasmatic samples were treated by protein precipitation and the method was validated to quantify ibrutinib and PCI-45227 in real biological plasmatic samples from 5.00 to 491 ng·mL-1. Lastly, for both matrices, accuracy profiles were plotted from the trueness and precision results using a 20% α-risk (ß = 80%) and the tolerance intervals were comprised within the acceptance limits fixed at ±25% for the LLOQ and ±15% for the other concentrations. Finally, these methods were successfully applied to quantify ibrutinib and PCI-45227 in real human CSF and plasma samples.


Subject(s)
Adenine/analogs & derivatives , Chromatography, High Pressure Liquid/methods , Pyrazoles/cerebrospinal fluid , Pyrimidines/cerebrospinal fluid , Tandem Mass Spectrometry/methods , Adenine/blood , Adenine/cerebrospinal fluid , Adenine/chemistry , Adenine/therapeutic use , Humans , Limit of Detection , Lymphoma, B-Cell/drug therapy , Piperidines , Pyrazoles/blood , Pyrazoles/chemistry , Pyrazoles/therapeutic use , Pyrimidines/blood , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Reproducibility of Results
6.
Med Mal Infect ; 47(8): 532-539, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28823390

ABSTRACT

OBJECTIVES: Adults with hematological malignancies are at high-risk of Clostridium difficile infection (CDI), but no guidelines for CDI treatment are available in this population. Our primary objective was to evaluate the clinical outcomes in CDI patients with hematological malignancies. Our secondary objectives were to describe CDI severity using the main clinical guidelines and to evaluate the compliance of treatment choice with published guidelines. PATIENTS AND METHODS: Single-center, retrospective, observational case series including every consecutive adult patient with a confirmed diagnosis of CDI admitted in the hematology unit of our teaching hospital. Each CDI episode was classified as moderate, severe, or complicated according to the main clinical guidelines (IDSA 2010, AJG 2013, ESCMID 2014). RESULTS: Twenty-three episodes of CDI in 19 patients admitted to the hematology unit occurred between June 2012 and October 2013. Clinical cure was achieved for 20 episodes (87%). Ten weeks after diagnosis, global cure was reached for 14 episodes (61%) whereas recurrence occurred in two episodes (10%). The mortality rate reached 37% (7/19) but the attributable mortality rate was 5% (1/19). ESCMID criteria more frequently classified patients in the severe category compared with the two other classifications. Prescription compliance with clinical guidelines was observed in 61% of episodes with IDSA criteria, 43% with AJG, and 9% with ESCMID. CONCLUSIONS: IDSA and AJG assessment may underestimate the potential risk of unfavorable clinical outcome. Further prospective studies on a larger cohort are needed to develop adequate treatment guidelines for CDI in hematology settings.


Subject(s)
Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Hematologic Neoplasms/complications , Practice Guidelines as Topic , Adult , Clostridium Infections/epidemiology , Cross Infection/drug therapy , Cross Infection/microbiology , Disease Management , Female , Hospital Units , Hospitals, Teaching , Humans , Immunocompromised Host , Male , Medication Adherence , Metronidazole/therapeutic use , Middle Aged , Recurrence , Retrospective Studies , Severity of Illness Index , Treatment Outcome , Vancomycin/therapeutic use
7.
Ann Oncol ; 28(4): 843-848, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28031174

ABSTRACT

Background: Plasmablastic lymphoma (PBL), initially described in 1997 in the oral cavity of HIV positive patients, is now recognized as a distinct aggressive and rare entity of diffuse large B-cells lymphoma by the World Health Organization (WHO) classification. Since the original description, others cases have been reported. However, these are largely derived from case reports or small series limiting any definitive conclusions on clinical characteristics and outcome. Patients and methods: The clinical, biological, pathological features and outcome of a cohort including 135 patients with PBL, from LYSA centers in France and Belgium, were reported and analyzed. Results: The median age was 58 years, with a male predominance. The cohort was divided into 56 HIV-positive patients, 17 post-transplant patients and 62 HIV-negative/non-transplanted patients. Within HIV-negative/non-transplanted, a relative immunosuppression was found in most cases (systemic inflammatory disease, history of cancer, increased age associated with weakened immune system). We have also described a new subtype, PBL arising in a chronic localized inflammatory site, without any sign of immunosuppression. At presentation, 19% of patients showed oral involvement. Immunophenotype showed CD138 positivity in 88% of cases and CD20 negativity in 90% of cases. Chemotherapy was administered to 80% of patients, with a complete response (CR) rate of 55%. The median overall survival (OS) was 32 months. In univariate analysis, HIV positive status showed better OS when compared with HIV negative status. In multivariate analysis, International Prognostic Index score, chemotherapy and CR were associated with survival benefit. Conclusion(s): This cohort, the largest reported to date, increases the spectrum of knowledge on PBL, rarely described. However, specific guidelines to clarify treatment are lacking, and may improve the poor prognosis of this rare disease.


Subject(s)
Plasmablastic Lymphoma , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Belgium , Comorbidity , Female , France , HIV Infections/epidemiology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Plasmablastic Lymphoma/epidemiology , Plasmablastic Lymphoma/immunology , Plasmablastic Lymphoma/pathology , Proportional Hazards Models , Transplant Recipients , Young Adult
9.
Rev Med Interne ; 36(9): 613-8, 2015 Sep.
Article in French | MEDLINE | ID: mdl-26257103

ABSTRACT

Once characterized by a very poor outcome, multiple myeloma (MM) now has a significantly prolonged survival, with major improvements allowed by the use of "novel agents": proteasome inhibitors (first-in-class bortezomib) and immunomodulatory compounds (IMiDs; first-in-class thalidomide and lenalidomide). However, the vast majority - if not all - of patients with MM ultimately end up being refractory to all existing drugs, including these efficient novel agents. There is a clear unmet medical need in this situation, which warrants the development of the next generation of proteasome inhibitors and IMiDs, as well as new drug classes. This review focuses on pomalidomide, the next generation IMiD, recently approved by the US FDA and the EMA for patients with relapsed or refractory MM who have received at least two prior therapies, including lenalidomide and bortezomib, and have demonstrated disease progression on their last therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Multiple Myeloma/drug therapy , Thalidomide/analogs & derivatives , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Clinical Trials as Topic/methods , Humans , Molecular Conformation , Multiple Myeloma/epidemiology , Structure-Activity Relationship , Thalidomide/chemistry , Thalidomide/pharmacokinetics , Thalidomide/therapeutic use
10.
Rev Med Interne ; 35(4): 243-9, 2014 Apr.
Article in French | MEDLINE | ID: mdl-24050785

ABSTRACT

Smoldering multiple myeloma (SMM) is an asymptomatic plasma cell neoplasia, characterized by monoclonal plasma cell proliferation in the absence of end-organ damage, but with a high risk of progression to multiple myeloma. It has therefore to be distinguished from monoclonal gammapathy of undetermined significance (MGUS), which has a much lower risk of progression, but also from multiple myeloma, which remains an incurable disease and requires a specific treatment. The critical question in the management of SMM is whether an early therapeutic strategy could help delaying the progression to multiple myeloma, in order to lower the risk of serious complications related to this progression, or even to cure the disease. This early treatment could not be proposed to all SMM patients, who are indeed asymptomatic, and in whom the risk of toxicity could make it difficult to justify the potential benefit obtained. The challenge is to target early at diagnosis SMM patients with a high risk of progression, using available routine tests sufficiently reliable to warrant the therapeutic sanction which relies on it. Today however, apart from randomized studies, recommendations are to maintain therapeutic abstention in SMM patients.


Subject(s)
Multiple Myeloma , Disease Progression , Humans , Monitoring, Physiologic/methods , Monoclonal Gammopathy of Undetermined Significance/pathology , Multiple Myeloma/diagnosis , Multiple Myeloma/epidemiology , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Prognosis
11.
Leukemia ; 27(11): 2165-76, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23628958

ABSTRACT

Chromosomal rearrangements of the human MLL (mixed lineage leukemia) gene are associated with high-risk infant, pediatric, adult and therapy-induced acute leukemias. We used long-distance inverse-polymerase chain reaction to characterize the chromosomal rearrangement of individual acute leukemia patients. We present data of the molecular characterization of 1590 MLL-rearranged biopsy samples obtained from acute leukemia patients. The precise localization of genomic breakpoints within the MLL gene and the involved translocation partner genes (TPGs) were determined and novel TPGs identified. All patients were classified according to their gender (852 females and 745 males), age at diagnosis (558 infant, 416 pediatric and 616 adult leukemia patients) and other clinical criteria. Combined data of our study and recently published data revealed a total of 121 different MLL rearrangements, of which 79 TPGs are now characterized at the molecular level. However, only seven rearrangements seem to be predominantly associated with illegitimate recombinations of the MLL gene (≈ 90%): AFF1/AF4, MLLT3/AF9, MLLT1/ENL, MLLT10/AF10, ELL, partial tandem duplications (MLL PTDs) and MLLT4/AF6, respectively. The MLL breakpoint distributions for all clinical relevant subtypes (gender, disease type, age at diagnosis, reciprocal, complex and therapy-induced translocations) are presented. Finally, we present the extending network of reciprocal MLL fusions deriving from complex rearrangements.


Subject(s)
Chromosome Breakage , Gene Rearrangement , Leukemia/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Translocation, Genetic/genetics , Acute Disease , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Animals , Child , Child, Preschool , Female , Histone-Lysine N-Methyltransferase , Humans , Infant , Infant, Newborn , Leukemia/classification , Male , Mice , Middle Aged , Polymerase Chain Reaction , Prognosis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...