125 research outputs found

    Risk and Response-Adapted Treatment in Multiple Myeloma

    Get PDF
    Myeloma therapeutic strategies have been adapted to patients' age and comorbidities for a long time. However, although cytogenetics and clinical presentations (plasmablastic cytology; extramedullary disease) are major prognostic factors, until recently, all patients received the same treatment whatever their initial risk. No strong evidence allows us to use a personalized treatment according to one cytogenetic abnormality in newly diagnosed myeloma. Retrospective studies showed a benefit of a double autologous transplant in high-risk cytogenetics according to the International Myeloma Working Group definition (t(4;14), t(14;16) or del(17p)). Moreover, this definition has to be updated since other independent abnormalities, namely gain 1q, del(1p32), and trisomies 5 or 21, as well as TP53 mutations, are also prognostic. Another very strong predictive tool is the response to treatment assessed by the evaluation of minimal residual disease (MRD). We are convinced that the time has come to use it to adapt the strategy to a dynamic risk. Many trials are ongoing to answer many questions: when and how should we adapt the therapy, its intensity and duration. Nevertheless, we also have to take into account the clinical outcome for one patient, especially adverse events affecting his or her quality of life and his or her preferences for continuous/fixed duration treatment

    Subgroup analysis of ICARIA-MM study in relapsed/refractory multiple myeloma patients with high-risk cytogenetics

    Get PDF
    Treatment benefit in multiple myeloma (MM) patients with high-risk cytogenetics remains suboptimal. The phase 3 ICARIA-MM trial (NCT02990338) showed that isatuximab plus pomalidomide-dexamethasone prolongs median progression-free survival (mPFS) in patients with relapsed/refractory MM (RRMM). This subgroup analysis of ICARIA-MM compared the benefit of isatuximab in high-risk [defined by the presence of del(17p), t(4;14) or t(14;16)] versus standard-risk patients. The efficacy of isatuximab in patients with gain(1q21) abnormality was also assessed in a retrospective subgroup analysis. In ICARIA-MM, 307 patients received isatuximab-pomalidomide-dexamethasone (n = 154) or pomalidomide-dexamethasone (n = 153). Isatuximab (10 mg/kg intravenously) was given weekly in the first 28-day cycle, and every other week thereafter. Standard pomalidomide-dexamethasone doses were given. Isatuximab-pomalidomide-dexamethasone improved mPFS (7 center dot 5 vs 3 center dot 7 months; HR, 0 center dot 66; 95% CI, 0 center dot 33-1 center dot 28) and overall response rate (ORR, 50 center dot 0% vs 16 center dot 7%) in high-risk patients. In patients with isolated gain(1q21), isatuximab addition improved mPFS (11 center dot 2 vs 4 center dot 6 months; HR, 0 center dot 50; 95% CI, 0 center dot 28-0 center dot 88) and ORR (53 center dot 6% vs 27 center dot 6%). More grade >= 3 adverse events occurred in high-risk patients receiving isatuximab (95 center dot 7%) versus the control group (67 center dot 6%); however, isatuximab did not increase events leading to discontinuation or treatment-related mortality. Isatuximab-pomalidomide-dexamethasone provides a consistent benefit over pomalidomide-dexamethasone treatment in RRMM patients regardless of cytogenetic risk

    LocoMMotion:a prospective, non-interventional, multinational study of real-life current standards of care in patients with relapsed and/or refractory multiple myeloma

    Get PDF
    Despite treatment advances, patients with multiple myeloma (MM) often progress through standard drug classes including proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), and anti-CD38 monoclonal antibodies (mAbs). LocoMMotion (ClinicalTrials.gov identifier: NCT04035226) is the first prospective study of real-life standard of care (SOC) in triple-class exposed (received at least a PI, IMiD, and anti-CD38 mAb) patients with relapsed/refractory MM (RRMM). Patients (N = 248; ECOG performance status of 0–1, ≄3 prior lines of therapy or double refractory to a PI and IMiD) were treated with median 4.0 (range, 1–20) cycles of SOC therapy. Overall response rate was 29.8% (95% CI: 24.2–36.0). Median progression-free survival (PFS) and median overall survival (OS) were 4.6 (95% CI: 3.9–5.6) and 12.4 months (95% CI: 10.3–NE). Treatment-emergent adverse events (TEAEs) were reported in 83.5% of patients (52.8% grade 3/4). Altogether, 107 deaths occurred, due to progressive disease (n = 74), TEAEs (n = 19), and other reasons (n = 14). The 92 varied regimens utilized demonstrate a lack of clear SOC for heavily pretreated, triple-class exposed patients with RRMM in real-world practice and result in poor outcomes. This supports a need for new treatments with novel mechanisms of action

    Similar NF-ÎșB Gene Signatures in TNF-α Treated Human Endothelial Cells and Breast Tumor Biopsies

    Get PDF
    BACKGROUND: Endothelial dysfunction has been implicated in the pathogenesis of diverse pathologies ranging from vascular and immune diseases to cancer. TNF-α is one of the mediators of endothelial dysfunction through the activation of transcription factors, including NF-ÎșB. While HUVEC (macrovascular cells) have been largely used in the past, here, we documented an NF-ÎșB gene signature in TNFα-stimulated microvascular endothelial cells HMEC often used in tumor angiogenesis studies. METHODOLOGY/PRINCIPAL FINDINGS: We measured mRNA expression of 55 NF-ÎșB related genes using quantitative RT-PCR in HUVEC and HMEC. Our study identified twenty genes markedly up-regulated in response to TNFα, including adhesion molecules, cytokines, chemokines, and apoptosis regulators, some of them being identified as TNF-α-inducible genes for the first time in endothelial cells (two apoptosis regulators, TNFAIP3 and TNFRSF10B/Trail R2 (DR5), the chemokines GM-CSF/CSF2 and MCF/CSF1, and CD40 and TNF-α itself, as well as NF-ÎșB components (RELB, NFKB1 or 50/p105 and NFKB2 or p52/p100). For eight genes, the fold induction was much higher in HMEC, as compared to HUVEC. Most importantly, our study described for the first time a connection between NF-ÎșB activation and the induction of most, if not all, of these genes in HMEC as evaluated by pharmacological inhibition and RelA expression knock-down by RNA interference. Moreover, since TNF-α is highly expressed in tumors, we further applied the NF-ÎșB gene signature documented in TNFα-stimulated endothelial cells to human breast tumors. We found a significant positive correlation between TNF and the majority (85 %) of the identified endothelial TNF-induced genes in a well-defined series of 96 (48 ERα positive and 48 ERα negative) breast tumors. CONCLUSION/SIGNIFICANCE: Taken together these data suggest the potential use of this NF-ÎșB gene signature in analyzing the role of TNF-α in the endothelial dysfunction, as well as in breast tumors independently of the presence of ERα

    Daratumumab plus lenalidomide and dexamethasone for untreated myeloma

    Get PDF
    This is an accepted manuscript of an article published by Massachusetts Medical Society in New England Journal of Medicine on 30/05/2019, available online: https://doi.org/10.1056/NEJMoa1817249 The accepted version of the publication may differ from the final published version.Copyright © 2019 Massachusetts Medical Society. Lenalidomide plus dexamethasone is a standard treatment for patients with newly diagnosed multiple myeloma who are ineligible for autologous stem-cell transplantation. We sought to determine whether the addition of daratumumab would significantly reduce the risk of disease progression or death in this population. METHODS We randomly assigned 737 patients with newly diagnosed multiple myeloma who were ineligible for autologous stem-cell transplantation to receive daratumumab plus lenalidomide and dexamethasone (daratumumab group) or lenalidomide and dexamethasone alone (control group). Treatment was to continue until the occurrence of disease progression or unacceptable side effects. The primary end point was progression-free survival. RESULTS At a median follow-up of 28.0 months, disease progression or death had occurred in 240 patients (97 of 368 patients [26.4%] in the daratumumab group and 143 of 369 patients [38.8%] in the control group). The estimated percentage of patients who were alive without disease progression at 30 months was 70.6% (95% confidence interval [CI], 65.0 to 75.4) in the daratumumab group and 55.6% (95% CI, 49.5 to 61.3) in the control group (hazard ratio for disease progression or death, 0.56; 95% CI, 0.43 to 0.73; P<0.001). The percentage of patients with a complete response or better was 47.6% in the daratumumab group and 24.9% in the control group (P<0.001). A total of 24.2% of the patients in the daratumumab group, as compared with 7.3% of the patients in the control group, had results below the threshold for minimal residual disease (1 tumor cell per 105 white cells) (P<0.001). The most common adverse events of grade 3 or 4 were neutropenia (50.0% in the daratumumab group vs. 35.3% in the control group), anemia (11.8% vs. 19.7%), lymphopenia (15.1% vs. 10.7%), and pneumonia (13.7% vs. 7.9%).Published versio

    Exome sequencing identifies germline variants in DIS3 in familial multiple myeloma

    Get PDF
    [Excerpt] Multiple myeloma (MM) is the third most common hematological malignancy, after Non-Hodgkin Lymphoma and Leukemia. MM is generally preceded by Monoclonal Gammopathy of Undetermined Significance (MGUS) [1], and epidemiological studies have identified older age, male gender, family history, and MGUS as risk factors for developing MM [2]. The somatic mutational landscape of sporadic MM has been increasingly investigated, aiming to identify recurrent genetic events involved in myelomagenesis. Whole exome and whole genome sequencing studies have shown that MM is a genetically heterogeneous disease that evolves through accumulation of both clonal and subclonal driver mutations [3] and identified recurrently somatically mutated genes, including KRAS, NRAS, FAM46C, TP53, DIS3, BRAF, TRAF3, CYLD, RB1 and PRDM1 [3,4,5]. Despite the fact that family-based studies have provided data consistent with an inherited genetic susceptibility to MM compatible with Mendelian transmission [6], the molecular basis of inherited MM predisposition is only partly understood. Genome-Wide Association (GWAS) studies have identified and validated 23 loci significantly associated with an increased risk of developing MM that explain ~16% of heritability [7] and only a subset of familial cases are thought to have a polygenic background [8]. Recent studies have identified rare germline variants predisposing to MM in KDM1A [9], ARID1A and USP45 [10], and the implementation of next-generation sequencing technology will allow the characterization of more such rare variants. [...]French National Cancer Institute (INCA) and the Fondation Française pour la Recherche contre le Myélome et les Gammapathies (FFMRG), the Intergroupe Francophone du Myélome (IFM), NCI R01 NCI CA167824 and a generous donation from Matthew Bell. This work was supported in part through the computational resources and staff expertise provided by Scientific Computing at the Icahn School of Medicine at Mount Sinai. Research reported in this paper was supported by the Office of Research Infrastructure of the National Institutes of Health under award number S10OD018522. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The authors thank the Association des Malades du Myélome Multiple (AF3M) for their continued support and participation. Where authors are identified as personnel of the International Agency for Research on Cancer / World Health Organization, the authors alone are responsible for the views expressed in this article and they do not necessarily represent the decisions, policy or views of the International Agency for Research on Cancer / World Health Organizatio

    Apport d'une approche protéomique dans l'étude des mécanismes d'activation de néoplasies lymphoïdes B

    No full text
    CLL is characterized by a strong heterogeneity of clinical and biological presentation with indolent forms (mutated IgVH, ZAP-70-) and aggressive forms (unmutated IgVH, ZAP-70 +). BCR and the downstream signaling pathways have been the subject of a study of the transcriptional response to a strong stimulation of the BCR. We continued with a proteomic approach. WM is a chronic lymphoproliferative disorder whose pathophysiology remains poorly understood, although a recurrent mutation has recently been described. We have shown that the overall 48 proteomic profiles analysis allowed to distinguish between CLL cells M and UM before stimulation. Among the differentially expressed proteins include HCLS1 including protein, whose role has already been explored in CLL. Furthermore, stimulation of the BCR induces a specific response in proteomics aggressive LLC cells, corresponding to protein expression changes involved in cellular signaling, regulation of the immune response, protein metabolism, cell growth and apoptosis. The decrease in expression of two proteins, and RAD23B PDCD4 after stimulation aggressive cells was confirmed by Western blotting in 19 patients. This DIGE technology also allows the study of different protein isoforms (especially phosphorylation isoforms), we observed phosphorylation state changes more involved in the cytoskeleton after stimulation of RCC (rolled, vimentin ....). A proteomic study by two-dimensional electrophoresis E2D DIGE on primary cells of blood and marrow from carriers MW previously untreated patients, in comparison to other lymphoproliferative disorders such as marginal zone lymphoma (MZL) or CLL, helped to highlight a specific proteomic profile of cell MW. Among the spots differentially expressed polypeptide is to highlight the under-expression of Ku70 protein in patients MW compared with other lymphoproliferative disorders. The confirmation of this under-expression of Ku70 was confirmed at the transcriptional level by conventional PCR and at the protein level by Western blotting in a larger cohort of patients. We were able to highlight specific proteomic profiles aggressive forms and identification of differently expressed proteins allowed to identify new proteins involved in aggressiveness and pathophysiology of diseases, opening the way for new studies will focus on the regulation of these molecules of interestLa LLC est caractĂ©risĂ©e par une forte hĂ©tĂ©rogĂ©nĂ©itĂ© de prĂ©sentation clinico-biologique avec description de formes indolentes (IGVH mutes, ZAP-70-) et de formes agressives (IGVH non mutes, ZAP-70+). Le BCR et les voies de signalisation en aval ont fait l’objet d'une Ă©tude transcriptionnelle de la rĂ©ponse Ă  une forte stimulation du BCR, que nous avons poursuivi par une approche protĂ©omique. La MW est un syndrome lymphoprolifĂ©ratif chronique dont la physiopathologie reste actuellement mal comprise mĂȘme si une mutation rĂ©currente a Ă©tĂ© rĂ©cemment dĂ©crite. Nous avons pu montrer que l’analyse globale de 48 profils protĂ©omiques permettait de distinguer les cellules de LLC M et UM avant toute stimulation. Parmi les protĂ©ines diffĂ©rentiellement exprimĂ©es, on peut citer notamment la protĂ©ine HCLS1, dont le rĂŽle a dĂ©jĂ  Ă©tĂ© explore dans la LLC. De plus, la stimulation du BCR induit une rĂ©ponse protĂ©omique spĂ©cifique dans les cellules de LLC agressives, correspondant a des variations d’expression de protĂ©ines impliquĂ©es dans la signalisation cellulaire, la rĂ©gulation de la rĂ©ponse immunologique, le mĂ©tabolisme protĂ©ique, la croissance cellulaire et l’apoptose. La diminution d’expression de 2 protĂ©ines, RAD23B et PDCD4, aprĂšs stimulation du BCR de cellules de LLC agressives a Ă©tĂ© confirmĂ©e par Western-Blot chez 19 patients. Cette technologie DIGE permettant Ă©galement l’étude de diffĂ©rents isoformes protĂ©iques (et notamment d’isoformes de phosphorylation), nous avons observe des modifications d’état de phosphorylation de plusieurs protĂ©ines impliquĂ©es dans le cytosquelette aprĂšs stimulation du BCR (lamines, vimentine
.). Une Ă©tude protĂ©omique par Ă©lectrophorĂšse bidimensionnelle E2D DIGE sur des cellules primaires de sang et de moelle issues de patients porteurs de MW non prĂ©alablement traitĂ©s, en comparaison a d’autres syndromes lymphoprolifĂ©ratifs tels les lymphomes de la zone marginale (LZM) ou la LLC, a permis de mettre en Ă©vidence un profil protĂ©omique spĂ©cifique des cellules de MW. Parmi les spots polypeptidiques diffĂ©rentiellement exprimĂ©s, est Ă  souligner la sous-expression de la protĂ©ine Ku70 chez les patients porteurs de MW par rapport aux autres lymphoprolifĂ©rations. La confirmation de cette sous-expression de Ku70 a Ă©tĂ© validĂ©e au niveau transcriptionnel par PCR classique et au niveau protĂ©ique par Western-Blot dans une plus grande cohorte de patients. La mise en Ă©vidence de ces protĂ©ines d'intĂ©rĂȘt dans l'agressivitĂ© et la physiopathologie de ces nĂ©oplasies lymphoĂŻdes ouvrent la voie Ă  de nouvelles Ă©tudes portant sur la rĂ©gulation de ces molĂ©cule

    Proteomics approach to study B cell lymphoid neoplasms

    No full text
    La LLC est caractĂ©risĂ©e par une forte hĂ©tĂ©rogĂ©nĂ©itĂ© de prĂ©sentation clinico-biologique avec description de formes indolentes (IGVH mutes, ZAP-70-) et de formes agressives (IGVH non mutes, ZAP-70+). Le BCR et les voies de signalisation en aval ont fait l’objet d'une Ă©tude transcriptionnelle de la rĂ©ponse Ă  une forte stimulation du BCR, que nous avons poursuivi par une approche protĂ©omique. La MW est un syndrome lymphoprolifĂ©ratif chronique dont la physiopathologie reste actuellement mal comprise mĂȘme si une mutation rĂ©currente a Ă©tĂ© rĂ©cemment dĂ©crite. Nous avons pu montrer que l’analyse globale de 48 profils protĂ©omiques permettait de distinguer les cellules de LLC M et UM avant toute stimulation. Parmi les protĂ©ines diffĂ©rentiellement exprimĂ©es, on peut citer notamment la protĂ©ine HCLS1, dont le rĂŽle a dĂ©jĂ  Ă©tĂ© explore dans la LLC. De plus, la stimulation du BCR induit une rĂ©ponse protĂ©omique spĂ©cifique dans les cellules de LLC agressives, correspondant a des variations d’expression de protĂ©ines impliquĂ©es dans la signalisation cellulaire, la rĂ©gulation de la rĂ©ponse immunologique, le mĂ©tabolisme protĂ©ique, la croissance cellulaire et l’apoptose. La diminution d’expression de 2 protĂ©ines, RAD23B et PDCD4, aprĂšs stimulation du BCR de cellules de LLC agressives a Ă©tĂ© confirmĂ©e par Western-Blot chez 19 patients. Cette technologie DIGE permettant Ă©galement l’étude de diffĂ©rents isoformes protĂ©iques (et notamment d’isoformes de phosphorylation), nous avons observe des modifications d’état de phosphorylation de plusieurs protĂ©ines impliquĂ©es dans le cytosquelette aprĂšs stimulation du BCR (lamines, vimentine
.). Une Ă©tude protĂ©omique par Ă©lectrophorĂšse bidimensionnelle E2D DIGE sur des cellules primaires de sang et de moelle issues de patients porteurs de MW non prĂ©alablement traitĂ©s, en comparaison a d’autres syndromes lymphoprolifĂ©ratifs tels les lymphomes de la zone marginale (LZM) ou la LLC, a permis de mettre en Ă©vidence un profil protĂ©omique spĂ©cifique des cellules de MW. Parmi les spots polypeptidiques diffĂ©rentiellement exprimĂ©s, est Ă  souligner la sous-expression de la protĂ©ine Ku70 chez les patients porteurs de MW par rapport aux autres lymphoprolifĂ©rations. La confirmation de cette sous-expression de Ku70 a Ă©tĂ© validĂ©e au niveau transcriptionnel par PCR classique et au niveau protĂ©ique par Western-Blot dans une plus grande cohorte de patients. La mise en Ă©vidence de ces protĂ©ines d'intĂ©rĂȘt dans l'agressivitĂ© et la physiopathologie de ces nĂ©oplasies lymphoĂŻdes ouvrent la voie Ă  de nouvelles Ă©tudes portant sur la rĂ©gulation de ces molĂ©culesCLL is characterized by a strong heterogeneity of clinical and biological presentation with indolent forms (mutated IgVH, ZAP-70-) and aggressive forms (unmutated IgVH, ZAP-70 +). BCR and the downstream signaling pathways have been the subject of a study of the transcriptional response to a strong stimulation of the BCR. We continued with a proteomic approach. WM is a chronic lymphoproliferative disorder whose pathophysiology remains poorly understood, although a recurrent mutation has recently been described. We have shown that the overall 48 proteomic profiles analysis allowed to distinguish between CLL cells M and UM before stimulation. Among the differentially expressed proteins include HCLS1 including protein, whose role has already been explored in CLL. Furthermore, stimulation of the BCR induces a specific response in proteomics aggressive LLC cells, corresponding to protein expression changes involved in cellular signaling, regulation of the immune response, protein metabolism, cell growth and apoptosis. The decrease in expression of two proteins, and RAD23B PDCD4 after stimulation aggressive cells was confirmed by Western blotting in 19 patients. This DIGE technology also allows the study of different protein isoforms (especially phosphorylation isoforms), we observed phosphorylation state changes more involved in the cytoskeleton after stimulation of RCC (rolled, vimentin ....). A proteomic study by two-dimensional electrophoresis E2D DIGE on primary cells of blood and marrow from carriers MW previously untreated patients, in comparison to other lymphoproliferative disorders such as marginal zone lymphoma (MZL) or CLL, helped to highlight a specific proteomic profile of cell MW. Among the spots differentially expressed polypeptide is to highlight the under-expression of Ku70 protein in patients MW compared with other lymphoproliferative disorders. The confirmation of this under-expression of Ku70 was confirmed at the transcriptional level by conventional PCR and at the protein level by Western blotting in a larger cohort of patients. We were able to highlight specific proteomic profiles aggressive forms and identification of differently expressed proteins allowed to identify new proteins involved in aggressiveness and pathophysiology of diseases, opening the way for new studies will focus on the regulation of these molecules of interes
    • 

    corecore